Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Eur Heart J ; 34(19): 1437-47, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22261894

RESUMO

AIMS: G protein-coupled receptor kinase 2 (GRK2), which is markedly upregulated in failing human myocardium, has been implicated as a contributing factor or consequence of heart failure (HF). Importantly, cardiac-specific GRK2 knockout mice have recently proved the pathological nature of GRK2 in HF. Targeted inhibition of GRK2 is possible using a peptide inhibitor known as the ßARKct, which has rescued several disparate small animal HF models. This study was designed to evaluate long-term ßARKct expression in a clinically relevant large animal HF model, using stable myocardial gene delivery with adeno-associated virus serotype 6 (AAV6). METHODS AND RESULTS: A porcine model of HF subsequent to left ventricular (LV) myocardial infarction (MI) was used to study the effects of retrograde injection into the anterior interventricular vein of either AAV6.ßARKct or AAV6.luciferase as a control 2 weeks after MI. Echocardiography and LV hemodynamics were performed before and 6 weeks after gene transfer. Robust and long-term ßARKct expression was found after AAV6-mediated delivery, leading to significant amelioration of LV haemodynamics and contractile function in HF pigs compared with AAV6.luciferase-treated control animals that showed a continued decline in cardiac function. Interestingly, the neurohormonal axis was virtually normalized in AVV6.ßARKct-treated HF animals, represented by reductions in plasma norepinephrine levels, whereas AAV6.luciferase-treated pigs showed further increases in plasma catecholamine levels. As a result, LV remodelling and foetal gene expression was reversed by AVV6.ßARKct gene therapy. CONCLUSION: These data--showing sustained amelioration of cardiac function in a post-MI pig HF model--demonstrate the therapeutic potential of ßARKct gene therapy for HF.


Assuntos
Terapia Genética/métodos , Insuficiência Cardíaca/terapia , Peptídeos/uso terapêutico , Receptores CCR10/antagonistas & inibidores , Proteínas Recombinantes/uso terapêutico , Adenoviridae , Animais , Catecolaminas/metabolismo , Vasos Coronários , Ecocardiografia , Técnicas de Transferência de Genes , Vetores Genéticos , Insuficiência Cardíaca/fisiopatologia , Hemodinâmica/fisiologia , Luciferases/genética , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Peptídeos/genética , Peptídeos/metabolismo , Distribuição Aleatória , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sus scrofa , Transgenes/genética , Remodelação Ventricular/fisiologia
2.
Circ Res ; 108(1): 27-39, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21106943

RESUMO

RATIONALE: The G(ßγ)-sequestering peptide ß-adrenergic receptor kinase (ßARK)ct derived from the G-protein-coupled receptor kinase (GRK)2 carboxyl terminus has emerged as a promising target for gene-based heart failure therapy. Enhanced downstream cAMP signaling has been proposed as the underlying mechanism for increased ß-adrenergic receptor (ßAR) responsiveness. However, molecular targets mediating improved cardiac contractile performance by ßARKct and its impact on G(ßγ)-mediated signaling have yet to be fully elucidated. OBJECTIVE: We sought to identify G(ßγ)-regulated targets and signaling mechanisms conveying ßARKct-mediated enhanced ßAR responsiveness in normal (NC) and failing (FC) adult rat ventricular cardiomyocytes. METHODS AND RESULTS: Assessing viral-based ßARKct gene delivery with electrophysiological techniques, analysis of contractile performance, subcellular Ca²(+) handling, and site-specific protein phosphorylation, we demonstrate that ßARKct enhances the cardiac L-type Ca²(+) channel (LCC) current (I(Ca)) both in NCs and FCs on ßAR stimulation. Mechanistically, ßARKct augments I(Ca) by preventing enhanced inhibitory interaction between the α1-LCC subunit (Cav1.2α) and liberated G(ßγ) subunits downstream of activated ßARs. Despite improved ßAR contractile responsiveness, ßARKct neither increased nor restored cAMP-dependent protein kinase (PKA) and calmodulin-dependent kinase II signaling including unchanged protein kinase (PK)Cε, extracellular signal-regulated kinase (ERK)1/2, Akt, ERK5, and p38 activation both in NCs and FCs. Accordingly, although ßARKct significantly increases I(Ca) and Ca²(+) transients, being susceptible to suppression by recombinant G(ßγ) protein and use-dependent LCC blocker, ßARKct-expressing cardiomyocytes exhibit equal basal and ßAR-stimulated sarcoplasmic reticulum Ca²(+) load, spontaneous diastolic Ca²(+) leakage, and survival rates and were less susceptible to field-stimulated Ca²(+) waves compared with controls. CONCLUSION: Our study identifies a G(ßγ)-dependent signaling pathway attenuating cardiomyocyte I(Ca) on ßAR as molecular target for the G(ßγ)-sequestering peptide ßARKct. Targeted interruption of this inhibitory signaling pathway by ßARKct confers improved ßAR contractile responsiveness through increased I(Ca) without enhancing regular or restoring abnormal cAMP-signaling. ßARKct-mediated improvement of I(Ca) rendered cardiomyocytes neither susceptible to ßAR-induced damage nor arrhythmogenic sarcoplasmic reticulum Ca²(+) leakage.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Cardiotônicos/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Terapia Genética/métodos , Insuficiência Cardíaca , Contração Miocárdica/genética , Miócitos Cardíacos/metabolismo , Peptídeos/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Sobrevivência Celular/genética , Subunidades beta da Proteína de Ligação ao GTP/genética , Subunidades gama da Proteína de Ligação ao GTP/genética , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/terapia , Ventrículos do Coração/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Peptídeos/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Ratos , Retículo Sarcoplasmático/genética , Retículo Sarcoplasmático/metabolismo
3.
J Cell Mol Med ; 16(12): 3028-36, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22970922

RESUMO

Progressive cardiomyopathy is a major cause of death in Duchenne muscular dystrophy (DMD) patients. Coupling between Ca(2+) handling and contractile properties in dystrophic hearts is poorly understood. It is also not clear whether developing cardiac failure is dominated by alterations in Ca(2+) pathways or more related to the contractile apparatus. We simultaneously recorded force and Ca(2+) transients in field-stimulated papillary muscles from young (10-14 weeks) wild-type (wt) and dystrophic mdx mice. Force amplitudes were fivefold reduced in mdx muscles despite only 30% reduction in fura-2 ratio amplitudes. This indicated mechanisms other than systolic Ca(2+) to additionally account for force decrements in mdx muscles. pCa-force relations revealed decreased mdx myofibrillar Ca(2+) sensitivity. 'In vitro' motility assays, studied in mdx hearts here for the first time, showed significantly slower sliding velocities. mdx MLC/MHC isoforms were not grossly altered. Dystrophic hearts showed echocardiography signs of early ventricular wall hypertrophy with a significantly enlarged end-diastolic diameter 'in vivo'. However, fractional shortening was still comparable to wt mice. Changes in the contractile apparatus satisfactorily explained force drop in mdx hearts. We give first evidence of early hypertrophy in mdx mice and possible mechanisms for already functional impairment of cardiac muscle in DMD.


Assuntos
Coração/fisiopatologia , Distrofia Muscular de Duchenne/fisiopatologia , Contração Miocárdica , Músculos Papilares/fisiopatologia , Animais , Cálcio/metabolismo , Cardiomegalia , Cardiomiopatias , Células Cultivadas , Ecocardiografia , Camundongos , Camundongos Endogâmicos mdx , Miocárdio/citologia , Miocárdio/metabolismo
4.
Neuromuscul Disord ; 29(3): 231-241, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30782477

RESUMO

So far effective strategies to treat cardiomyopathy in patients with muscular dystrophies are still not clearly defined. Previously, treatment with ß-blockers showed beneficial effects on the development of cardiomyopathy in dystrophin-deficient (mdx) mice, but not in δ-sarcoglycan-deficient (Sgcd-/-) mice. We therefore aimed to study a more specific approach to target maladaptive ß-adrenergic signalling in these mice. It has been shown that lowering cardiac G-protein-coupled-receptor-kinase-2 (GRK2) activity with ßARKct expression, a peptide inhibitor of protein-coupled-receptor-kinase-2 (GRK2), results in improvement of heart failure in several different animal models. We therefore investigated whether adeno-associated virus type 9 (AAV9)-mediated gene delivery of ßARKct, could ameliorate cardiac pathology in mdx and Sgcd-/- mice. We found that long-term treatment with AAV9- ßARKct-cDNA with a cardiac-specific promoter significantly improves left ventricular systolic function and reduces myocardial hypertrophy in mdx mice, whereas only mild beneficial effects on cardiac function is observed in Sgcd-/- mice. Interestingly, in contrast to mdx mice neither GRK2 nor nuclear-factor-kappaB (NFκB) were upregulated in Sgcd-/- mice. Taken together, effectiveness of AAV-mediated ßARKct therapy may vary between different genetic mutations and presumably depend on the state of adrenergic dysregulation mediated through the upregulation of GRK2.


Assuntos
Cardiomiopatias/genética , Dependovirus , Distrofina/deficiência , Distrofias Musculares/genética , Sarcoglicanas/genética , Animais , Dependovirus/genética , Distrofina/genética , Terapia Genética/métodos , Coração/fisiopatologia , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/fisiopatologia , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Distrofias Musculares/metabolismo , Função Ventricular Esquerda/genética
5.
Eur J Hum Genet ; 22(1): 119-25, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23695275

RESUMO

So far, the role of mutations in the δ-sarcogylcan (Sgcd) gene in causing autosomal dominant dilated cardiomyopathy (DCM) remains inconclusive. A p.S151A missense mutation in exon 6 of the Sgcd gene was reported to cause severe isolated autosomal dominant DCM without affecting skeletal muscle. This is controversial to our previous findings in a large consanguineous family where this p.S151A mutation showed no relevance for cardiac disease. In this study, the potential of the p.S151A mutation to cause DCM was investigated by using two different approaches: (1) engineering and characterization of heterozygous knock-in (S151A-) mice carrying the p.S151A mutation and (2) evaluation of the potential of adeno-associated virus (AAV) 9-based cardiac-specific transfer of p.S151A-mutated Sgcd cDNA to rescue the cardiac phenotype in Sgcd-deficient (Sgcd-null) mice as it has been demonstrated for intact, wild-type Sgcd cDNA. Heterozygous S151A knock-in mice developed a rather mild phenotype of cardiomyopathy. Increased heart to body weight suggests cardiac enlargement in 1-year-old S151A knock-in mice. However, at this age cardiac function, assessed by echocardiography, is maintained and histopathology completely absent. Myocardial expression of p.S151A cDNA, similar to intact Sgcd cDNA, restores cardiac function, although not being able to prevent myocardial histopathology in Sgcd-null mice completely. Our results suggest that the p.S151A mutation causes a mild, subclinical phenotype of cardiomyopathy, which is prone to be overseen in patients carrying such sequence variants. Furthermore, this study shows the suitability of an AAV-mediated cardiac gene transfer approach to analyze whether a sequence variant is a disease-causing mutation.


Assuntos
Cardiomiopatias/genética , Mutação de Sentido Incorreto , Sarcoglicanas/genética , Animais , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Dependovirus , Técnicas de Introdução de Genes , Heterozigoto , Humanos , Camundongos , Miocárdio/patologia , Fenótipo
6.
Hum Gene Ther ; 23(6): 566-75, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22248393

RESUMO

Dystrophin plays an important role in muscle contraction, linking the intracellular cytoskeleton to the extracellular matrix. Mutations of the dystrophin gene leading to a complete loss of the protein cause Duchenne muscular dystrophy (DMD), frequently associated with severe cardiomyopathy. Early clinical trials in DMD using gene transfer to skeletal muscle are underway, but gene transfer to dystrophic cardiac muscle has not yet been tested in humans. The aim of this study was to develop an optimized protocol for cardiac gene therapy in the mouse model of dystrophin deficiency (mdx), using a cardiac promoter for expression of a microdystrophin (µDys) transgene packaged into an adeno-associated virus serotype 9 vector (AAV9). In this study adult mdx mice were intravenously injected with 1×10(12) genomic particles of AAV9 vectors carrying a cDNA encoding µDys under the control of either a ubiquitously active cytomegalovirus (CMV) promoter or a cardiac-specific CMV-enhanced myosin light chain (MLC0.26) promoter. After 10 months, both AAV9 vectors led to sustained µDys expression in cardiac muscle, but the MLC promoter conferred about 4-fold higher protein levels. AAV9-CMV-MLC0.26-µDys resulted in significant protection of cardiac morphology and function as assessed by histopathology, echocardiography, and left ventricular catheterization. In conclusion, we established an AAV9-mediated gene transfer approach for efficient and specific long-term µDys expression in the hearts of mdx mice, resulting in a sustained therapeutic effect. Thus, this approach might be a basis for further translation into a treatment strategy for DMD-associated cardiomyopathy.


Assuntos
Dependovirus/genética , Distrofina/genética , Terapia Genética , Miocárdio/citologia , Animais , Western Blotting , Modelos Animais de Doenças , Distrofina/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos mdx , Miocárdio/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas
7.
Sci Transl Med ; 3(92): 92ra64, 2011 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-21775667

RESUMO

As a prerequisite for clinical application, we determined the long-term therapeutic effectiveness and safety of adeno-associated virus (AAV)-S100A1 gene therapy in a preclinical large animal model of heart failure. S100A1, a positive inotropic regulator of myocardial contractility, becomes depleted in failing cardiomyocytes in humans and animals, and myocardial-targeted S100A1 gene transfer rescues cardiac contractile function by restoring sarcoplasmic reticulum calcium (Ca(2+)) handling in acutely and chronically failing hearts in small animal models. We induced heart failure in domestic pigs by balloon occlusion of the left circumflex coronary artery, resulting in myocardial infarction. After 2 weeks, when the pigs displayed significant left ventricular contractile dysfunction, we administered, by retrograde coronary venous delivery, AAV serotype 9 (AAV9)-S100A1 to the left ventricular, non-infarcted myocardium. AAV9-luciferase and saline treatment served as control. At 14 weeks, both control groups showed significantly decreased myocardial S100A1 protein expression along with progressive deterioration of cardiac performance and left ventricular remodeling. AAV9-S100A1 treatment prevented and reversed these functional and structural changes by restoring cardiac S100A1 protein levels. S100A1 treatment normalized cardiomyocyte Ca(2+) cycling, sarcoplasmic reticulum calcium handling, and energy homeostasis. Transgene expression was restricted to cardiac tissue, and extracardiac organ function was uncompromised. This translational study shows the preclinical feasibility of long-term therapeutic effectiveness of and a favorable safety profile for cardiac AAV9-S100A1 gene therapy in a preclinical model of heart failure. Our results present a strong rationale for a clinical trial of S100A1 gene therapy for human heart failure that could potentially complement current strategies to treat end-stage heart failure.


Assuntos
Dependovirus/genética , Modelos Animais de Doenças , Terapia Genética , Insuficiência Cardíaca/terapia , Isquemia Miocárdica/complicações , Miocárdio/metabolismo , Proteínas S100/genética , Proteínas S100/uso terapêutico , Animais , Biomarcadores/sangue , Cálcio/metabolismo , Metabolismo Energético , Técnicas de Transferência de Genes , Terapia Genética/efeitos adversos , Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/fisiopatologia , Testes de Função Cardíaca , Homeostase , Humanos , Infarto do Miocárdio/sangue , Infarto do Miocárdio/complicações , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/sangue , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/terapia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Especificidade de Órgãos , Retículo Sarcoplasmático/metabolismo , Sus scrofa
8.
Cardiovasc Res ; 82(3): 404-10, 2009 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-19218289

RESUMO

AIMS: Delta-sarcoglycan is a member of the dystrophin-associated glycoprotein complex linking the cytoskeleton to the extracellular matrix. Similar to patients with defects in the gene encoding delta-sarcoglycan (Sgcd), knockout mice develop cardiomyopathy and muscular dystrophy. The aim of our study was to develop an approach for preventing cardiomyopathy in Sgcd-deficient mice by cardiac expression of the intact cDNA upon systemic delivery of adeno-associated viral (AAV) vectors. METHODS AND RESULTS: We packaged the Sgcd cDNA under transcriptional control of a myosin light chain-promoter fused with a cytomegalovirus enhancer into AAV-9 capsids. Vectors carrying either the Sgcd cDNA or an enhanced green fluorescent protein (EGFP) reporter gene were intravenously injected into adult Sgcd knockout mice. After 6 months, immunohistochemistry revealed almost complete reconstitution of the sarcoglycan subcomplex in heart but not skeletal muscle of mice with the Sgcd vector. Furthermore, Sgcd gene transfer resulted in prevention of cardiac fibrosis and significantly increased running distance measured by voluntary wheel running. Left ventricular function remained stable in mice expressing Sgcd while it deteriorated in EGFP controls within 6 months, paralleled by increased expression of brain natriuretic peptide, a molecular marker of heart failure. CONCLUSION: Our study establishes an approach to specifically treat hereditary cardiomyopathies by targeting gene expression into the myocardium upon systemic application of AAV vectors.


Assuntos
Cardiomiopatias/prevenção & controle , Dependovirus , Terapia Genética , Miocárdio/metabolismo , Sarcoglicanas/genética , Animais , Teste de Esforço , Técnicas de Transferência de Genes , Vetores Genéticos , Insuficiência Cardíaca/prevenção & controle , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Miocárdio/patologia , Sarcoglicanas/metabolismo , Função Ventricular Esquerda
9.
Scand J Gastroenterol ; 41(8): 989-92, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16803699

RESUMO

This is the first report of a case of biliopancreatic fistula complicating a pancreatic pseudocyst diagnosed correctly by transabdominal ultrasound. The diagnosis was confirmed by magnetic resonance and endoscopic retrograde cholangiopancreatography. The fistula was treated successfully with biliary stenting. The clinical and imaging features of this exceptional complication are presented along with a brief review of the topic.


Assuntos
Fístula Biliar/diagnóstico por imagem , Colangiopancreatografia Retrógrada Endoscópica , Doenças do Ducto Colédoco/diagnóstico por imagem , Fístula Pancreática/diagnóstico por imagem , Pseudocisto Pancreático/diagnóstico por imagem , Idoso , Fístula Biliar/complicações , Fístula Biliar/terapia , Doenças do Ducto Colédoco/complicações , Doenças do Ducto Colédoco/terapia , Humanos , Masculino , Fístula Pancreática/complicações , Fístula Pancreática/terapia , Pseudocisto Pancreático/complicações , Pseudocisto Pancreático/terapia , Ultrassonografia Doppler
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA