Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.511
Filtrer
1.
Physiol Res ; 73(3): 405-413, 2024 07 17.
Article de Anglais | MEDLINE | ID: mdl-39027957

RÉSUMÉ

Cisplatin is a widely used chemotherapy drug for the treatment of various cancers. However, although cisplatin is effective in targeting cancer cells, it has severe side effects including skeletal muscle atrophy. In this study, we aimed to characterize the role of Dihydromyricetin in cisplatin-induced muscle atrophy in mice. 5-week-old male C57BL/6 mice were treated with Dihydromyricetin for 14 days orally followed by in intraperitoneally cisplatin administration for 6 days. Gastrocnemius muscles were isolated for the following experiments. Antioxidative stress were determined by peroxidative product malondialdehyde (MDA) and antioxidants superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities. Quadriceps muscle mass and grip strength were significantly restored by Dihydromyricetin in a dose-dependent manner. Moreover, muscle fibers were improved in Dihydromyricetin treated group. Excessive skeletal muscle E3 ubiquitin-protein ligases in cisplatin group were significantly repressed by Dihydromyricetin treatment. Dihydromyricetin significantly reduced oxidative stress induced by cisplatin by decreasing MDA level and restored SOD and GPx activities. In addition, ferroptosis was significantly reduced by Dihydromyricetin characterized by reduced iron level and ferritin heavy chain 1 and improved Gpx4 level. The present study demonstrated that Dihydromyricetin attenuated cisplatin-induced muscle atrophy by reducing skeletal muscle E3 ubiquitin-protein ligases, oxidative stress, and ferroptosis.


Sujet(s)
Cisplatine , Ferroptose , Flavonols , Souris de lignée C57BL , Amyotrophie , Stress oxydatif , Animaux , Mâle , Flavonols/pharmacologie , Flavonols/usage thérapeutique , Amyotrophie/induit chimiquement , Amyotrophie/anatomopathologie , Amyotrophie/métabolisme , Amyotrophie/prévention et contrôle , Amyotrophie/traitement médicamenteux , Ferroptose/effets des médicaments et des substances chimiques , Cisplatine/toxicité , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/anatomopathologie , Muscles squelettiques/métabolisme , Antinéoplasiques/toxicité , Antioxydants/pharmacologie
2.
Skelet Muscle ; 14(1): 17, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39044305

RÉSUMÉ

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is characterized by progressive motor neuron (MN) degeneration, leading to neuromuscular junction (NMJ) dismantling and severe muscle atrophy. The nuclear receptor interaction protein (NRIP) functions as a multifunctional protein. It directly interacts with calmodulin or α-actinin 2, serving as a calcium sensor for muscle contraction and maintaining sarcomere integrity. Additionally, NRIP binds with the acetylcholine receptor (AChR) for NMJ stabilization. Loss of NRIP in muscles results in progressive motor neuron degeneration with abnormal NMJ architecture, resembling ALS phenotypes. Therefore, we hypothesize that NRIP could be a therapeutic factor for ALS. METHODS: We used SOD1 G93A mice, expressing human SOD1 with the ALS-linked G93A mutation, as an ALS model. An adeno-associated virus vector encoding the human NRIP gene (AAV-NRIP) was generated and injected into the muscles of SOD1 G93A mice at 60 days of age, before disease onset. Pathological and behavioral changes were measured to evaluate the therapeutic effects of AAV-NRIP on the disease progression of SOD1 G93A mice. RESULTS: SOD1 G93A mice exhibited lower NRIP expression than wild-type mice in both the spinal cord and skeletal muscle tissues. Forced NRIP expression through AAV-NRIP intramuscular injection was observed in skeletal muscles and retrogradely transduced into the spinal cord. AAV-NRIP gene therapy enhanced movement distance and rearing frequencies in SOD1 G93A mice. Moreover, AAV-NRIP increased myofiber size and slow myosin expression, ameliorated NMJ degeneration and axon terminal denervation at NMJ, and increased the number of α-motor neurons (α-MNs) and compound muscle action potential (CMAP) in SOD1 G93A mice. CONCLUSIONS: AAV-NRIP gene therapy ameliorates muscle atrophy, motor neuron degeneration, and axon terminal denervation at NMJ, leading to increased NMJ transmission and improved motor functions in SOD1 G93A mice. Collectively, AAV-NRIP could be a potential therapeutic drug for ALS.


Sujet(s)
Sclérose latérale amyotrophique , Dependovirus , Modèles animaux de maladie humaine , Thérapie génétique , Souris transgéniques , Motoneurones , Amyotrophie , Animaux , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/thérapie , Sclérose latérale amyotrophique/métabolisme , Sclérose latérale amyotrophique/anatomopathologie , Thérapie génétique/méthodes , Amyotrophie/génétique , Amyotrophie/thérapie , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Motoneurones/métabolisme , Motoneurones/anatomopathologie , Dependovirus/génétique , Souris , Humains , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Jonction neuromusculaire/métabolisme , Jonction neuromusculaire/anatomopathologie , Vecteurs génétiques/administration et posologie , Dégénérescence nerveuse/génétique , Dégénérescence nerveuse/thérapie , Mâle , Superoxide dismutase-1/génétique , Superoxide dismutase-1/métabolisme
3.
Cell Mol Biol Lett ; 29(1): 99, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38978023

RÉSUMÉ

Skeletal muscular atrophy is a complex disease involving a large number of gene expression regulatory networks and various biological processes. Despite extensive research on this topic, its underlying mechanisms remain elusive, and effective therapeutic approaches are yet to be established. Recent studies have shown that epigenetics play an important role in regulating skeletal muscle atrophy, influencing the expression of numerous genes associated with this condition through the addition or removal of certain chemical modifications at the molecular level. This review article comprehensively summarizes the different types of modifications to DNA, histones, RNA, and their known regulators. We also discuss how epigenetic modifications change during the process of skeletal muscle atrophy, the molecular mechanisms by which epigenetic regulatory proteins control skeletal muscle atrophy, and assess their translational potential. The role of epigenetics on muscle stem cells is also highlighted. In addition, we propose that alternative splicing interacts with epigenetic mechanisms to regulate skeletal muscle mass, offering a novel perspective that enhances our understanding of epigenetic inheritance's role and the regulatory network governing skeletal muscle atrophy. Collectively, advancements in the understanding of epigenetic mechanisms provide invaluable insights into the study of skeletal muscle atrophy. Moreover, this knowledge paves the way for identifying new avenues for the development of more effective therapeutic strategies and pharmaceutical interventions.


Sujet(s)
Épigenèse génétique , Muscles squelettiques , Amyotrophie , Humains , Amyotrophie/génétique , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Muscles squelettiques/anatomopathologie , Muscles squelettiques/métabolisme , Animaux , Histone/métabolisme , Histone/génétique , Méthylation de l'ADN/génétique , Épissage alternatif/génétique
4.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-39000606

RÉSUMÉ

Sarcopenia refers to an age-related decrease in muscle mass and strength. The gut-muscle axis has been proposed as a promising target to alleviate muscle atrophy. The effect of KL-Biome-a postbiotic preparation comprising heat-killed Lactiplantibacillus plantarum KM-2, its metabolites, and an excipient (soybean powder)-on muscle atrophy was evaluated using dexamethasone (DEX)-induced atrophic C2C12 myoblasts and C57BL/6J mice. KL-Biome significantly downregulated the expression of genes (Atrogin-1 and MuRF1) associated with skeletal muscle degradation but increased the anabolic phosphorylation of FoxO3a, Akt, and mTOR in C2C12 cells. Oral administration of KL-Biome (900 mg/kg) for 8 weeks significantly improved muscle mass, muscle function, and serum lactate dehydrogenase levels in DEX-treated mice. KL-Biome administration increased gut microbiome diversity and reversed DEX-mediated gut microbiota alterations. Furthermore, it significantly increased the relative abundances of the genera Subdologranulum, Alistipes, and Faecalibacterium prausnitzii, which are substantially involved in short-chain fatty acid production. These findings suggest that KL-Biome exerts beneficial effects on muscle atrophy by regulating gut microbiota.


Sujet(s)
Dexaméthasone , Microbiome gastro-intestinal , Souris de lignée C57BL , Muscles squelettiques , Amyotrophie , Animaux , Amyotrophie/traitement médicamenteux , Amyotrophie/métabolisme , Amyotrophie/induit chimiquement , Souris , Dexaméthasone/pharmacologie , Dexaméthasone/effets indésirables , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Mâle , Protéines du muscle/métabolisme , Protéines du muscle/génétique , Protéine O3 à motif en tête de fourche/métabolisme , Protéine O3 à motif en tête de fourche/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , SKP cullin F-box protein ligases/métabolisme , SKP cullin F-box protein ligases/génétique , Probiotiques/administration et posologie , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Sarcopénie/traitement médicamenteux , Sarcopénie/métabolisme , Sarcopénie/anatomopathologie , Sérine-thréonine kinases TOR/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Lignée cellulaire , Lactobacillus plantarum
5.
Physiol Rep ; 12(13): e16145, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39001580

RÉSUMÉ

The dystrophin protein has well-characterized roles in force transmission and maintaining membrane integrity during muscle contraction. Studies have reported decreased expression of dystrophin in atrophying muscles during wasting conditions, and that restoration of dystrophin can attenuate atrophy, suggesting a role in maintaining muscle mass. Phosphorylation of S3059 within the cysteine-rich region of dystrophin enhances binding between dystrophin and ß-dystroglycan, and mimicking phosphorylation at this site by site-directed mutagenesis attenuates myotube atrophy in vitro. To determine whether dystrophin phosphorylation can attenuate muscle wasting in vivo, CRISPR-Cas9 was used to generate mice with whole body mutations of S3059 to either alanine (DmdS3059A) or glutamate (DmdS3059E), to mimic a loss of, or constitutive phosphorylation of S3059, on all endogenous dystrophin isoforms, respectively. Sciatic nerve transection was performed on these mice to determine whether phosphorylation of dystrophin S3059 could attenuate denervation atrophy. At 14 days post denervation, atrophy of tibialis anterior (TA) but not gastrocnemius or soleus muscles, was partially attenuated in DmdS3059E mice relative to WT mice. Attenuation of atrophy was associated with increased expression of ß-dystroglycan in TA muscles of DmdS3059E mice. Dystrophin S3059 phosphorylation can partially attenuate denervation-induced atrophy, but may have more significant impact in less severe modes of muscle wasting.


Sujet(s)
Dystrophine , Muscles squelettiques , Amyotrophie , Animaux , Phosphorylation , Souris , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/génétique , Muscles squelettiques/métabolisme , Muscles squelettiques/innervation , Muscles squelettiques/anatomopathologie , Dystrophine/métabolisme , Dystrophine/génétique , Mâle , Dénervation musculaire/méthodes , Souris de lignée C57BL
6.
Biomed Res ; 45(4): 163-172, 2024.
Article de Anglais | MEDLINE | ID: mdl-39010192

RÉSUMÉ

Exercise training increases brain-derived neurotrophic factor (BDNF) expression and improves cognitive function. However, the dynamics of BDNF during inactivity and the effects of exercise intervention on BDNF levels have rarely been examined. Therefore, we aimed to examine changes in serum, skeletal muscle, and brain BDNF levels under these conditions. Mice were divided into control (Co), cast immobilization (CI), reloading (RL), and exercise (Ex) groups. Muscle atrophy was induced by cast immobilization for 2 weeks in the CI, RL, and Ex groups. After cast removal, the RL and Ex groups underwent regrounding and treadmill exercise, respectively, for 2 weeks. Serum, skeletal muscle, and brain BDNF levels showed a similar decreasing trend in the CI group, recovery in the RL group, and a further increase in the Ex group compared with those in the Co group. This indicates that BDNF levels change in parallel with the degree of activity. However, the magnitude of variation differed among the tissues in the order of serum > skeletal muscle > brain tissue. These results suggest that different mechanisms in different tissues regulate BDNF expression. BDNF could potentially act as an objective measure of the impact of both inactivity and exercise-based interventions.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Encéphale , Muscles squelettiques , Conditionnement physique d'animal , Animaux , Mâle , Souris , Encéphale/métabolisme , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/sang , Cinétique , Muscles squelettiques/métabolisme , Amyotrophie/métabolisme , Amyotrophie/thérapie
7.
Int J Mol Sci ; 25(14)2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-39062775

RÉSUMÉ

Breast cancer (BC) stands out as the most commonly type of cancer diagnosed in women worldwide, and chemotherapy, a key component of treatment, exacerbates cancer-induced skeletal muscle wasting, contributing to adverse health outcomes. Notably, the impact of chemotherapy on skeletal muscle seems to surpass that of the cancer itself, with inflammation identified as a common trigger for muscle wasting in both contexts. In skeletal muscle, pro-inflammatory cytokines modulate pathways crucial for the delicate balance between protein synthesis and breakdown, as well as satellite cell activation and myonuclear accretion. Physical exercise consistently emerges as a crucial therapeutic strategy to counteract cancer and chemotherapy-induced muscle wasting, ultimately enhancing patients' quality of life. However, a "one size fits all" approach does not apply to the prescription of exercise for BC patients, with factors such as age, menopause and comorbidities influencing the response to exercise. Hence, tailored exercise regimens, considering factors such as duration, frequency, intensity, and type, are essential to maximize efficacy in mitigating muscle wasting and improving disease outcomes. Despite the well-established anti-inflammatory role of aerobic exercise, resistance exercise proves equally or more beneficial in terms of mass and strength gain, as well as enhancing quality of life. This review comprehensively explores the molecular pathways affected by distinct exercise regimens in the skeletal muscle of cancer patients during chemotherapy, providing critical insights for precise exercise implementation to prevent skeletal muscle wasting.


Sujet(s)
Tumeurs du sein , Exercice physique , Muscles squelettiques , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Femelle , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Qualité de vie , Traitement par les exercices physiques/méthodes , Antinéoplasiques/effets indésirables , Antinéoplasiques/usage thérapeutique , Amyotrophie/étiologie , Amyotrophie/métabolisme
8.
Int J Chron Obstruct Pulmon Dis ; 19: 1591-1601, 2024.
Article de Anglais | MEDLINE | ID: mdl-39005647

RÉSUMÉ

Background: Exercise is an indispensable component of pulmonary rehabilitation with strong anti-inflammatory effects. However, the mechanisms by which exercise prevents diaphragmatic atrophy in COPD (chronic obstructive pulmonary disease) remain unclear. Methods: Forty male C57BL/6 mice were assigned to the control (n=16) and smoke (n=24) groups. Mice in the smoke group were exposed to the cigarette smoke (CS) for six months. They were then divided into model and exercise training groups for 2 months. Histological changes were observed in lung and diaphragms. Subsequently, agonist U46639 and antagonist Y27632 of RhoA/ROCK were subjected to mechanical stretching in LPS-treated C2C12 myoblasts. The expression levels of Atrogin-1, MuRF-1, MyoD, Myf5, IL-1ß, TNF-α, and RhoA/ROCK were determined by Western blotting. Results: Diaphragmatic atrophy and increased RhoA/ROCK expression were observed in COPD mice. Exercise training attenuated diaphragmatic atrophy, decreased the expression of MuRF-1, and increased MyoD expression in COPD diaphragms. Exercise also affects the upregulation of RhoA/ROCK and inflammation-related proteins. In in vitro experiments with C2C12 myoblasts, LPS remarkably increased the level of inflammation and protein degradation, whereas Y27632 or combined with mechanical stretching prevented this phenomenon considerably. Conclusion: RhoA/ROCK plays an important role in the prevention of diaphragmatic atrophy in COPD.


Sujet(s)
Muscle diaphragme , Modèles animaux de maladie humaine , Souris de lignée C57BL , Amyotrophie , Broncho-pneumopathie chronique obstructive , Transduction du signal , rho-Associated Kinases , Protéine G RhoA , Animaux , Broncho-pneumopathie chronique obstructive/métabolisme , Broncho-pneumopathie chronique obstructive/physiopathologie , rho-Associated Kinases/métabolisme , Mâle , Amyotrophie/prévention et contrôle , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/physiopathologie , Amyotrophie/étiologie , Protéine G RhoA/métabolisme , Muscle diaphragme/métabolisme , Muscle diaphragme/physiopathologie , Muscle diaphragme/anatomopathologie , Lignée cellulaire , Protéines G rho/métabolisme , Traitement par les exercices physiques/méthodes , Souris , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/physiopathologie , Médiateurs de l'inflammation/métabolisme , Conditionnement physique d'animal
9.
J Cachexia Sarcopenia Muscle ; 15(4): 1601-1615, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39031684

RÉSUMÉ

BACKGROUNDS: Fat infiltration of skeletal muscle has been recognized as a common feature of many degenerative muscle disorders. Retinol binding protein 4 (RBP4) is an adipokine that has been demonstrated to be correlated with the presence and severity of sarcopenia in the elderly. However, the exact role and the underlying mechanism of RBP4 in muscle atrophy remains unclear. METHODS: Denervation-induced muscle atrophy model was constructed in wild-type and RBP4 knockout mice. To modify the expression of RBP4, mice were received intramuscular injection of retinol-free RBP4 (apo-RBP4), retinol-bound RBP4 (holo-RBP4) or oral gavage of RBP4 inhibitor A1120. Holo-RBP4-stimulated C2C12 myotubes were treated with siRNAs or specific inhibitors targeting signalling receptor and transporter of retinol 6 (STRA6)/Janus kinase 2 (JAK2)/Signal transducer and activator of transcription 3 (STAT3) pathway. Fat accumulation, myofibre cross-sectional area, myotube diameter and the expression of muscle atrophy markers and myogenesis markers were analysed. RESULTS: The expression levels of RBP4 in skeletal muscles were significantly up-regulated more than 2-fold from 7 days and sustained for 28 days after denervation. Immunofluorescence analysis indicated that increased RBP4 was localized in the infiltrated fatty region in denervated skeletal muscles. Knockout of RBP4 alleviated denervation-induced fatty infiltration and muscle atrophy together with decreased expression of atrophy marker Atrogin-1 and MuRF1 as well as increased expression of myogenesis regulators MyoD and MyoG. By contrast, injection of retinol-bound holo-RBP4 aggregated denervation-induced ectopic fat accumulation and muscle atrophy. Consistently, holo-RBP4 stimulation also had a dose-dependent effect on the reduction of C2C12 myotube diameter and myofibre cross-sectional area, as well as on the increase of Atrogin-1and MuRF1 expression and decrease of MyoD and MyoG expression. Mechanistically, holo-RBP4 treatment increased the expression of its membrane receptor STRA6 (>3-fold) and promoted the phosphorylation of downstream JAK2 and STAT3. Inhibition of STRA6/JAK2/STAT3 pathway either by specific siRNAs or inhibitors could decrease the expression of Atrogin-1 and MuRF1 (>50%) and decrease the expression of MyoD and MyoG (>3-fold) in holo-RBP4-treated C2C12 myotube. RBP4 specific pharmacological antagonist A1120 significantly inhibited the activation of STRA6/JAK2/STAT3 pathway, ameliorated ectopic fat infiltration and protected against denervation-induced muscle atrophy (30% increased myofibre cross-sectional area) in mice. CONCLUSIONS: In conclusion, our data reveal that RBP4 promotes fat infiltration and muscle atrophy through a STRA6-dependent and JAK2/STAT3 pathway-mediated mechanism in denervated skeletal muscle. Our results suggest that lowering RBP4 levels might serve as a promising therapeutic approach for prevention and treatment of muscle atrophy.


Sujet(s)
Amyotrophie , Protéines plasmatiques de liaison au rétinol , Transduction du signal , Animaux , Protéines plasmatiques de liaison au rétinol/métabolisme , Souris , Amyotrophie/métabolisme , Amyotrophie/étiologie , Protéines membranaires/métabolisme , Souris knockout , Modèles animaux de maladie humaine , Facteur de transcription STAT-3/métabolisme , Muscles squelettiques/anatomopathologie , Muscles squelettiques/métabolisme , Mâle , Kinase Janus-2/métabolisme
10.
Int J Mol Sci ; 25(11)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38892242

RÉSUMÉ

Skeletal muscle atrophy (SMA) is caused by a rise in muscle breakdown and a decline in protein synthesis, with a consequent loss of mass and function. This study characterized the effect of an amino acid mixture (AA) in models of SMA, focusing on mitochondria. C57/Bl6 mice underwent immobilization of one hindlimb (I) or cardiotoxin-induced muscle injury (C) and were compared with controls (CTRL). Mice were then administered AA in drinking water for 10 days and compared to a placebo group. With respect to CTRL, I and C reduced running time and distance, along with grip strength; however, the reduction was prevented by AA. Tibialis anterior (TA) muscles were used for histology and mitochondria isolation. I and C resulted in TA atrophy, characterized by a reduction in both wet weight and TA/body weight ratio and smaller myofibers than those of CTRL. Interestingly, these alterations were lightly observed in mice treated with AA. The mitochondrial yield from the TA of I and C mice was lower than that of CTRL but not in AA-treated mice. AA also preserved mitochondrial bioenergetics in TA muscle from I and C mice. To conclude, this study demonstrates that AA prevents loss of muscle mass and function in SMA by protecting mitochondria.


Sujet(s)
Acides aminés , Métabolisme énergétique , Souris de lignée C57BL , Muscles squelettiques , Amyotrophie , Animaux , Souris , Métabolisme énergétique/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/anatomopathologie , Acides aminés/pharmacologie , Acides aminés/métabolisme , Amyotrophie/métabolisme , Amyotrophie/traitement médicamenteux , Amyotrophie/anatomopathologie , Amyotrophie/étiologie , Mâle , Modèles animaux de maladie humaine , Mitochondries du muscle/métabolisme , Mitochondries du muscle/effets des médicaments et des substances chimiques , Mitochondries du muscle/anatomopathologie , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques
11.
Clin Sci (Lond) ; 138(12): 741-756, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38895777

RÉSUMÉ

Periods of skeletal muscle disuse lead to rapid declines in muscle mass (atrophy), which is fundamentally underpinned by an imbalance between muscle protein synthesis (MPS) and muscle protein breakdown (MPB). The complex interplay of molecular mechanisms contributing to the altered regulation of muscle protein balance during disuse have been investigated but rarely synthesised in the context of humans. This narrative review discusses human models of muscle disuse and the ensuing inversely exponential rate of muscle atrophy. The molecular processes contributing to altered protein balance are explored, with a particular focus on growth and breakdown signalling pathways, mitochondrial adaptations and neuromuscular dysfunction. Finally, key research gaps within the disuse atrophy literature are highlighted providing future avenues to enhance our mechanistic understanding of human disuse atrophy.


Sujet(s)
Protéines du muscle , Muscles squelettiques , Amyotrophie , Humains , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Protéines du muscle/métabolisme , Transduction du signal , Immobilisation/effets indésirables , Amyotrophies/métabolisme , Amyotrophies/anatomopathologie , Amyotrophies/physiopathologie
12.
J Orthop Surg Res ; 19(1): 325, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38822418

RÉSUMÉ

OBJECTIVE: Muscle wasting frequently occurs following joint trauma. Previous research has demonstrated that joint distraction in combination with treadmill exercise (TRE) can mitigate intra-articular inflammation and cartilage damage, consequently delaying the advancement of post-traumatic osteoarthritis (PTOA). However, the precise mechanism underlying this phenomenon remains unclear. Hence, the purpose of this study was to examine whether the mechanism by which TRE following joint distraction delays the progression of PTOA involves the activation of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), as well as its impact on muscle wasting. METHODS: Quadriceps samples were collected from patients with osteoarthritis (OA) and normal patients with distal femoral fractures, and the expression of PGC-1α was measured. The hinged external fixator was implanted in the rabbit PTOA model. One week after surgery, a PGC-1α agonist or inhibitor was administered for 4 weeks prior to TRE. Western blot analysis was performed to detect the expression of PGC-1α and Muscle atrophy gene 1 (Atrogin-1). We employed the enzyme-linked immunosorbent assay (ELISA) technique to examine pro-inflammatory factors. Additionally, we utilized quantitative real-time polymerase chain reaction (qRT-PCR) to analyze genes associated with cartilage regeneration. Synovial inflammation and cartilage damage were evaluated through hematoxylin-eosin staining. Furthermore, we employed Masson's trichrome staining and Alcian blue staining to analyze cartilage damage. RESULTS: The decreased expression of PGC-1α in skeletal muscle in patients with OA is correlated with the severity of OA. In the rabbit PTOA model, TRE following joint distraction inhibited the expressions of muscle wasting genes, including Atrogin-1 and muscle ring finger 1 (MuRF1), as well as inflammatory factors such as interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) in skeletal muscle, potentially through the activation of PGC-1α. Concurrently, the production of IL-1ß, IL-6, TNF-α, nitric oxide (NO), and malondialdehyde (MDA) in the synovial fluid was down-regulated, while the expression of type II collagen (Col2a1), Aggrecan (AGN), SRY-box 9 (SOX9) in the cartilage, and superoxide dismutase (SOD) in the synovial fluid was up-regulated. Additionally, histological staining results demonstrated that TRE after joint distraction reduced cartilage degeneration, leading to a significant decrease in OARSI scores.TRE following joint distraction could activate PGC-1α, inhibit Atrogin-1 expression in skeletal muscle, and reduce C-telopeptides of type II collagen (CTX-II) in the blood compared to joint distraction alone. CONCLUSION: Following joint distraction, TRE might promote the activation of PGC-1α in skeletal muscle during PTOA progression to exert anti-inflammatory effects in skeletal muscle and joint cavity, thereby inhibiting muscle wasting and promoting cartilage regeneration, making it a potential therapeutic intervention for treating PTOA.


Sujet(s)
Évolution de la maladie , Muscles squelettiques , Amyotrophie , Arthrose , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Animaux , Lapins , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Arthrose/étiologie , Arthrose/métabolisme , Arthrose/prévention et contrôle , Amyotrophie/étiologie , Amyotrophie/prévention et contrôle , Amyotrophie/métabolisme , Muscles squelettiques/métabolisme , Mâle , Humains , Conditionnement physique d'animal/physiologie , Femelle , Modèles animaux de maladie humaine
13.
Mol Metab ; 86: 101976, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38925248

RÉSUMÉ

OBJECTIVES: A high proportion of women with advanced epithelial ovarian cancer (EOC) experience weakness and cachexia. This relationship is associated with increased morbidity and mortality. EOC is the most lethal gynecological cancer, yet no preclinical cachexia model has demonstrated the combined hallmark features of metastasis, ascites development, muscle loss and weakness in adult immunocompetent mice. METHODS: Here, we evaluated a new model of ovarian cancer-induced cachexia with the advantages of inducing cancer in adult immunocompetent C57BL/6J mice through orthotopic injections of EOC cells in the ovarian bursa. We characterized the development of metastasis, ascites, muscle atrophy, muscle weakness, markers of inflammation, and mitochondrial stress in the tibialis anterior (TA) and diaphragm ∼45, ∼75 and ∼90 days after EOC injection. RESULTS: Primary ovarian tumour sizes were progressively larger at each time point while severe metastasis, ascites development, and reductions in body, fat and muscle weights occurred by 90 Days. There were no changes in certain inflammatory (TNFα), atrogene (MURF1 and Atrogin) or GDF15 markers within both muscles whereas IL-6 was increased at 45 and 90 Day groups in the diaphragm. TA weakness in 45 Day preceded atrophy and metastasis that were observed later (75 and 90 Day, respectively). The diaphragm demonstrated both weakness and atrophy in 45 Day. In both muscles, this pre-severe-metastatic muscle weakness corresponded with considerable reprogramming of gene pathways related to mitochondrial bioenergetics as well as reduced functional measures of mitochondrial pyruvate oxidation and creatine-dependent ADP/ATP cycling as well as increased reactive oxygen species emission (hydrogen peroxide). Remarkably, muscle force per unit mass at 90 days was partially restored in the TA despite the presence of atrophy and severe metastasis. In contrast, the diaphragm demonstrated progressive weakness. At this advanced stage, mitochondrial pyruvate oxidation in both muscles exceeded control mice suggesting an apparent metabolic super-compensation corresponding with restored indices of creatine-dependent adenylate cycling. CONCLUSIONS: This mouse model demonstrates the concurrent development of cachexia and metastasis that occurs in women with EOC. The model provides physiologically relevant advantages of inducing tumour development within the ovarian bursa in immunocompetent adult mice. Moreover, the model reveals that muscle weakness in both TA and diaphragm precedes severe metastasis while weakness also precedes atrophy in the TA. An underlying mitochondrial bioenergetic stress corresponded with this early weakness. Collectively, these discoveries can direct new research towards the development of therapies that target pre-atrophy and pre-severe-metastatic weakness during EOC in addition to therapies targeting cachexia.


Sujet(s)
Cachexie , Modèles animaux de maladie humaine , Souris de lignée C57BL , Mitochondries , Faiblesse musculaire , Tumeurs de l'ovaire , Animaux , Cachexie/métabolisme , Cachexie/étiologie , Cachexie/anatomopathologie , Femelle , Souris , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/complications , Faiblesse musculaire/métabolisme , Faiblesse musculaire/étiologie , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Amyotrophie/métabolisme , Amyotrophie/étiologie , Amyotrophie/anatomopathologie , Métastase tumorale , Carcinome épithélial de l'ovaire/métabolisme , Carcinome épithélial de l'ovaire/anatomopathologie , Lignée cellulaire tumorale
14.
Int J Mol Sci ; 25(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891908

RÉSUMÉ

Chronic inflammation causes muscle wasting. Because most inflammatory cytokine signals are mediated via TGF-ß-activated kinase-1 (TAK1) activation, inflammatory cytokine-induced muscle wasting may be ameliorated by the inhibition of TAK1 activity. The present study was undertaken to clarify whether TAK1 inhibition can ameliorate inflammation-induced muscle wasting. SKG/Jcl mice as an autoimmune arthritis animal model were treated with a small amount of mannan as an adjuvant to enhance the production of TNF-α and IL-1ß. The increase in these inflammatory cytokines caused a reduction in muscle mass and strength along with an induction of arthritis in SKG/Jcl mice. Those changes in muscle fibers were mediated via the phosphorylation of TAK1, which activated the downstream signaling cascade via NF-κB, p38 MAPK, and ERK pathways, resulting in an increase in myostatin expression. Myostatin then reduced the expression of muscle proteins not only via a reduction in MyoD1 expression but also via an enhancement of Atrogin-1 and Murf1 expression. TAK1 inhibitor, LL-Z1640-2, prevented all the cytokine-induced changes in muscle wasting. Thus, TAK1 inhibition can be a new therapeutic target of not only joint destruction but also muscle wasting induced by inflammatory cytokines.


Sujet(s)
Cytokines , MAP Kinase Kinase Kinases , Amyotrophie , Animaux , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/étiologie , Amyotrophie/traitement médicamenteux , Souris , Cytokines/métabolisme , Faiblesse musculaire/métabolisme , Faiblesse musculaire/traitement médicamenteux , Myostatine/métabolisme , Myostatine/antagonistes et inhibiteurs , Protéines du muscle/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Modèles animaux de maladie humaine , Interleukine-1 bêta/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Muscles squelettiques/effets des médicaments et des substances chimiques , Zéaralénone/pharmacologie , Zéaralénone/analogues et dérivés
15.
Int J Mol Sci ; 25(11)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38892252

RÉSUMÉ

Muscular atrophy is a complex catabolic condition that develops due to several inflammatory-related disorders, resulting in muscle loss. Tumor necrosis factor alpha (TNF-α) is believed to be one of the leading factors that drive inflammatory response and its progression. Until now, the link between inflammation and muscle wasting has been thoroughly investigated, and the non-coding RNA machinery is a potential connection between the candidates. This study aimed to identify specific miRNAs for muscular atrophy induced by TNF-α in the C2C12 murine myotube model. The difference in expression of fourteen known miRNAs and two newly identified miRNAs was recorded by next-generation sequencing between normal muscle cells and treated myotubes. After validation, we confirmed the difference in the expression of one novel murine miRNA (nov-mmu-miRNA-1) under different TNF-α-inducing conditions. Functional bioinformatic analyses of nov-mmu-miRNA-1 revealed the potential association with inflammation and muscle atrophy. Our results suggest that nov-mmu-miRNA-1 may trigger inflammation and muscle wasting by the downregulation of LIN28A/B, an anti-inflammatory factor in the let-7 family. Therefore, TNF-α is involved in muscle atrophy through the modulation of the miRNA cellular machinery. Here, we describe for the first time and propose a mechanism for the newly discovered miRNA, nov-mmu-miRNA-1, which may regulate inflammation and promote muscle atrophy.


Sujet(s)
microARN , Amyotrophie , Facteur de nécrose tumorale alpha , Animaux , microARN/génétique , microARN/métabolisme , Souris , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/génétique , Amyotrophie/génétique , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/induit chimiquement , Lignée cellulaire , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/anatomopathologie , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Séquençage nucléotidique à haut débit
16.
In Vivo ; 38(4): 1520-1529, 2024.
Article de Anglais | MEDLINE | ID: mdl-38936901

RÉSUMÉ

Sarcopenia is a prevalent and clinically significant condition, particularly among older age groups and those with chronic disease. Patients with cancer frequently suffer from sarcopenia and progressive loss of muscle mass, strength, and function. The complex interplay between cancer and its treatment, including medical therapy, radiotherapy, and surgery, significantly contributes to the onset and worsening of sarcopenia. Cancer induces muscle wasting through inflammatory processes, metabolic alterations, and hormonal imbalance. Moreover, medical and radiation therapies exert direct toxic effects on muscles, contributing to the impairment of physical function. Loss of appetite, malnutrition, and physical inactivity further exacerbate muscle wasting in cancer patients. Imaging techniques are the cornerstones for sarcopenia diagnosis. Magnetic resonance imaging, computed tomography, and dual-energy X-ray absorptiometry provide valuable insights into muscle structure and quality. Although each modality has advantages and limitations, magnetic resonance imaging produces high-resolution images and provides dynamic information about muscle function. Despite these challenges, addressing sarcopenia is essential for optimizing treatment outcomes and improving survival rates in patients with cancer. This review explored the factors contributing to sarcopenia in oncologic patients, emphasizing the importance of early detection and comprehensive management strategies.


Sujet(s)
Muscles squelettiques , Tumeurs , Sarcopénie , Humains , Sarcopénie/étiologie , Sarcopénie/thérapie , Tumeurs/complications , Tumeurs/thérapie , Tumeurs/métabolisme , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Muscles squelettiques/physiopathologie , Muscles squelettiques/imagerie diagnostique , Amyotrophie/étiologie , Amyotrophie/métabolisme , Imagerie par résonance magnétique/méthodes
17.
Sci Rep ; 14(1): 13282, 2024 06 10.
Article de Anglais | MEDLINE | ID: mdl-38858416

RÉSUMÉ

Recent research has emphasized the role of macrophage-secreted factors on skeletal muscle metabolism. We studied Sargassum Serratifolium ethanol extract (ESS) in countering lipopolysaccharide (LPS)-induced changes in the macrophage transcriptome and their impact on skeletal muscle. Macrophage-conditioned medium (MCM) from LPS-treated macrophages (LPS-MCM) and ESS-treated macrophages (ESS-MCM) affected C2C12 myotube cells. LPS-MCM upregulated muscle atrophy genes and reduced glucose uptake, while ESS-MCM reversed these effects. RNA sequencing revealed changes in the immune system and cytokine transport pathways in ESS-treated macrophages. Protein analysis in ESS-MCM showed reduced levels of key muscle atrophy-related proteins, TNF-α, IL-6, IL-1, and GDF-15. These proteins play crucial roles in muscle function. These findings highlight the intricate relationship between the macrophage transcriptome and their secreted factors in either impairing or enhancing skeletal muscle function. ESS treatment has the potential to reduce macrophage-derived cytokines, preserving skeletal muscle function.


Sujet(s)
Macrophages , Amyotrophie , Extraits de plantes , Sargassum , Sargassum/composition chimique , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Animaux , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Souris , Amyotrophie/métabolisme , Amyotrophie/traitement médicamenteux , Amyotrophie/anatomopathologie , Transcriptome , Lipopolysaccharides , Cytokines/métabolisme , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/anatomopathologie , Lignée cellulaire , Milieux de culture conditionnés/pharmacologie , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques
18.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38928418

RÉSUMÉ

Breast cancer is the type of cancer with the highest prevalence in women worldwide. Skeletal muscle atrophy is an important prognostic factor in women diagnosed with breast cancer. This atrophy stems from disrupted skeletal muscle homeostasis, triggered by diminished anabolic signalling and heightened inflammatory conditions, culminating in an upregulation of skeletal muscle proteolysis gene expression. The importance of delving into research on modulators of skeletal muscle atrophy, such as microRNAs (miRNAs), which play a crucial role in regulating cellular signalling pathways involved in skeletal muscle protein synthesis and degradation, has been recognised. This holds true for conditions of homeostasis as well as pathologies like cancer. However, the determination of specific miRNAs that modulate skeletal muscle atrophy in breast cancer conditions has not yet been explored. In this narrative review, we aim to identify miRNAs that could directly or indirectly influence skeletal muscle atrophy in breast cancer models to gain an updated perspective on potential therapeutic targets that could be modulated through resistance exercise training, aiming to mitigate the loss of skeletal muscle mass in breast cancer patients.


Sujet(s)
Tumeurs du sein , microARN , Muscles squelettiques , Amyotrophie , Humains , microARN/génétique , microARN/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Femelle , Amyotrophie/métabolisme , Amyotrophie/génétique , Amyotrophie/anatomopathologie , Amyotrophie/étiologie , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Animaux , Développement musculaire/génétique
19.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38928510

RÉSUMÉ

The decline in the function and mass of skeletal muscle during aging or other pathological conditions increases the incidence of aging-related secondary diseases, ultimately contributing to a decreased lifespan and quality of life. Much effort has been made to surmise the molecular mechanisms underlying muscle atrophy and develop tools for improving muscle function. Enhancing mitochondrial function is considered critical for increasing muscle function and health. This study is aimed at evaluating the effect of an aqueous extract of Gloiopeltis tenax (GTAE) on myogenesis and muscle atrophy caused by dexamethasone (DEX). The GTAE promoted myogenic differentiation, accompanied by an increase in peroxisome proliferator-activated receptor γ coactivator α (PGC-1α) expression and mitochondrial content in myoblast cell culture. In addition, the GTAE alleviated the DEX-mediated myotube atrophy that is attributable to the Akt-mediated inhibition of the Atrogin/MuRF1 pathway. Furthermore, an in vivo study using a DEX-induced muscle atrophy mouse model demonstrated the efficacy of GTAE in protecting muscles from atrophy and enhancing mitochondrial biogenesis and function, even under conditions of atrophy. Taken together, this study suggests that the GTAE shows propitious potential as a nutraceutical for enhancing muscle function and preventing muscle wasting.


Sujet(s)
Dexaméthasone , Développement musculaire , Amyotrophie , Extraits de plantes , Animaux , Amyotrophie/induit chimiquement , Amyotrophie/métabolisme , Amyotrophie/traitement médicamenteux , Amyotrophie/anatomopathologie , Dexaméthasone/effets indésirables , Dexaméthasone/pharmacologie , Développement musculaire/effets des médicaments et des substances chimiques , Souris , Extraits de plantes/pharmacologie , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Myoblastes/effets des médicaments et des substances chimiques , Myoblastes/métabolisme , Lignée cellulaire , Protéines du muscle/métabolisme , Mâle , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/métabolisme , Fibres musculaires squelettiques/anatomopathologie , Souris de lignée C57BL , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Rhodophyta
20.
J Nutr Sci Vitaminol (Tokyo) ; 70(3): 219-227, 2024.
Article de Anglais | MEDLINE | ID: mdl-38945887

RÉSUMÉ

This study investigated the protective effect of carnosine and its components (L-histidine and ß-alanine [HA]) against dexamethasone (Dex)-induced muscle atrophy in C2C12 myotubes. Myotubes were treated with Dex (10 µM) to induce muscle atrophy manifested by decreased myotube diameter, low myosin heavy chain content, and increased expression of muscle atrophy-associated ubiquitin ligases (Atrogin-1, MuRF-1, and Cbl-b). Carnosine (20 mM) treatment significantly improved the myotube diameter and MyHC protein expression level in Dex-treated C2C12 myotubes. It also downregulated the expression of Atrogin-1, MuRF-1, and Cbl-b and suppressed the expression of forkhead box O3 (FoxO3a) mediated by Dex. Furthermore, reactive oxygen species production was increased by Dex but was ameliorated by carnosine treatment. However, HA (20 mM), the component of carnosine, treatment was found ineffective in preventing Dex-induced protein damage. Therefore, based on above results it can be suggested that carnosine could be a potential therapeutic agent to prevent Dex-induced muscle atrophy compared to its components HA.


Sujet(s)
Carnosine , Dexaméthasone , Fibres musculaires squelettiques , Protéines du muscle , Amyotrophie , Espèces réactives de l'oxygène , SKP cullin F-box protein ligases , Carnosine/pharmacologie , Dexaméthasone/pharmacologie , Amyotrophie/induit chimiquement , Amyotrophie/prévention et contrôle , Amyotrophie/métabolisme , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/métabolisme , Animaux , Souris , Protéines du muscle/métabolisme , Lignée cellulaire , Espèces réactives de l'oxygène/métabolisme , SKP cullin F-box protein ligases/métabolisme , Ubiquitin-protein ligases/métabolisme , Protéine O3 à motif en tête de fourche/métabolisme , Protéines à motif tripartite/métabolisme , Chaînes lourdes de myosine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE