Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 29(6): 944-954, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-31976522

RESUMO

Duchenne muscular dystrophy (DMD) is a devastating neuromuscular disease that causes progressive muscle wasting and cardiomyopathy. This X-linked disease results from mutations of the DMD allele on the X-chromosome resulting in the loss of expression of the protein dystrophin. Dystrophin loss causes cellular dysfunction that drives the loss of healthy skeletal muscle and cardiomyocytes. As gene therapy strategies strive toward dystrophin restoration through micro-dystrophin delivery or exon skipping, preclinical models have shown that incomplete restoration in the heart results in heterogeneous dystrophin expression throughout the myocardium. This outcome prompts the question of how much dystrophin restoration is sufficient to rescue the heart from DMD-related pathology. Female DMD carrier hearts can shed light on this question, due to their mosaic cardiac dystrophin expression resulting from random X-inactivation. In this work, a dystrophinopathy carrier mouse model was derived by breeding male or female dystrophin-null mdx mice with a wild type mate. We report that these carrier hearts are significantly susceptible to injury induced by one or multiple high doses of isoproterenol, despite expressing ~57% dystrophin. Importantly, only carrier mice with dystrophic mothers showed mortality after isoproterenol. These findings indicate that dystrophin restoration in approximately half of the heart still allows for marked vulnerability to injury. Additionally, the discovery of divergent stress-induced mortality based on parental origin in mice with equivalent dystrophin expression underscores the need for better understanding of the epigenetic, developmental, and even environmental factors that may modulate vulnerability in the dystrophic heart.


Assuntos
Cardiomiopatias/patologia , Modelos Animais de Doenças , Distrofina/genética , Distrofia Muscular de Duchenne/complicações , Mutação , Miocárdio/patologia , Animais , Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx
2.
J Mol Cell Cardiol ; 128: 51-61, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30664850

RESUMO

BACKGROUND: Duchenne muscular dystrophy (DMD) is an X-linked disease characterized by skeletal muscle degeneration and a significant cardiomyopathy secondary to cardiomyocyte damage and myocardial loss. The molecular basis of DMD lies in the absence of the protein dystrophin, which plays critical roles in mechanical membrane integrity and protein localization at the sarcolemma. A popular mouse model of DMD is the mdx mouse, which lacks dystrophin and displays mild cardiac and skeletal pathology that can be exacerbated to advance the disease state. In clinical and pre-clinical studies of DMD, angiotensin signaling pathways have emerged as therapeutic targets due to their adverse influence on muscle remodeling and oxidative stress. Here we aim to establish a physiologically relevant cardiac injury model in the mdx mouse, and determine whether acute blockade of the angiotensin II type 1 receptor (AT1R) may be utilized for prevention of dystrophic injury. METHODS AND RESULTS: A single IP injection of isoproterenol (Iso, 10 mg/kg) was used to induce cardiac stress and injury in mdx and wild type (C57Bl/10) mice. Mice were euthanized 8 h, 30 h, 1 week, or 1 month following the injection, and hearts were harvested for injury evaluation. At 8 and 30 h post-injury, mdx hearts showed 2.2-fold greater serum cTnI content and 3-fold more extensive injury than wild type hearts. Analysis of hearts 1 week and 1 month after injury revealed significantly higher fibrosis in mdx hearts, with a more robust and longer-lasting immune response compared to wild type hearts. In the 30-hour group, losartan treatment initiated 1 h before Iso injection protected dystrophic hearts from cardiac damage, reducing mdx acute injury area by 2.8-fold, without any significant effect on injury in wild type hearts. However, both wild type and dystrophic hearts showed a 2-fold reduction in the magnitude of the macrophage response to injury 30 h after Iso with losartan. CONCLUSIONS: This work demonstrates that acute blockade of AT1R has the potential for robust injury prevention in a model of Iso-induced dystrophic heart injury. In addition to selectively limiting dystrophic cardiac damage, blocking AT1R may serve to limit the inflammatory nature of the immune response to injury in all hearts. Our findings strongly suggest that earlier adoption of angiotensin receptor blockers in DMD patients could limit myocardial damage and subsequent cardiomyopathy.


Assuntos
Cardiomiopatias/tratamento farmacológico , Coração/efeitos dos fármacos , Distrofia Muscular de Duchenne/tratamento farmacológico , Receptor Tipo 1 de Angiotensina/genética , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Cardiomiopatias/genética , Cardiomiopatias/patologia , Distrofina/genética , Coração/fisiopatologia , Humanos , Isoproterenol/farmacologia , Losartan/farmacologia , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Sarcolema/metabolismo , Sarcolema/patologia
3.
Environ Sci Technol ; 50(14): 7480-8, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27356268

RESUMO

Despite its wide use as a veterinary pharmaceutical, environmental fate data is lacking for altrenogest, a potent synthetic progestin. Here, it is reported that direct photolysis of altrenogest under environmentally relevant conditions was extremely efficient and rapid (half-life ∼25 s). Photolysis rates (observed rate constant kobs = 2.7 ± 0.2 × 10(-2) s(-1)) were unaffected by changes in pH or temperature but were sensitive to oxygen concentrations (N2-saturated kobs = 9.10 ± 0.32 × 10(-2) s(-1); O2-saturated kobs = 1.38 ± 0.11 × 10(-2) s(-1)). The primary photoproduct was identified as an isomer formed via an internal 2 + 2 cycloaddition reaction; the triplet lifetime (8.4 ± 0.2 µs) and rate constant (8 × 10(4) s(-1)) of this reaction were measured using transient absorption spectroscopy. Subsequent characterization determined that this primary cycloaddition photoproduct undergoes photohydration. The resultant photostable secondary photoproducts are subject to thermal dehydration in dark conditions, leading to reversion to the primary cycloaddition photoproduct on a time scale of hours to days, with the photohydration and dehydration repeatable over several light/dark cycles. This dehydration reaction occurs more rapidly at higher temperatures and is also accelerated at both high and low pH values. In vitro androgen receptor (AR)-dependent gene transcriptional activation cell assays and in silico nuclear hormone receptor screening revealed that certain photoproducts retain significant androgenic activity, which has implications for exposure risks associated with the presence and cycling of altrenogest and its photoproducts in the environment.


Assuntos
Fotoquímica , Fotólise , Meio Ambiente , Meia-Vida , Temperatura
5.
Sci Rep ; 10(1): 17195, 2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-33057067

RESUMO

Understanding the energetic state of the heart is essential for unraveling the central tenets of cardiac physiology. The heart uses a tremendous amount of energy and reductions in that energy supply can have lethal consequences. While ischemic events clearly result in significant metabolic perturbations, heart failure with both preserved and reduced ejection fraction display reductions in energetic status. To date, most cardiac energetics have been performed using 31P-NMR, which requires dedicated access to a specialized NMR spectrometer. This has limited the availability of this method to a handful of centers around the world. Here we present a method of assessing myocardial energetics in the isolated mouse heart using 1H-NMR spectrometers that are widely available in NMR core facilities. In addition, this methodology provides information on many other important metabolites within the heart, including unique metabolic differences between the hypoxic and ischemic hearts. Furthermore, we demonstrate the correlation between myocardial energetics and measures of contractile function in the mouse heart. These methods will allow a broader examination of myocardial energetics providing a valuable tool to aid in the understanding of the nature of these energetic deficits and to develop therapies directed at improving myocardial energetics in failing hearts.


Assuntos
Metabolismo Energético/fisiologia , Coração/fisiologia , Miocárdio/metabolismo , Animais , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Contração Miocárdica/fisiologia , N-Glicosil Hidrolases/metabolismo , Espectroscopia de Prótons por Ressonância Magnética/métodos
6.
Sci Rep ; 9(1): 12306, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31444390

RESUMO

Duchenne muscular dystrophy (DMD) is a uniformly fatal condition of striated muscle wasting resulting in premature death from respiratory and/or cardiac failure. Symptomatic therapy has prolonged survival by limiting deaths resulting from respiratory insufficiency, but there is currently no effective therapy for most patients with DMD. This grim prognosis has led patients and their families to seek unproven therapeutic approaches. One such approach is the use of hyperbaric therapies, which 14% of DMD patients self-report using. The primary goal of this study was to determine if intermittent hyperbaric exposure altered the muscle function of the mdx mouse, a genetic model of DMD. To do this, mdx mice were exposed to three daily 90-minute 1.3 atmosphere hyperbaric exposures for 4 weeks. Skeletal muscle, respiratory, and cardiac function were assessed in treated and untreated wild type and dystrophic mice. The results of these studies find that hyperbaric and hyperoxic approaches resulted in increased cardiac fibrosis in dystrophic mice and no beneficial effects on the functional parameters measured. These data suggest that these oxygen-based therapies are unlikely to provide therapeutic benefit to DMD patients.


Assuntos
Traumatismos Cardíacos/etiologia , Traumatismos Cardíacos/fisiopatologia , Oxigenoterapia Hiperbárica/efeitos adversos , Músculo Esquelético/fisiopatologia , Respiração , Animais , Fibrose , Força da Mão , Coração/fisiopatologia , Hemodinâmica , Hiperóxia/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Condicionamento Físico Animal , Pletismografia , Pressão
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa