Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 177(6): 1507-1521.e16, 2019 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-31031004

RESUMO

Friedreich's ataxia (FRDA) is a devastating, multisystemic disorder caused by recessive mutations in the mitochondrial protein frataxin (FXN). FXN participates in the biosynthesis of Fe-S clusters and is considered to be essential for viability. Here we report that when grown in 1% ambient O2, FXN null yeast, human cells, and nematodes are fully viable. In human cells, hypoxia restores steady-state levels of Fe-S clusters and normalizes ATF4, NRF2, and IRP2 signaling events associated with FRDA. Cellular studies and in vitro reconstitution indicate that hypoxia acts through HIF-independent mechanisms that increase bioavailable iron as well as directly activate Fe-S synthesis. In a mouse model of FRDA, breathing 11% O2 attenuates the progression of ataxia, whereas breathing 55% O2 hastens it. Our work identifies oxygen as a key environmental variable in the pathogenesis associated with FXN depletion, with important mechanistic and therapeutic implications.


Assuntos
Hipóxia/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Feminino , Ataxia de Friedreich/metabolismo , Células HEK293 , Humanos , Hipóxia/fisiopatologia , Ferro/metabolismo , Proteína 2 Reguladora do Ferro/metabolismo , Proteínas de Ligação ao Ferro/fisiologia , Proteínas Ferro-Enxofre/fisiologia , Células K562 , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Saccharomyces cerevisiae/metabolismo , Enxofre/metabolismo , Frataxina
2.
Vascular ; : 17085381231162733, 2023 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-36888982

RESUMO

BACKGROUND: Half of those undergoing major lower limb amputation for peripheral arterial disease die within 1 year. Advance care planning reduces days in hospital and increases the chance of dying in a preferred place. AIM: To investigate the prevalence and content of advance care planning for people having a lower limb amputation due to acute or chronic limb-threatening ischaemia or diabetes. Secondary aims were to explore its association with mortality, and length of hospital stay. DESIGN: A retrospective observational cohort study. The intervention was advance care planning. SETTING/PARTICIPANTS: Patients admitted to the South West England Major Arterial Centre between 1 January 2019 and 1 January 2021 who received unilateral or bilateral below, above, or through knee amputation due to acute or chronic limb-threatening ischaemia or diabetes. RESULTS: 116 patients were included in the study. 20.7% (n = 24) died within 1 year. 40.5% (n = 47) had an advance care planning discussion of which all included cardiopulmonary resuscitation decisions with few exploring other options. Patients who were more likely to have advance care planning discussions were ≥75 years (aOR = 5.58, 95%CI 1.56-20.0), female (aOR = 3.24, 95%CI 1.21-8.69), and had multimorbidity (Charlson Comorbidity Index ≥5, aOR = 2.97, 95%CI 1.11-7.92). Discussions occurred more often in the emergency pathway and were predominantly initiated by physicians. Advance care planning was associated with increased mortality (aHR = 2.63, 95%CI 1.01, 5.02) and longer hospital stay (aHR = 0.52, 95%CI 0.32-0.83). CONCLUSIONS: Despite a high risk of death for all patients in the months following amputation, advance care planning occurred in fewer than half of people and mostly focused on resuscitation.

3.
Hum Mol Genet ; 29(13): 2162-2170, 2020 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-32472139

RESUMO

Laminin-α2 related congenital muscular dystrophy (LAMA2-CMD) is a fatal muscle disease caused by mutations in the LAMA2 gene. Laminin-α2 is critical for the formation of laminin-211 and -221 heterotrimers in the muscle basal lamina. LAMA2-CMD patients exhibit hypotonia from birth and progressive muscle loss that results in developmental delay, confinement to a wheelchair, respiratory insufficiency and premature death. There is currently no cure or effective treatment for LAMA2-CMD. Several studies have shown laminin-111 can serve as an effective protein-replacement therapy for LAMA2-CMD. Studies have demonstrated early treatment with laminin-111 protein results in an increase in life expectancy and improvements in muscle pathology and function. Since LAMA2-CMD patients are often diagnosed after advanced disease, it is unclear if laminin-111 protein therapy at an advanced stage of the disease can have beneficial outcomes. In this study, we tested the efficacy of laminin-111 protein therapy after disease onset in a mouse model of LAMA2-CMD. Our results showed laminin-111 treatment after muscle disease onset increased life expectancy, promoted muscle growth and increased muscle stiffness. Together these studies indicate laminin-111 protein therapy either early or late in the disease process could serve as an effective protein replacement therapy for LAMA2-CMD.


Assuntos
Laminina/farmacologia , Doenças Musculares/genética , Distrofias Musculares/genética , Animais , Membrana Basal/efeitos dos fármacos , Membrana Basal/crescimento & desenvolvimento , Modelos Animais de Doenças , Humanos , Laminina/genética , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/crescimento & desenvolvimento , Doenças Musculares/patologia , Distrofias Musculares/patologia , Mutação/genética
4.
Arch Toxicol ; 96(12): 3363-3371, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36195745

RESUMO

Electronic cigarettes (e-cigarettes) have been used widely as an alternative to conventional cigarettes and have become particularly popular among young adults. A growing body of evidence has shown that e-cigarettes are associated with acute lung injury and adverse effects in multiple other organs. Previous studies showed that high emissions of aldehydes (formaldehyde and acetaldehyde) in aerosols were associated with increased usage of the same e-cigarette coils. However, the impact on lung function of using aged coils has not been reported. We investigated the relationship between coil age and acute lung injury in mice exposed to experimental vaping for 1 h (2 puffs/min, 100 ml/puff). The e-liquid contains propylene glycol and vegetable glycerin (50:50, vol) only. The concentrations of formaldehyde and acetaldehyde in the vaping aerosols increased with age of the nichrome coils starting at 1200 puffs. Mice exposed to e-cigarette aerosols produced from 1800, but not 0 or 900, puff-aged coils caused acute lung injury, increased lung wet/dry weight ratio, and induced lung inflammation (IL-6, TNF-α, IL-1ß, MIP-2). Exposure to vaping aerosols from 1800 puff-aged coils decreased heart rate, respiratory rate, and oxygen saturation in mice compared to mice exposed to air or aerosols from new coils. In conclusion, we observed that the concentration of aldehydes (formaldehyde and acetaldehyde) increased with repeated and prolonged usage of e-cigarette coils. Exposure to high levels of aldehyde in vaping aerosol was associated with acute lung injury in mice. These findings show significant risk of lung injury associated with prolonged use of e-cigarette devices.


Assuntos
Lesão Pulmonar Aguda , Sistemas Eletrônicos de Liberação de Nicotina , Vaping , Animais , Camundongos , Acetaldeído , Lesão Pulmonar Aguda/induzido quimicamente , Aldeídos/toxicidade , Formaldeído/toxicidade , Glicerol , Interleucina-6 , Propilenoglicol/toxicidade , Aerossóis e Gotículas Respiratórios , Fator de Necrose Tumoral alfa
5.
Mol Genet Metab ; 133(1): 83-93, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33752971

RESUMO

Leigh syndrome is a severe mitochondrial neurodegenerative disease with no effective treatment. In the Ndufs4-/- mouse model of Leigh syndrome, continuously breathing 11% O2 (hypoxia) prevents neurodegeneration and leads to a dramatic extension (~5-fold) in lifespan. We investigated the effect of hypoxia on the brain metabolism of Ndufs4-/- mice by studying blood gas tensions and metabolite levels in simultaneously sampled arterial and cerebral internal jugular venous (IJV) blood. Relatively healthy Ndufs4-/- and wildtype (WT) mice breathing air until postnatal age ~38 d were compared to Ndufs4-/- and WT mice breathing air until ~38 days old followed by 4-weeks of breathing 11% O2. Compared to WT control mice, Ndufs4-/- mice breathing air have reduced brain O2 consumption as evidenced by an elevated partial pressure of O2 in IJV blood (PijvO2) despite a normal PO2 in arterial blood, and higher lactate/pyruvate (L/P) ratios in IJV plasma revealed by metabolic profiling. In Ndufs4-/- mice, hypoxia treatment normalized the cerebral venous PijvO2 and L/P ratios, and decreased levels of nicotinate in IJV plasma. Brain concentrations of nicotinamide adenine dinucleotide (NAD+) were lower in Ndufs4-/- mice breathing air than in WT mice, but preserved at WT levels with hypoxia treatment. Although mild hypoxia (17% O2) has been shown to be an ineffective therapy for Ndufs4-/- mice, we find that when combined with nicotinic acid supplementation it provides a modest improvement in neurodegeneration and lifespan. Therapies targeting both brain hyperoxia and NAD+ deficiency may hold promise for treating Leigh syndrome.


Assuntos
Encéfalo/metabolismo , Complexo I de Transporte de Elétrons/genética , Doença de Leigh/metabolismo , NAD/genética , Oxigênio/metabolismo , Animais , Encéfalo/patologia , Hipóxia Celular/fisiologia , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/metabolismo , Humanos , Doença de Leigh/genética , Doença de Leigh/terapia , Metabolômica , Camundongos , Mitocôndrias , NAD/deficiência , Doenças Neurodegenerativas , Respiração/genética
6.
Connect Tissue Res ; 62(1): 40-52, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32867551

RESUMO

PURPOSE: To describe potential signaling (cross-talk) between dystrophic skeletal muscle and tendon in Duchenne muscular dystrophy. MATERIALS AND METHODS: Review of Duchenne muscular dystrophy and associated literature relevant to muscle-tendon cross-talk. RESULTS AND CONCLUSIONS: Duchenne muscular dystrophy results from the absence of the protein dystrophin and the associated dystrophin - glycoprotein complex, which are thought to provide both structural support and signaling functions for the muscle fiber. In addition, there are other potential signal pathways that could represent cross-talk between muscle and tendon, particularly at the myotendinous junction. Duchenne muscular dystrophy is characterized by multiple pathophysiologic mechanisms. Herein, we explore three of these: (1) the extracellular matrix, fibrosis, and fat deposition; (2) satellite cells; and (3) tensegrity. A key signaling protein that emerged in each was transforming growth factor - beta one (TGF-ß1).].


Assuntos
Distrofia Muscular de Duchenne , Distrofina , Humanos , Músculo Esquelético , Tendões , Fator de Crescimento Transformador beta
7.
Mol Ther ; 28(2): 382-393, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-31784415

RESUMO

Multiple clinical trials employing recombinant adeno-associated viral (rAAV) vectors have been initiated for neuromuscular disorders, including Duchenne and limb-girdle muscular dystrophies, spinal muscular atrophy, and recently X-linked myotubular myopathy (XLMTM). Our previous work on a canine model of XLMTM showed that a single rAAV8-cMTM1 systemic infusion corrected structural abnormalities within the muscle and restored contractile function, with affected dogs surviving more than 4 years post injection. This remarkable therapeutic efficacy presents a unique opportunity to identify the downstream molecular drivers of XLMTM pathology and to what extent the whole muscle transcriptome is restored to normal after gene transfer. Herein, RNA-sequencing was used to examine the transcriptomes of the Biceps femoris and Vastus lateralis in a previously described canine cohort that showed dose-dependent clinical improvements after rAAV8-cMTM1 gene transfer. Our analysis confirmed several dysregulated genes previously observed in XLMTM mice but also identified transcripts linked to XLMTM pathology. We demonstrated XLMTM transcriptome remodeling and dose-dependent normalization of gene expression after gene transfer and created metrics to pinpoint potential biomarkers of disease progression and correction.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Músculo Esquelético/metabolismo , Miopatias Congênitas Estruturais/genética , Transcriptoma , Animais , Biomarcadores , Modelos Animais de Doenças , Cães , Dosagem de Genes , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Transdução Genética
8.
Exerc Sport Sci Rev ; 48(2): 74-82, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32168170

RESUMO

Hypothalamic pro-opiomelanocortin (POMC) neurons are key sensory neurons for energy balance. The basic helix-loop-helix transcription factor NHLH2 is expressed in POMC neurons, and Nhlh2 knockout mice show adult-onset obesity with low exercise behavior. Evidence is presented to explore the hypothesis that NHLH2 transcriptional activity within POMC neurons is crucial for maintaining motivated spontaneous activity and enforced exercise.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Exercício Físico/fisiologia , Hipotálamo/metabolismo , Motivação/fisiologia , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Transcrição Gênica , Animais , Exercício Físico/psicologia , Humanos , Modelos Animais
9.
J Mol Cell Cardiol ; 135: 160-171, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31445917

RESUMO

Novel therapeutic strategies to treat mitochondrial deficiencies in acute coronary syndromes are needed. Complex I of the mitochondrial electron transport system is damaged following ischemia/reperfusion (I/R) injury. This disruption contributes to aberrant electron transport, diminished bioenergetics, an altered redox environment, and mitochondrial damage involved in tissue injury. In this study, we determined the cardiac and mitochondrial effects of idebenone, a benzoquinone currently in several clinical trials with purported 'antioxidant' effects. We employed complimentary models of ischemia/reperfusion injury in perfused hearts, permeabilized cardiac fibers, isolated mitochondria, and in cells to elucidate idebenone's cardioprotective mechanism(s). In ex vivo whole hearts, infarct size was markedly reduced with post-ischemic idebenone treatment (25 ±â€¯5% area at risk, AAR) compared to controls (56 ±â€¯6% AAR, P < .05). Several parameters of hemodynamic function were also significantly improved after idebenone treatment. Parallel studies of anoxia/reoxygenation were conducted using isolated mitochondria and permeabilized ventricular fibers. In isolated mitochondria, we simultaneously monitored respiration and ROS emission. Idebenone treatment modestly elevated succinate-derived H2O2 production when compared to vehicle control (1.34 ±â€¯0.05 vs 1.21 ±â€¯0.05%, H2O2/O2 respectively, P < .05). Isolated mitochondria subjected to anoxia/reoxygenation demonstrated higher rates of respiration with idebenone treatment (2360 ±â€¯69 pmol/s*mg) versus vehicle control (1995 ±â€¯101 pmol/s*mg). Both mitochondria and permeabilized cardiac fibers produced high rates of H2O2 after anoxia/reoxygenation, with idebenone showing no discernable attenuation on H2O2 production. These insights were further investigated with studies in mitochondria isolated from reperfused ventricle. The profound decrease in complex-I dependent respiration after ischemia/reperfusion (701 ±â€¯59 pmolO2/s*mg compared to 1816 ±â€¯105 pmol O2/s*mg in normoxic mitochondria) was attenuated with idebenone treatment (994 ±â€¯76 vs pmol O2/s*mg, P < .05). Finally, the effects of idebenone were determined using permeabilized cell models with chemical inhibition of complex I. ADP-dependent oxidative phosphorylation capacity was significantly higher in complex-I inhibited cells treated acutely with idebenone (89.0 ±â€¯4.2 pmol/s*million cells versus 70.1 ±â€¯8.2 pmol/s*million cells in untreated cells). Taken together, these data indicate that the cardioprotective effects of idebenone treatment do not involve ROS-scavenging but appear to involve augmentation of the quinone pool, thus providing reducing equivalents downstream of complex I. As this compound is already in clinical trials for other indications, it may provide a safe and useful approach to mitigate ischemia/reperfusion injury in patients.


Assuntos
Complexo I de Transporte de Elétrons/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Traumatismo por Reperfusão/tratamento farmacológico , Ubiquinona/análogos & derivados , Animais , Modelos Animais de Doenças , Complexo I de Transporte de Elétrons/genética , Humanos , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/patologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fosforilação Oxidativa/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Ubiquinona/farmacologia
10.
Am J Physiol Lung Cell Mol Physiol ; 316(2): L391-L399, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30520688

RESUMO

Hypoxic pulmonary vasoconstriction (HPV) is a physiological vasomotor response that maintains systemic oxygenation by matching perfusion to ventilation during alveolar hypoxia. Although mitochondria appear to play an essential role in HPV, the impact of mitochondrial dysfunction on HPV remains incompletely defined. Mice lacking the mitochondrial complex I (CI) subunit Ndufs4 ( Ndufs4-/-) develop a fatal progressive encephalopathy and serve as a model for Leigh syndrome, the most common mitochondrial disease in children. Breathing normobaric 11% O2 prevents neurological disease and improves survival in Ndufs4-/- mice. In this study, we found that either genetic Ndufs4 deficiency or pharmacological inhibition of CI using piericidin A impaired the ability of left mainstem bronchus occlusion (LMBO) to induce HPV. In mice breathing air, the partial pressure of arterial oxygen during LMBO was lower in Ndufs4-/- and in piericidin A-treated Ndufs4+/+ mice than in respective controls. Impairment of HPV in Ndufs4-/- mice was not a result of nonspecific dysfunction of the pulmonary vascular contractile apparatus or pulmonary inflammation. In Ndufs4-deficient mice, 3 wk of breathing 11% O2 restored HPV in response to LMBO. When compared with Ndufs4-/- mice breathing air, chronic hypoxia improved systemic oxygenation during LMBO. The results of this study show that, when breathing air, mice with a congenital Ndufs4 deficiency or chemically inhibited CI function have impaired HPV. Our study raises the possibility that patients with inborn errors of mitochondrial function may also have defects in HPV.


Assuntos
Complexo I de Transporte de Elétrons/deficiência , Hipóxia/fisiopatologia , Doença de Leigh/fisiopatologia , Vasoconstrição/fisiologia , Animais , Brônquios/metabolismo , Modelos Animais de Doenças , Hipóxia/metabolismo , Pulmão/metabolismo , Pulmão/fisiopatologia , Camundongos Transgênicos , Mitocôndrias/metabolismo , Artéria Pulmonar/metabolismo , Circulação Pulmonar/fisiologia
11.
Mol Ther ; 25(4): 839-854, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28237839

RESUMO

X-linked myotubular myopathy (XLMTM) results from MTM1 gene mutations and myotubularin deficiency. Most XLMTM patients develop severe muscle weakness leading to respiratory failure and death, typically within 2 years of age. Our objective was to evaluate the efficacy and safety of systemic gene therapy in the p.N155K canine model of XLMTM by performing a dose escalation study. A recombinant adeno-associated virus serotype 8 (rAAV8) vector expressing canine myotubularin (cMTM1) under the muscle-specific desmin promoter (rAAV8-cMTM1) was administered by simple peripheral venous infusion in XLMTM dogs at 10 weeks of age, when signs of the disease are already present. A comprehensive analysis of survival, limb strength, gait, respiratory function, neurological assessment, histology, vector biodistribution, transgene expression, and immune response was performed over a 9-month study period. Results indicate that systemic gene therapy was well tolerated, prolonged lifespan, and corrected the skeletal musculature throughout the body in a dose-dependent manner, defining an efficacious dose in this large-animal model of the disease. These results support the development of gene therapy clinical trials for XLMTM.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/genética , Músculo Esquelético/metabolismo , Miopatias Congênitas Estruturais/genética , Animais , Biópsia , Dependovirus/classificação , Modelos Animais de Doenças , Progressão da Doença , Cães , Marcha , Expressão Gênica , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Vetores Genéticos/farmacocinética , Imunidade Celular , Imunidade Humoral , Estimativa de Kaplan-Meier , Força Muscular , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Músculo Esquelético/ultraestrutura , Miopatias Congênitas Estruturais/diagnóstico , Miopatias Congênitas Estruturais/mortalidade , Miopatias Congênitas Estruturais/terapia , Proteínas Tirosina Fosfatases não Receptoras/genética , Recuperação de Função Fisiológica , Reflexo , Testes de Função Respiratória , Distribuição Tecidual , Transgenes/genética , Transgenes/imunologia , Resultado do Tratamento
12.
Muscle Nerve ; 56(5): 943-953, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28370029

RESUMO

INTRODUCTION: X-linked myotubular myopathy (XLMTM), a devastating pediatric disease caused by the absence of the protein myotubularin, results from mutations in the MTM1 gene. While there is no cure for XLMTM, we previously reported effects of MTM1 gene therapy using adeno-associated virus (AAV) vector on muscle weakness and pathology in MTM1-mutant dogs. Here, we followed 2 AAV-infused dogs over 4 years. METHODS: We evaluated gait, strength, respiration, neurological function, muscle pathology, AAV vector copy number (VCN), and transgene expression. RESULTS: Four years following AAV-mediated gene therapy, gait, respiratory performance, neurological function and pathology in AAV-infused XLMTM dogs remained comparable to their healthy littermate controls despite a decline in VCN and muscle strength. CONCLUSIONS: AAV-mediated gene transfer of MTM1 in young XLMTM dogs results in long-term expression of myotubularin transgene with normal muscular performance and neurological function in the absence of muscle pathology. These findings support a clinical trial in patients. Muscle Nerve 56: 943-953, 2017.


Assuntos
Terapia Genética , Miopatias Congênitas Estruturais/terapia , Proteínas Tirosina Fosfatases não Receptoras/uso terapêutico , Adenosina Trifosfatases/metabolismo , Animais , Dependovirus/genética , Modelos Animais de Doenças , Cães , Feminino , Transtornos Neurológicos da Marcha/etiologia , Glucuronidase/genética , Glucuronidase/metabolismo , Humanos , Estudos Longitudinais , Microscopia Eletrônica , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Mutação/genética , Miopatias Congênitas Estruturais/complicações , Miopatias Congênitas Estruturais/genética , Miopatias Congênitas Estruturais/veterinária , NAD/metabolismo , Exame Neurológico , Proteínas Tirosina Fosfatases não Receptoras/genética , Transtornos Respiratórios/etiologia , Transdução Genética
13.
Ann Neurol ; 78(2): 222-34, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25925726

RESUMO

OBJECTIVE: We have recently reported on the pathology of the neuromuscular junction (NMJ) in Pompe disease, reflecting disruption of neuronal and muscle homeostasis as a result of glycogen accumulation. The aim of this study was to examine how the alteration of NMJ physiology contributes to Pompe disease pathology; we performed molecular, physiological, and histochemical analyses of NMJ-related measures of the tibialis anterior muscles of young-, mid-, and late-stage alpha-glucosidase (GAA)-deficient mice. METHODS: We performed intramuscular injection of an adeno-associated virus (AAV)9 vector expressing GAA (AAV9-hGAA) into the tibialis anterior muscle of Gaa(-/-) mice at early, mid, and severe pathological time points. We analyzed expression of NMJ-related genes, in situ muscle force production, and clearance of glycogen in conjunction with histological assessment of the NMJ. RESULTS: Our data demonstrate that AAV9-hGAA is able to replace GAA to the affected tissue and modify AChR mRNA expression, muscle force production, motor endplate area, and innervation status. Importantly, the degree of restoration for these outcomes is limited by severity of disease. Early restoration of GAA activity was most effective, whereas late correction of GAA expression was not effective in modifying parameters reflecting NMJ structure and function nor in force restoration despite resolution of glycogen storage in muscle. INTERPRETATION: Our data provide new mechanistic insight into the pathology of Pompe disease and suggest that early systemic correction to both neural and muscle tissues may be essential for successful correction of neuromuscular function in Pompe disease. Ann Neurol 2015;78:222-234.


Assuntos
Terapia Genética , Doença de Depósito de Glicogênio Tipo II/genética , Força Muscular/fisiologia , Músculo Esquelético/metabolismo , Junção Neuromuscular/metabolismo , RNA Mensageiro/metabolismo , Receptores Colinérgicos/genética , alfa-Glucosidases/genética , Animais , Dependovirus , Modelos Animais de Doenças , Vetores Genéticos , Glicogênio/metabolismo , Doença de Depósito de Glicogênio Tipo II/metabolismo , Doença de Depósito de Glicogênio Tipo II/fisiopatologia , Membro Posterior , Injeções Intramusculares , Contração Isométrica , Camundongos , Camundongos Knockout , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Junção Neuromuscular/patologia , Junção Neuromuscular/fisiopatologia , Receptores Colinérgicos/metabolismo , Fatores de Tempo
14.
Am J Physiol Cell Physiol ; 308(1): C33-40, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25298424

RESUMO

Duchenne muscular dystrophy (DMD) is one of the most frequent types of muscular dystrophy. Alterations in intracellular calcium (Ca(2+)) handling are thought to contribute to the disease severity in DMD, possibly due to the activation of Ca(2+)-activated proteases. The purpose of this study was twofold: 1) to determine whether prolonged excitation-contraction (E-C) coupling disruption following repeated contractions is greater in animals lacking both dystrophin and utrophin (mdx/Utr(-/-)) compared with mice lacking only dystrophin (mdx); and 2) to assess whether protease inhibition can prevent E-C coupling failure following repeated tetani in these dystrophic mouse models. Excitation-contraction coupling was assessed using Fura-2 ratio, as an index of intracellular free Ca(2+) concentration, in response to electrical stimulation of single muscle fibers from the flexor digitorum brevis muscle. Resting Fura-2 ratio was higher in dystrophic compared with control (Con) fibers, but peak Fura-2 ratios during stimulation were similar in dystrophic and Con fibers. One hour after a series of repeated tetani, peak Fura-2 ratios were reduced by 30 ± 5.6%, 23 ± 2%, and 36 ± 3.1% in mdx, mdx/Utr(+/-), and mdx/Utr(-/-), respectively, with the greatest reduction in mdx/Utr(-/-) fibers (P < 0.05). Protease inhibition attenuated this decrease in peak Fura-2 ratio. These data indicate that E-C coupling impairment after repeated contractions is greatest in fibers lacking both dystrophin and utrophin and that prevention of protease activation can mitigate the prolonged E-C coupling impairment. These data further suggest that acute protease inhibition may be useful in reducing muscle weakness in DMD.


Assuntos
Cálcio/metabolismo , Acoplamento Excitação-Contração , Fibras Musculares Esqueléticas/enzimologia , Músculo Esquelético/enzimologia , Distrofia Muscular de Duchenne/enzimologia , Peptídeo Hidrolases/metabolismo , Animais , Modelos Animais de Doenças , Estimulação Elétrica , Acoplamento Excitação-Contração/efeitos dos fármacos , Camundongos Endogâmicos mdx , Camundongos Knockout , Fibras Musculares Esqueléticas/efeitos dos fármacos , Força Muscular , Debilidade Muscular , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Inibidores de Proteases/farmacologia , Fatores de Tempo , Utrofina/deficiência , Utrofina/genética
15.
Hum Mol Genet ; 22(8): 1525-38, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23307925

RESUMO

No effective treatment exists for patients with X-linked myotubular myopathy (XLMTM), a fatal congenital muscle disease caused by deficiency of the lipid phosphatase, myotubularin. The Mtm1δ4 and Mtm1 p.R69C mice model severely and moderately symptomatic XLMTM, respectively, due to differences in the degree of myotubularin deficiency. Contractile function of intact extensor digitorum longus (EDL) and soleus muscles from Mtm1δ4 mice, which produce no myotubularin, is markedly impaired. Contractile forces generated by chemically skinned single fiber preparations from Mtm1δ4 muscle were largely preserved, indicating that weakness was largely due to impaired excitation contraction coupling. Mtm1 p.R69C mice, which produce small amounts of myotubularin, showed impaired contractile function only in EDL muscles. Short-term replacement of myotubularin with a prototypical targeted protein replacement agent (3E10Fv-MTM1) in Mtm1δ4 mice improved contractile function and muscle pathology. These promising findings suggest that even low levels of myotubularin protein replacement can improve the muscle weakness and reverse the pathology that characterizes XLMTM.


Assuntos
Terapia de Reposição de Enzimas , Miopatias Congênitas Estruturais/patologia , Miopatias Congênitas Estruturais/terapia , Proteínas Tirosina Fosfatases não Receptoras/genética , Animais , Modelos Animais de Doenças , Fadiga/metabolismo , Fadiga/fisiopatologia , Feminino , Humanos , Camundongos , Debilidade Muscular/genética , Debilidade Muscular/terapia , Músculo Esquelético/fisiopatologia , Músculos/enzimologia , Músculos/metabolismo , Músculos/patologia , Miopatias Congênitas Estruturais/enzimologia , Miopatias Congênitas Estruturais/genética , Proteínas Tirosina Fosfatases não Receptoras/biossíntese , Proteínas Tirosina Fosfatases não Receptoras/deficiência
16.
J Cell Sci ; 125(Pt 22): 5329-37, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22956541

RESUMO

In response to severe injury, adult skeletal muscle exhibits a remarkable regenerative capacity due to a resident muscle stem/progenitor cell population. While a number of factors are expressed in the muscle progenitor cell (MPC) population, the molecular networks that govern this cell population remain an area of active investigation. In this study, utilizing knockdown techniques and overexpression of Foxk1 in the myogenic lineage, we observed dysregulation of Foxo and Mef2 downstream targets. Utilizing an array of technologies, we establish that Foxk1 represses the transcriptional activity of Foxo4 and Mef2 and physically interacts with Foxo4 and Mef2, thus promoting MPC proliferation and antagonizing the myogenic lineage differentiation program, respectively. Correspondingly, knockdown of Foxk1 in C2C12 myoblasts results in cell cycle arrest, and Foxk1 overexpression in C2C12CAR myoblasts retards muscle differentiation. Collectively, we have established that Foxk1 promotes MPC proliferation by repressing Foxo4 transcriptional activity and inhibits myogenic differentiation by repressing Mef2 activity. These studies enhance our understanding of the transcriptional networks that regulate the MPC population and muscle regeneration.


Assuntos
Diferenciação Celular , Fatores de Transcrição Forkhead/metabolismo , Desenvolvimento Muscular , Fatores de Regulação Miogênica/metabolismo , Animais , Ciclo Celular , Proteínas de Ciclo Celular , Proliferação de Células , DNA/metabolismo , Fatores de Transcrição MEF2 , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Ligação Proteica , Regeneração , Proteínas Repressoras/metabolismo , Transcrição Gênica
17.
FASEB J ; 27(4): 1585-99, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23325319

RESUMO

Mutations in the human SEPN1 gene, encoding selenoprotein N (SepN), cause SEPN1-related myopathy (SEPN1-RM) characterized by muscle weakness, spinal rigidity, and respiratory insufficiency. As with other members of the selenoprotein family, selenoprotein N incorporates selenium in the form of selenocysteine (Sec). Most selenoproteins that have been functionally characterized are involved in oxidation-reduction (redox) reactions, with the Sec residue located at their catalytic site. To model SEPN1-RM, we generated a Sepn1-knockout (Sepn1(-/-)) mouse line. Homozygous Sepn1(-/-) mice are fertile, and their weight and lifespan are comparable to wild-type (WT) animals. Under baseline conditions, the muscle histology of Sepn1(-/-) mice remains normal, but subtle core lesions could be detected in skeletal muscle after inducing oxidative stress. Ryanodine receptor (RyR) calcium release channels showed lower sensitivity to caffeine in SepN deficient myofibers, suggesting a possible role of SepN in RyR regulation. SepN deficiency also leads to abnormal lung development characterized by enlarged alveoli, which is associated with decreased tissue elastance and increased quasi-static compliance of Sepn1(-/-) lungs. This finding raises the possibility that the respiratory syndrome observed in patients with SEPN1 mutations may have a primary pulmonary component in addition to the weakness of respiratory muscles.


Assuntos
Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Doenças Musculares/genética , Selenoproteínas/deficiência , Animais , Humanos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mutação/genética , Estresse Oxidativo/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Selenocisteína/genética , Selenoproteínas/metabolismo
18.
Br J Nutr ; 111(10): 1791-800, 2014 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-24524266

RESUMO

Duchenne muscular dystrophy (DMD) is a severe muscle disease that affects afflicted males from a young age, and the mdx mouse is an animal model of this disease. Although new drugs are in development, it is also essential to assess potential dietary therapies that could assist in the management of DMD. In the present study, we compared two diets, high-MUFA diet v. high-PUFA diet, in mdx mice. To generate the high-PUFA diet, a portion of dietary MUFA (oleic acid) was replaced with the dietary essential n-3 PUFA α-linolenic acid (ALA). We sought to determine whether ALA, compared with oleic acid, was beneficial in mdx mice. Consumption of the high-PUFA diet resulted in significantly higher n-3 PUFA content and reduced arachidonic acid content in skeletal muscle phospholipids (PL), while the high-MUFA diet led to higher oleate content in PL. Mdx mice on the high-MUFA diet exhibited 2-fold lower serum creatine kinase activity than those on the high-PUFA diet (P< 0·05) as well as a lower body fat percentage (P< 0·05), but no significant difference in skeletal muscle histopathology results. There was no significant difference between the dietary groups with regard to phosphorylated p65 (an inflammatory marker) in skeletal muscle. In conclusion, alteration of PL fatty acid (FA) composition by the high-PUFA diet made mdx muscle more susceptible to sarcolemmal leakiness, while the high-MUFA diet exhibited a more favourable impact. These results may be important for designing dietary treatments for DMD patients, and future work on dietary FA profiles, such as comparing other FA classes and dose effects, is needed.


Assuntos
Creatina Quinase/sangue , Gorduras na Dieta/metabolismo , Ácidos Graxos Monoinsaturados/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/patologia , Fosfolipídeos/isolamento & purificação , Análise de Variância , Animais , Ácido Araquidônico/metabolismo , Biomarcadores/metabolismo , Cromatografia Líquida , Modelos Animais de Doenças , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/patologia , NF-kappa B/análise , Ácido Oleico/metabolismo , Fosforilação , Óleos de Plantas/metabolismo
19.
Proc Natl Acad Sci U S A ; 108(25): 10196-201, 2011 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-21633012

RESUMO

In response to physiological stimuli, skeletal muscle alters its myofiber composition to significantly affect muscle performance and metabolism. This process requires concerted regulation of myofiber-specific isoforms of sarcomeric and calcium regulatory proteins that couple action potentials to the generation of contractile force. Here, we identify Sox6 as a fast myofiber-enriched repressor of slow muscle gene expression in vivo. Mice lacking Sox6 specifically in skeletal muscle have an increased number of slow myofibers, elevated mitochondrial activity, and exhibit down-regulation of the fast myofiber gene program, resulting in enhanced muscular endurance. In addition, microarray profiling of Sox6 knockout muscle revealed extensive muscle fiber-type remodeling, and identified numerous genes that display distinctive fiber-type enrichment. Sox6 directly represses the transcription of slow myofiber-enriched genes by binding to conserved cis-regulatory elements. These results identify Sox6 as a robust regulator of muscle contractile phenotype and metabolism, and elucidate a mechanism by which functionally related muscle fiber-type specific gene isoforms are collectively controlled.


Assuntos
Regulação da Expressão Gênica , Fibras Musculares de Contração Rápida/fisiologia , Fibras Musculares de Contração Lenta/fisiologia , Músculo Esquelético/fisiologia , Fatores de Transcrição SOXD/metabolismo , Animais , Cálcio/metabolismo , Perfilação da Expressão Gênica , Camundongos , Camundongos Knockout , Análise em Microsséries , Contração Muscular/fisiologia , Fibras Musculares de Contração Rápida/citologia , Fibras Musculares de Contração Lenta/citologia , Músculo Esquelético/citologia , Fatores de Transcrição SOXD/genética
20.
Function (Oxf) ; 5(1): zqad066, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38111538

RESUMO

Alzheimer's disease (AD) develops along a continuum that spans years prior to diagnosis. Decreased muscle function and mitochondrial respiration occur years earlier in those that develop AD; however, it is unknown what causes these peripheral phenotypes in a disease of the brain. Exercise promotes muscle, mitochondria, and cognitive health and is proposed to be a potential therapeutic for AD, but no study has investigated how skeletal muscle adapts to exercise training in an AD-like context. Utilizing 5xFAD mice, an AD model that develops ad-like pathology and cognitive impairments around 6 mo of age, we examined in vivo neuromuscular function and exercise adapations (mitochondrial respiration and RNA sequencing) before the manifestation of overt cognitive impairment. We found 5xFAD mice develop neuromuscular dysfunction beginning as early as 4 mo of age, characterized by impaired nerve-stimulated muscle torque production and compound nerve action potential of the sciatic nerve. Furthermore, skeletal muscle in 5xFAD mice had altered, sex-dependent, adaptive responses (mitochondrial respiration and gene expression) to exercise training in the absence of overt cognitive impairment. Changes in peripheral systems, specifically neural communication to skeletal muscle, may be harbingers for AD and have implications for lifestyle interventions, like exercise, in AD.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Camundongos , Animais , Doença de Alzheimer/genética , Camundongos Transgênicos , Encéfalo/metabolismo , Disfunção Cognitiva/etiologia , Mitocôndrias/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA