Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.959
Filtrar
Más filtros











Intervalo de año de publicación
1.
Physiol Res ; 73(2): 285-294, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38710059

RESUMEN

This study aimed to determine whether electrical stimulation-based twitch exercise is effective in inhibiting the progression of immobilization-induced muscle fibrosis. 19 Wistar rats were randomly divided into a control group (n=6), an immobilization group (n=6; with immobilization only), and a Belt group (n=7; with immobilization and twitch exercise through the belt electrode device, beginning 2 weeks after immobilization). The bilateral soleus muscles were harvested after the experimental period. The right soleus muscles were used for histological analysis, and the left soleus muscles were used for biochemical and molecular biological analysis. As a result, in the picrosirius red images, the perimysium and endomysium were thicker in both the immobilization and Belt groups compared to the control group. However, the perimysium and endomysium thickening were suppressed in the Belt group. The hydroxyproline content and alpha-SMA, TGF-beta1, and HIF-1alpha mRNA expressions were significantly higher in the immobilization and belt groups than in the control group. These expressions were significantly lower in the Belt group than in the immobilization group. The capillary-to-myofiber ratio and the mRNA expressions of VEGF and PGC-1alpha were significantly lower in the immobilization and belt groups than in the control group, these were significantly higher in the Belt group than in the immobilization group. From these results, Electrical stimulation-based twitch exercise using the belt electrode device may prevent the progression of immobilization-induced muscle fibrosis caused by downregulating PGC-1alpha/VEGF pathway, we surmised that this intervention strategy might be effective against the progression of muscle contracture. Keywords: Immobilization, Skeletal muscle, Fibrosis, Electrical stimulation-based twitch exercise, PGC-1alpha/VEGF pathway.


Asunto(s)
Regulación hacia Abajo , Fibrosis , Músculo Esquelético , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Condicionamiento Físico Animal , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular , Animales , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Masculino , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Ratas , Condicionamiento Físico Animal/fisiología , Transducción de Señal/fisiología , Estimulación Eléctrica , Terapia por Estimulación Eléctrica/métodos , Progresión de la Enfermedad , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Enfermedades Musculares/prevención & control , Enfermedades Musculares/etiología
2.
Sci Rep ; 14(1): 10554, 2024 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719903

RESUMEN

Sarcopenia greatly reduces the quality of life of the elderly, and iron metabolism plays an important role in muscle loss. This study aimed to investigate the association between iron status and sarcopenia. A total of 286 adult patients hospitalized between 2019 and 2021 were included in this study, of which 117 were diagnosed with sarcopenia. Serum iron, total iron binding capacity (TIBC), transferrin, and transferrin saturation levels were compared between groups with and without sarcopenia and were included in the logistic analyses, with significant variables further included in the logistic regression model for the prediction of sarcopenia. Serum iron, TIBC, and transferrin levels decreased significantly in the sarcopenia group (p < 0.05), and were negatively associated with handgrip strength, relative skeletal muscle index, and multiple test performances (p < 0.05). Multivariate logistic analysis showed that sex, age, body mass index (BMI), and serum iron level were independent risk factors for sarcopenia. In the final logistic regression model, male sex (odds ratio [OR] 3.65, 95% confidence interval [CI] 1.67-7.98), age > 65 years (OR 5.40, 95% CI 2.25-12.95), BMI < 24 kg/m2 (OR 0.17, 95% CI 0.08-0.36), and serum iron < 10.95 µmol/L (OR 0.39, 95% CI 0.16-0.93) were included. Our study supported the impact of iron metabolism on muscle strength and performance.


Asunto(s)
Hierro , Sarcopenia , Transferrina , Humanos , Sarcopenia/sangre , Masculino , Femenino , Hierro/sangre , Anciano , Persona de Mediana Edad , Estudios Retrospectivos , Transferrina/metabolismo , Transferrina/análisis , Índice de Masa Corporal , Fuerza de la Mano , Factores de Riesgo , Músculo Esquelético/metabolismo , Modelos Logísticos , Anciano de 80 o más Años
3.
PLoS One ; 19(5): e0298827, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38722949

RESUMEN

Glutathione peroxidase 2 (GPX2) is a selenium-dependent enzyme and protects cells against oxidative damage. Recently, GPX2 has been identified as a candidate gene for backfat and feed efficiency in pigs. However, it is unclear whether GPX2 regulates the development of porcine preadipocytes and skeletal muscle cells. In this study, adenoviral gene transfer was used to overexpress GPX2. Our findings suggest that overexpression of GPX2 gene inhibited proliferation of porcine preadipocytes. And the process is accompanied by the reduction of the p-p38. GPX2 inhibited adipogenic differentiation and promoted lipid degradation, while ERK1/2 was reduced and p-p38 was increased. Proliferation of porcine skeletal muscle cells was induced after GPX2 overexpression, was accompanied by activation in JNK, ERK1/2, and p-p38. Overexpression methods confirmed that GPX2 has a promoting function in myoblastic differentiation. ERK1/2 pathway was activated and p38 was suppressed during the process. This study lays a foundation for the functional study of GPX2 and provides theoretical support for promoting subcutaneous fat reduction and muscle growth.


Asunto(s)
Adipocitos , Glutatión Peroxidasa , Sistema de Señalización de MAP Quinasas , Animales , Glutatión Peroxidasa/metabolismo , Glutatión Peroxidasa/genética , Adipocitos/metabolismo , Adipocitos/citología , Porcinos , Diferenciación Celular/genética , Proliferación Celular , Adipogénesis/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/citología
4.
Sci Rep ; 14(1): 10631, 2024 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724633

RESUMEN

Higher fat-to-muscle mass ratio (FMR) is reported to be a risk factor for various diseases, including type 2 diabetes and cardiovascular diseases, and mortality. Although this association suggests that reducing FMR may help to prevent certain diseases and mortality, the relationship between FMR and lifestyle factors is unclear. Therefore, we performed a cross-sectional study with the aim to elucidate this relationship. This cross-sectional study included 1518 healthy Japanese adults aged 30 to 64 years. We measured FMR in the whole body, arms, legs, and trunk and assessed various lifestyle factors. Then, we performed forced entry multiple regression analyses for FMR with the following variables: sex, age, physical activity, dietary intake, sleep quality, cigarette smoking, stress levels, and body mass index. As a result, whole-body and regional FMRs were correlated with female sex (ß = 0.71); age (ß = 0.06); physical activity (ß = - 0.07); dietary intake of protein (ß = - 0.12), carbohydrate (ß = 0.04), sodium (ß = 0.13), and fiber (ß = - 0.16); and body mass index (ß = 0.70). The results suggest that in the Japanese middle-aged population, low FMR is associated with certain lifestyle factors, i.e. higher physical activity and a diet with higher protein and fiber and lower carbohydrate and sodium, independent of age, sex, and body mass index.


Asunto(s)
Carbohidratos de la Dieta , Fibras de la Dieta , Proteínas en la Dieta , Ejercicio Físico , Humanos , Masculino , Femenino , Estudios Transversales , Persona de Mediana Edad , Adulto , Ejercicio Físico/fisiología , Fibras de la Dieta/administración & dosificación , Proteínas en la Dieta/administración & dosificación , Carbohidratos de la Dieta/administración & dosificación , Índice de Masa Corporal , Japón , Músculo Esquelético/fisiología , Músculo Esquelético/metabolismo , Sodio en la Dieta/administración & dosificación , Tejido Adiposo/metabolismo , Composición Corporal , Estilo de Vida
5.
Redox Rep ; 29(1): 2347139, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38718286

RESUMEN

OBJECTIVES: The objective of this study was to investigate whether skeletal muscle cystathionine γ-lyase (CTH) contributes to high-fat diet (HFD)-induced metabolic disorders using skeletal muscle Cth knockout (CthΔskm) mice. METHODS: The CthΔskm mice and littermate Cth-floxed (Cthf/f) mice were fed with either HFD or chow diet for 13 weeks. Metabolomics and transcriptome analysis were used to assess the impact of CTH deficiency in skeletal muscle. RESULTS: Metabolomics coupled with transcriptome showed that CthΔskm mice displayed impaired energy metabolism and some signaling pathways linked to insulin resistance (IR) in skeletal muscle although the mice had normal insulin sensitivity. HFD led to reduced CTH expression and impaired energy metabolism in skeletal muscle in Cthf/f mice. CTH deficiency and HFD had some common pathways enriched in the aspects of amino acid metabolism, carbon metabolism, and fatty acid metabolism. CthΔskm+HFD mice exhibited increased body weight gain, fasting blood glucose, plasma insulin, and IR, and reduced glucose transporter 4 and CD36 expression in skeletal muscle compared to Cthf/f+HFD mice. Impaired mitochondria and irregular arrangement in myofilament occurred in CthΔskm+HFD mice. Omics analysis showed differential pathways enriched between CthΔskm mice and Cthf/f mice upon HFD. More severity in impaired energy metabolism, reduced AMPK signaling, and increased oxidative stress and ferroptosis occurred in CthΔskm+HFD mice compared to Cthf/f+HFD mice. DISCUSSION: Our results indicate that skeletal muscle CTH expression dysregulation contributes to metabolism disorders upon HFD.


Asunto(s)
Cistationina gamma-Liasa , Dieta Alta en Grasa , Hiperglucemia , Resistencia a la Insulina , Músculo Esquelético , Obesidad , Animales , Resistencia a la Insulina/fisiología , Músculo Esquelético/metabolismo , Ratones , Obesidad/metabolismo , Cistationina gamma-Liasa/metabolismo , Cistationina gamma-Liasa/genética , Cistationina gamma-Liasa/deficiencia , Dieta Alta en Grasa/efectos adversos , Hiperglucemia/metabolismo , Ratones Noqueados , Masculino , Metabolismo Energético
6.
J Clin Invest ; 134(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690726

RESUMEN

Proline substitutions within the coiled-coil rod region of the ß-myosin gene (MYH7) are the predominant mutations causing Laing distal myopathy (MPD1), an autosomal dominant disorder characterized by progressive weakness of distal/proximal muscles. We report that the MDP1 mutation R1500P, studied in what we believe to be the first mouse model for the disease, adversely affected myosin motor activity despite being in the structural rod domain that directs thick filament assembly. Contractility experiments carried out on isolated mutant muscles, myofibrils, and myofibers identified muscle fatigue and weakness phenotypes, an increased rate of actin-myosin detachment, and a conformational shift of the myosin heads toward the more reactive disordered relaxed (DRX) state, causing hypercontractility and greater ATP consumption. Similarly, molecular analysis of muscle biopsies from patients with MPD1 revealed a significant increase in sarcomeric DRX content, as observed in a subset of myosin motor domain mutations causing hypertrophic cardiomyopathy. Finally, oral administration of MYK-581, a small molecule that decreases the population of heads in the DRX configuration, significantly improved the limited running capacity of the R1500P-transgenic mice and corrected the increased DRX state of the myofibrils from patients. These studies provide evidence of the molecular pathogenesis of proline rod mutations and lay the groundwork for the therapeutic advancement of myosin modulators.


Asunto(s)
Sustitución de Aminoácidos , Miopatías Distales , Prolina , Animales , Ratones , Humanos , Prolina/genética , Prolina/metabolismo , Miopatías Distales/genética , Miopatías Distales/metabolismo , Miopatías Distales/patología , Mutación Missense , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Cadenas Pesadas de Miosina/química , Femenino , Masculino , Ratones Transgénicos , Contracción Muscular/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología
7.
Skelet Muscle ; 14(1): 9, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702726

RESUMEN

BACKGROUND: Adeno-associated virus (AAV)-based gene therapy is a promising strategy to treat muscle diseases. However, this strategy is currently confronted with challenges, including a lack of transduction efficiency across the entire muscular system and toxicity resulting from off-target tissue effects. Recently, novel myotropic AAVs named MyoAAVs and AAVMYOs have been discovered using a directed evolution approach, all separately demonstrating enhanced muscle transduction efficiency and liver de-targeting effects. However, these newly discovered AAV variants have not yet been compared. METHODS: In this study, we performed a comparative analysis of these various AAV9-derived vectors under the same experimental conditions following different injection time points in two distinct mouse strains. RESULTS: We highlight differences in transduction efficiency between AAV9, AAVMYO, MyoAAV2A and MyoAAV4A that depend on age at injection, doses and mouse genetic background. In addition, specific AAV serotypes appeared more potent to transduce skeletal muscles including diaphragm and/or to de-target heart or liver. CONCLUSIONS: Our study provides guidance for researchers aiming to establish proof-of-concept approaches for preventive or curative perspectives in mouse models, to ultimately lead to future clinical trials for muscle disorders.


Asunto(s)
Dependovirus , Terapia Genética , Vectores Genéticos , Ratones Endogámicos C57BL , Músculo Esquelético , Transducción Genética , Animales , Dependovirus/genética , Vectores Genéticos/administración & dosificación , Músculo Esquelético/metabolismo , Ratones , Transducción Genética/métodos , Terapia Genética/métodos , Masculino , Hígado/metabolismo , Ratones Endogámicos mdx
8.
J Clin Invest ; 134(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690729

RESUMEN

The myosin inhibitor mavacamten has transformed the management of obstructive hypertrophic cardiomyopathy (HCM) by targeting myosin ATPase activity to mitigate cardiac hypercontractility. This therapeutic mechanism has proven effective for patients with HCM independent of having a primary gene mutation in myosin. In this issue of the JCI, Buvoli et al. report that muscle hypercontractility is a mechanism of pathogenesis underlying muscle dysfunction in Laing distal myopathy, a disorder characterized by mutations altering the rod domain of ß myosin heavy chain. The authors performed detailed physiological, molecular, and biomechanical analyses and demonstrated that myosin ATPase inhibition can correct a large extent of muscle abnormalities. The findings offer a therapeutic avenue for Laing distal myopathy and potentially other myopathies. This Commentary underscores the importance of reevaluating myosin activity's role across myopathies in general for the potential development of targeted myosin inhibitors to treat skeletal muscle disorders.


Asunto(s)
Bencilaminas , Músculo Esquelético , Uracilo/análogos & derivados , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/genética , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Miopatías Distales/genética , Miopatías Distales/tratamiento farmacológico , Miopatías Distales/metabolismo , Miopatías Distales/patología , Animales , Mutación , Miosinas/metabolismo , Miosinas/genética
9.
Int J Mol Sci ; 25(9)2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38732238

RESUMEN

Efficient repair of skeletal muscle relies upon the precise coordination of cells between the satellite cell niche and innate immune cells that are recruited to the site of injury. The expression of pro-inflammatory cytokines and chemokines such as TNFα, IFNγ, CXCL1, and CCL2, by muscle and tissue resident immune cells recruits neutrophils and M1 macrophages to the injury and activates satellite cells. These signal cascades lead to highly integrated temporal and spatial control of muscle repair. Despite the therapeutic potential of these factors for improving tissue regeneration after traumatic and chronic injuries, their transcriptional regulation is not well understood. The transcription factor Mohawk (Mkx) functions as a repressor of myogenic differentiation and regulates fiber type specification. Embryonically, Mkx is expressed in all progenitor cells of the musculoskeletal system and is expressed in human and mouse myeloid lineage cells. An analysis of mice deficient for Mkx revealed a delay in postnatal muscle repair characterized by impaired clearance of necrotic fibers and smaller newly regenerated fibers. Further, there was a delay in the expression of inflammatory signals such as Ccl2, Ifnγ, and Tgfß. This was coupled with impaired recruitment of pro-inflammatory macrophages to the site of muscle damage. These studies demonstrate that Mkx plays a critical role in adult skeletal muscle repair that is mediated through the initial activation of the inflammatory response.


Asunto(s)
Inflamación , Músculo Esquelético , Animales , Músculo Esquelético/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Regeneración , Macrófagos/metabolismo , Macrófagos/inmunología , Ratones , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Desarrollo de Músculos
10.
Nutrients ; 16(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732615

RESUMEN

Adequate diet, physical activity, and dietary supplementation with muscle-targeted food for special medical purposes (FSMP) or dietary supplement (DS) are currently considered fundamental pillars in sarcopenia treatment. The aim of this study is to evaluate the effectiveness of a DS (containing hydroxy-methyl-butyrate, carnosine, and magnesium, for its action on muscle function and protein synthesis and butyrate and lactoferrin for their contribution to the regulation of gut permeability and antioxidant/anti-inflammation activity) on muscle mass (assessed by dual X-ray absorptiometry (DXA)), muscle function (by handgrip test, chair test, short physical performance battery (SPPB) test, and walking speed test), inflammation (tumor necrosis factor-alpha (TNF-a), C-reactive protein (CRP), and visceral adipose tissue (VAT)) and gut axis (by zonulin). A total of 59 participants (age 79.7 ± 4.8 years, body mass index 20.99 ± 2.12 kg/m2) were enrolled and randomly assigned to intervention (n = 30) or placebo (n = 28). The skeletal muscle index (SMI) significantly improved in the supplemented group compared to the placebo one, +1.02 (CI 95%: -0.77; 1.26), p = 0.001; a significant reduction in VAT was observed in the intervention group, -70.91 g (-13.13; -4.70), p = 0.036. Regarding muscle function, all the tests significantly improved (p = 0.001) in the supplemented group compared to the placebo one. CRP, zonulin, and TNF-alpha significantly decreased (p = 0.001) in intervention, compared to placebo, -0.74 mg/dL (CI 95%: -1.30; -0.18), -0.30 ng/mL (CI 95%: -0.37; -0.23), -6.45 pg/mL (CI 95%: -8.71; -4.18), respectively. This DS improves muscle mass and function, and the gut muscle has emerged as a new intervention target for sarcopenia.


Asunto(s)
Carnosina , Suplementos Dietéticos , Lactoferrina , Magnesio , Músculo Esquelético , Permeabilidad , Sarcopenia , Humanos , Masculino , Anciano , Femenino , Sarcopenia/tratamiento farmacológico , Sarcopenia/prevención & control , Carnosina/administración & dosificación , Lactoferrina/administración & dosificación , Lactoferrina/farmacología , Magnesio/administración & dosificación , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Permeabilidad/efectos de los fármacos , Anciano de 80 o más Años , Valeratos/administración & dosificación , Valeratos/farmacología , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Butiratos , Método Doble Ciego , Haptoglobinas , Proteína C-Reactiva/metabolismo , Proteína C-Reactiva/análisis , Precursores de Proteínas
11.
Int J Mol Sci ; 25(9)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38732077

RESUMEN

The skeletal muscles account for approximately 40% of the body weight and are crucial in movement, nutrient absorption, and energy metabolism. Muscle loss and decline in function cause a decrease in the quality of life of patients and the elderly, leading to complications that require early diagnosis. Positron emission tomography/computed tomography (PET/CT) offers non-invasive, high-resolution visualization of tissues. It has emerged as a promising alternative to invasive diagnostic methods and is attracting attention as a tool for assessing muscle function and imaging muscle diseases. Effective imaging of muscle function and pathology relies on appropriate radiopharmaceuticals that target key aspects of muscle metabolism, such as glucose uptake, adenosine triphosphate (ATP) production, and the oxidation of fat and carbohydrates. In this review, we describe how [18F]fluoro-2-deoxy-D-glucose ([18F]FDG), [18F]fluorocholine ([18F]FCH), [11C]acetate, and [15O]water ([15O]H2O) are suitable radiopharmaceuticals for diagnostic imaging of skeletal muscles.


Asunto(s)
Músculo Esquelético , Radiofármacos , Humanos , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/metabolismo , Tomografía de Emisión de Positrones/métodos , Fluorodesoxiglucosa F18 , Animales , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos
12.
Proc Natl Acad Sci U S A ; 121(19): e2313590121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38683978

RESUMEN

Myokines and exosomes, originating from skeletal muscle, are shown to play a significant role in maintaining brain homeostasis. While exercise has been reported to promote muscle secretion, little is known about the effects of neuronal innervation and activity on the yield and molecular composition of biologically active molecules from muscle. As neuromuscular diseases and disabilities associated with denervation impact muscle metabolism, we hypothesize that neuronal innervation and firing may play a pivotal role in regulating secretion activities of skeletal muscles. We examined this hypothesis using an engineered neuromuscular tissue model consisting of skeletal muscles innervated by motor neurons. The innervated muscles displayed elevated expression of mRNAs encoding neurotrophic myokines, such as interleukin-6, brain-derived neurotrophic factor, and FDNC5, as well as the mRNA of peroxisome-proliferator-activated receptor γ coactivator 1α, a key regulator of muscle metabolism. Upon glutamate stimulation, the innervated muscles secreted higher levels of irisin and exosomes containing more diverse neurotrophic microRNAs than neuron-free muscles. Consequently, biological factors secreted by innervated muscles enhanced branching, axonal transport, and, ultimately, spontaneous network activities of primary hippocampal neurons in vitro. Overall, these results reveal the importance of neuronal innervation in modulating muscle-derived factors that promote neuronal function and suggest that the engineered neuromuscular tissue model holds significant promise as a platform for producing neurotrophic molecules.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Exosomas , Músculo Esquelético , Exosomas/metabolismo , Animales , Músculo Esquelético/metabolismo , Músculo Esquelético/inervación , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ratones , Fibronectinas/metabolismo , Neuronas Motoras/metabolismo , Interleucina-6/metabolismo , MicroARNs/metabolismo , MicroARNs/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Neuronas/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Mioquinas
13.
Proc Natl Acad Sci U S A ; 121(19): e2321438121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38687782

RESUMEN

Successful CRISPR/Cas9-based gene editing in skeletal muscle is dependent on efficient propagation of Cas9 to all myonuclei in the myofiber. However, nuclear-targeted gene therapy cargos are strongly restricted to their myonuclear domain of origin. By screening nuclear localization signals and nuclear export signals, we identify "Myospreader," a combination of short peptide sequences that promotes myonuclear propagation. Appending Myospreader to Cas9 enhances protein stability and myonuclear propagation in myoblasts and myofibers. AAV-delivered Myospreader dCas9 better inhibits transcription of toxic RNA in a myotonic dystrophy mouse model. Furthermore, Myospreader Cas9 achieves higher rates of gene editing in CRISPR reporter and Duchenne muscular dystrophy mouse models. Myospreader reveals design principles relevant to all nuclear-targeted gene therapies and highlights the importance of the spatial dimension in therapeutic development.


Asunto(s)
Sistemas CRISPR-Cas , Núcleo Celular , Edición Génica , Terapia Genética , Músculo Esquelético , Distrofia Muscular de Duchenne , Edición Génica/métodos , Animales , Ratones , Músculo Esquelético/metabolismo , Núcleo Celular/metabolismo , Terapia Genética/métodos , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/genética , Humanos , Señales de Localización Nuclear/genética , Proteína 9 Asociada a CRISPR/metabolismo , Proteína 9 Asociada a CRISPR/genética , Modelos Animales de Enfermedad , Mioblastos/metabolismo
14.
Aging (Albany NY) ; 16(8): 7141-7152, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38643465

RESUMEN

Disrupted mitochondrial dynamics and mitophagy contribute to functional deterioration of skeletal muscle (SM) during aging, but the regulatory mechanisms are poorly understood. Our previous study demonstrated that the expression of thyroid hormone receptor α (TRα) decreased significantly in aged mice, suggesting that the alteration of thyroidal elements, especially the decreased TRα, might attenuate local THs action thus to cause the degeneration of SM with aging, while the underlying mechanism remains to be further explored. In this study, decreased expression of myogenic regulators Myf5, MyoD1, mitophagy markers Pink1, LC3II/I, p62, as well as mitochondrial dynamic factors Mfn1 and Opa1, accompanied by increased reactive oxygen species (ROS), showed concomitant changes with reduced TRα expression in aged mice. Further TRα loss- and gain-of-function studies in C2C12 revealed that silencing of TRα not only down-regulated the expression of above-mentioned myogenic regulators, mitophagy markers and mitochondrial dynamic factors, but also led to a significant decrease in mitochondrial activity and maximum respiratory capacity, as well as more mitochondrial ROS and damaged mitochondria. Notedly, overexpression of TRα could up-regulate the expression of those myogenic regulators, mitophagy markers and mitochondrial dynamic factors, meanwhile also led to an increase in mitochondrial activity and number. These results confirmed that TRα could concertedly regulate mitochondrial dynamics, autophagy, and activity, and myogenic regulators rhythmically altered with TRα expression. Summarily, these results suggested that the decline of TRα might cause the degeneration of SM with aging by regulating mitochondrial dynamics, mitophagy and myogenesis.


Asunto(s)
Envejecimiento , Mitofagia , Músculo Esquelético , Especies Reactivas de Oxígeno , Sarcopenia , Receptores alfa de Hormona Tiroidea , Animales , Sarcopenia/metabolismo , Sarcopenia/patología , Ratones , Receptores alfa de Hormona Tiroidea/genética , Receptores alfa de Hormona Tiroidea/metabolismo , Envejecimiento/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Especies Reactivas de Oxígeno/metabolismo , Mitocondrias Musculares/metabolismo , Masculino , Dinámicas Mitocondriales , Mitocondrias/metabolismo , Línea Celular
15.
Physiol Rep ; 12(8): e16003, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38631892

RESUMEN

Chemotherapy is a major contributor to cachexia, but studies often investigate male animals. Here, we investigated whether sex modifies the effects of chemotherapy on cachexia and BCAA metabolism. Ten-week-old CD2F1 male and female mice were treated with the chemotherapy drug cocktail folfiri (50 mg/kg 5-fluorouracil, 90 mg/kg leucovorin, and 24 mg/kg CPT11) (drug) or vehicle twice a week for 6 weeks. Insulin tolerance tests were conducted and BCAA levels and metabolism were measured in plasma and tissues. Drug treatment reduced body and skeletal muscle weights and anabolic signaling in both sexes, with females showing worsened outcomes (p < 0.05 for all). Drug treatment increased plasma BCAA only in males, but BCAA concentrations in the skeletal muscle of both sexes were decreased; this decrease was more profound in males (p = 0.0097). In addition, muscle expression of the BCAA transporter LAT1 was reduced; this reduction was more severe in females (p = 0.0264). In both sexes, the (inhibitory) phosphorylation of BCKD-E1αser293 was increased along with decreased BCKD activity. In the liver, drug treatment increased BCAA concentrations and LAT1 expression, but BCKD activity was suppressed in both sexes (p < 0.05 for all). Our results demonstrate that altered BCAA metabolism may contribute to chemotherapy-induced cachexia in a sex-dependent manner.


Asunto(s)
Caquexia , Caracteres Sexuales , Ratones , Femenino , Masculino , Animales , Caquexia/metabolismo , Aminoácidos de Cadena Ramificada/farmacología , Hígado/metabolismo , Fluorouracilo/farmacología , Músculo Esquelético/metabolismo
16.
Physiol Rep ; 12(7): e15991, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38605421

RESUMEN

Skeletal muscle mass is critical for activities of daily living. Resistance training maintains or increases muscle mass, and various strategies maximize the training adaptation. Mesenchymal stem cells (MSCs) are multipotent cells with differential potency in skeletal muscle cells and the capacity to secrete growth factors. However, little is known regarding the effect of intramuscular injection of MSCs on basal muscle protein synthesis and catabolic systems after resistance training. Here, we measured changes in basal muscle protein synthesis, the ubiquitin-proteasome system, and autophagy-lysosome system-related factors after bouts of resistance exercise by intramuscular injection of MSCs. Mice performed three bouts of resistance exercise (each consisting of 50 maximal isometric contractions elicited by electrical stimulation) on the right gastrocnemius muscle every 48 h, and immediately after the first bout, mice were intramuscularly injected with either MSCs (2.0 × 106 cells) labeled with green fluorescence protein (GFP) or vehicle only placebo. Seventy-two hours after the third exercise bout, GFP was detected only in the muscle injected with MSCs with concomitant elevation of muscle protein synthesis. The injection of MSCs also increased protein ubiquitination. These results suggest that the intramuscular injection of MSCs augmented muscle protein turnover at the basal state after consecutive resistance exercise.


Asunto(s)
Células Madre Mesenquimatosas , Entrenamiento de Fuerza , Humanos , Masculino , Ratones , Animales , Inyecciones Intramusculares , Proteínas Musculares/metabolismo , Actividades Cotidianas , Músculo Esquelético/metabolismo , Células Madre Mesenquimatosas/metabolismo
17.
Toxins (Basel) ; 16(4)2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38668589

RESUMEN

Coralsnakes (Micrurus spp.) are the only elapids found throughout the Americas. They are recognized for their highly neurotoxic venom, which is comprised of a wide variety of toxins, including the stable, low-mass toxins known as three-finger toxins (3FTx). Due to difficulties in venom extraction and availability, research on coralsnake venoms is still very limited when compared to that of other Elapidae snakes like cobras, kraits, and mambas. In this study, two previously described 3FTx from the venom of M. corallinus, NXH1 (3SOC1_MICCO), and NXH8 (3NO48_MICCO) were characterized. Using in silico, in vitro, and ex vivo experiments, the biological activities of these toxins were predicted and evaluated. The results showed that only NXH8 was capable of binding to skeletal muscle cells and modulating the activity of nAChRs in nerve-diaphragm preparations. These effects were antagonized by anti-rNXH8 or antielapidic sera. Sequence analysis revealed that the NXH1 toxin possesses eight cysteine residues and four disulfide bonds, while the NXH8 toxin has a primary structure similar to that of non-conventional 3FTx, with an additional disulfide bond on the first loop. These findings add more information related to the structural diversity present within the 3FTx class, while expanding our understanding of the mechanisms of the toxicity of this coralsnake venom and opening new perspectives for developing more effective therapeutic interventions.


Asunto(s)
Clonación Molecular , Serpientes de Coral , Venenos Elapídicos , Músculo Esquelético , Receptores Nicotínicos , Animales , Venenos Elapídicos/química , Venenos Elapídicos/toxicidad , Venenos Elapídicos/genética , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Secuencia de Aminoácidos , Masculino
18.
Gen Comp Endocrinol ; 353: 114513, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38604437

RESUMEN

Skeletal muscle, comprising a significant proportion (40 to 50 percent) of total body weight in humans, plays a critical role in maintaining normal physiological conditions. Muscle atrophy occurs when the rate of protein degradation exceeds protein synthesis. Sarcopenia refers to age-related muscle atrophy, while cachexia represents a more complex form of muscle wasting associated with various diseases such as cancer, heart failure, and AIDS. Recent research has highlighted the involvement of signaling pathways, including IGF1-Akt-mTOR, MuRF1-MAFbx, and FOXO, in regulating the delicate balance between muscle protein synthesis and breakdown. Myostatin, a member of the TGF-ß superfamily, negatively regulates muscle growth and promotes muscle atrophy by activating Smad2 and Smad3. It also interacts with other signaling pathways in cachexia and sarcopenia. Inhibition of myostatin has emerged as a promising therapeutic approach for sarcopenia and cachexia. Additionally, other TGF-ß family members, such as TGF-ß1, activin A, and GDF11, have been implicated in the regulation of skeletal muscle mass. Furthermore, myostatin cooperates with these family members to impair muscle differentiation and contribute to muscle loss. This review provides an overview of the significance of myostatin and other TGF-ß signaling pathway members in muscular dystrophy, sarcopenia, and cachexia. It also discusses potential novel therapeutic strategies targeting myostatin and TGF-ß signaling for the treatment of muscle atrophy.


Asunto(s)
Caquexia , Atrofia Muscular , Miostatina , Neoplasias , Sarcopenia , Transducción de Señal , Factor de Crecimiento Transformador beta , Humanos , Caquexia/metabolismo , Caquexia/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Sarcopenia/metabolismo , Sarcopenia/patología , Transducción de Señal/fisiología , Neoplasias/metabolismo , Neoplasias/complicaciones , Neoplasias/patología , Factor de Crecimiento Transformador beta/metabolismo , Miostatina/metabolismo , Animales , Músculo Esquelético/metabolismo , Músculo Esquelético/patología
19.
Biomed Pharmacother ; 174: 116556, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38636398

RESUMEN

Skeletal muscle atrophy is a common complication of chronic kidney disease (CKD) that affects the quality of life and prognosis of patients. We aimed to investigate the effects and mechanisms of caffeic acid (CA), a natural phenolic compound, on skeletal muscle atrophy in CKD rats. Male Sprague-Dawley rats underwent 5/6 nephrectomy (NPM) and were treated with CA (20, 40, or 80 mg/kg/day) for 10 weeks. The body and muscle weights, renal function, hemoglobin, and albumin were measured. The histological, molecular, and biochemical changes in skeletal muscles were evaluated using hematoxylin-eosin staining, quantitative real-time PCR, malondialdehyde/catalase/superoxide dismutase/glutathione level detection, and enzyme-linked immunosorbent assay. Western blotting and network pharmacology were applied to identify the potential targets and pathways of CA, CKD, and muscle atrophy. The results showed that CA significantly improved NPM-induced muscle-catabolic effects, reduced the expression of muscle atrophy-related proteins (muscle atrophy F-box and muscle RING finger 1) and proinflammatory cytokines (interleukin [IL]-6, tumor necrosis factor-alpha, and IL-1ß), and attenuated muscle oxidative stress. Network pharmacology revealed that CA modulated the response to oxidative stress and nuclear factor kappa B (NF-κB) signaling pathway and that Toll-like receptor 4 (TLR4) was a key target. In vivo experiment confirmed that CA inhibited the TLR4/myeloid differentiation primary response 88 (MYD88)/NF-kB signaling pathway, reduced muscle iron levels, and restored glutathione peroxidase 4 activity, thereby alleviating ferroptosis and inflammation in skeletal muscles. Thus, CA might be a promising therapeutic agent for preventing and treating skeletal muscle atrophy in CKD by modulating the TLR4/MYD88/NF-κB pathway and ferroptosis.


Asunto(s)
Ácidos Cafeicos , Atrofia Muscular , Factor 88 de Diferenciación Mieloide , Insuficiencia Renal Crónica , Transducción de Señal , Animales , Masculino , Ratas , Ácidos Cafeicos/farmacología , Citocinas/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Músculo Esquelético/metabolismo , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/patología , Atrofia Muscular/etiología , Atrofia Muscular/prevención & control , Atrofia Muscular/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Nefrectomía/efectos adversos , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo
20.
Sci Rep ; 14(1): 9132, 2024 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-38644379

RESUMEN

The diaphragm is a unique skeletal muscle due to its continuous activation pattern during the act of breathing. The ontogeny of macrophages, pivotal cells for skeletal muscle maintenance and regeneration, is primarily based on two distinct origins: postnatal bone marrow-derived monocytes and prenatal embryonic progenitors. Here we employed chimeric mice to study the dynamics of these two macrophage populations under different conditions. Traditional chimeric mice generated through whole body irradiation showed virtually complete elimination of the original tissue-resident macrophage pool. We then developed a novel method which employs lead shielding to protect the diaphragm tissue niche from irradiation. This allowed us to determine that up to almost half of tissue-resident macrophages in the diaphragm can be maintained independently from bone marrow-derived monocytes under steady-state conditions. These findings were confirmed by long-term (5 months) parabiosis experiments. Acute diaphragm injury shifted the macrophage balance toward an overwhelming predominance of bone marrow (monocyte)-derived macrophages. However, there was a remarkable reversion to the pre-injury ontological landscape after diaphragm muscle recovery. This diaphragm shielding method permits analysis of the dynamics of macrophage origin and corresponding function under different physiological and pathological conditions. It may be especially useful for studying diseases which are characterized by acute or chronic injury of the diaphragm and accompanying inflammation.


Asunto(s)
Diafragma , Homeostasis , Macrófagos , Animales , Macrófagos/metabolismo , Ratones , Monocitos , Músculo Esquelético/metabolismo , Regeneración , Ratones Endogámicos C57BL , Irradiación Corporal Total , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA