Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.327
Filtrer
1.
Trials ; 25(1): 356, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38835083

RÉSUMÉ

BACKGROUND: Patients diagnosed with pancreatic, biliary tract, and liver cancer often suffer from a progressive loss of muscle mass. Given the considerable functional impairments in these patients, high musculoskeletal weight loads may not be well tolerated by all individuals. The use of blood-flow restricted resistance training (BFR-T) which only requires low training loads may allow for a faster recovery of muscle due to avoidance of high levels of mechanical muscle stress associated with high-load resistance exercise. This study aims to investigate whether BFR-T can prevent or slow down the loss of skeletal muscle mass and enhance the functional capacity and mental health of patients with pancreatic, biliary tract, and liver cancer. METHODS: The PREV-Ex exercise trial is a multicenter two-armed randomized controlled trial. Patients will be randomized to an exercise program consisting of home-based low-load BFR-T during a combined pre- and postoperative period for a total of 6-10 weeks (prehabilitation and rehabilitation), or to a control group. Protein supplementation will be given to both groups to ensure adequate protein intake. The primary outcomes, skeletal muscle thickness and muscle cross-sectional area, will be assessed by ultrasound. Secondary outcomes include the following: (i) muscle catabolism-related and inflammatory bio-markers (molecular characteristics will be assessed from a vastus lateralis biopsy and blood samples will be obtained from a sub-sample of patients); (ii) patient-reported outcome measures (self-reported fatigue, health-related quality of life, and nutritional status will be assessed through validated questionnaires); (iii) physical fitness/performance/activity (validated tests will be used to evaluate physical function, cardiorespiratory fitness and maximal isometric muscle strength. Physical activity and sedentary behavior (assessed using an activity monitor); (iv) clinical outcomes: hospitalization rates and blood status will be recorded from the patients' medical records; (v) explorative outcomes of patients' experience of the exercise program which will be evaluated using focus group/individual interviews. DISCUSSION: It is worthwhile to investigate new strategies that have the potential to counteract the deterioration of skeletal muscle mass, muscle function, strength, and physical function, all of which have debilitating consequences for patients with pancreatic, biliary tract, and liver cancer. The expected findings could improve prognosis, help patients stay independent for longer, and possibly reduce treatment-related costs. TRIAL REGISTRATION: ClinicalTrials.gov NCT05044065. Registered on September 14, 2021.


Sujet(s)
Tumeurs des voies biliaires , Tumeurs du foie , Muscles squelettiques , Tumeurs du pancréas , Entraînement en résistance , Humains , Entraînement en résistance/méthodes , Tumeurs du pancréas/chirurgie , Tumeurs des voies biliaires/complications , Tumeurs des voies biliaires/chirurgie , Muscles squelettiques/physiopathologie , Tumeurs du foie/chirurgie , Essais contrôlés randomisés comme sujet , Études multicentriques comme sujet , Débit sanguin régional , Résultat thérapeutique , Qualité de vie , Force musculaire , Facteurs temps , Activité physique préopératoire , Amyotrophie/prévention et contrôle , Amyotrophie/étiologie , Amyotrophie/physiopathologie , Sarcopénie/prévention et contrôle , Sarcopénie/physiopathologie , Sarcopénie/étiologie
2.
Int J Mol Sci ; 25(9)2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38732203

RÉSUMÉ

Skeletal muscle unloading occurs during a wide range of conditions, from space flight to bed rest. The unloaded muscle undergoes negative functional changes, which include increased fatigue. The mechanisms of unloading-induced fatigue are far from complete understanding and cannot be explained by muscle atrophy only. In this review, we summarize the data concerning unloading-induced fatigue in different muscles and different unloading models and provide several potential mechanisms of unloading-induced fatigue based on recent experimental data. The unloading-induced changes leading to increased fatigue include both neurobiological and intramuscular processes. The development of intramuscular fatigue seems to be mainly contributed by the transformation of soleus muscle fibers from a fatigue-resistant, "oxidative" "slow" phenotype to a "fast" "glycolytic" one. This process includes slow-to-fast fiber-type shift and mitochondrial density decline, as well as the disruption of activating signaling interconnections between slow-type myosin expression and mitochondrial biogenesis. A vast pool of relevant literature suggests that these events are triggered by the inactivation of muscle fibers in the early stages of muscle unloading, leading to the accumulation of high-energy phosphates and calcium ions in the myoplasm, as well as NO decrease. Disturbance of these secondary messengers leads to structural changes in muscles that, in turn, cause increased fatigue.


Sujet(s)
Fatigue musculaire , Muscles squelettiques , Humains , Fatigue musculaire/physiologie , Animaux , Muscles squelettiques/métabolisme , Muscles squelettiques/physiopathologie , Amyotrophie/métabolisme , Amyotrophie/étiologie , Amyotrophie/anatomopathologie , Amyotrophie/physiopathologie
3.
Physiol Behav ; 281: 114575, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38692384

RÉSUMÉ

Fibromyalgia (FM) is characterized by chronic widespread musculoskeletal pain accompanied by fatigue and muscle atrophy. Although its etiology is not known, studies have shown that FM patients exhibit altered function of the sympathetic nervous system (SNS), which regulates nociception and muscle plasticity. Nevertheless, the precise SNS-mediated mechanisms governing hyperalgesia and skeletal muscle atrophy in FM remain unclear. Thus, we employed two distinct FM-like pain models, involving intramuscular injections of acidic saline (pH 4.0) or carrageenan in prepubertal female rats, and evaluated the catecholamine content, adrenergic signaling and overall muscle proteolysis. Subsequently, we assessed the contribution of the SNS to the development of hyperalgesia and muscle atrophy in acidic saline-injected rats treated with clenbuterol (a selective ß2-adrenergic receptor agonist) and in animals maintained under baseline conditions and subjected to epinephrine depletion through adrenodemedullation (ADM). Seven days after inducing an FM-like model with acidic saline or carrageenan, we observed widespread mechanical hyperalgesia along with loss of strength and/or muscle mass. These changes were associated with reduced catecholamine content, suggesting a common underlying mechanism. Notably, treatment with a ß2-agonist alleviated hyperalgesia and prevented muscle atrophy in acidic saline-induced FM-like pain, while epinephrine depletion induced mechanical hyperalgesia and increased muscle proteolysis in animals under baseline conditions. Together, the results suggest that reduced sympathetic activity is involved in the development of pain and muscle atrophy in the murine model of FM analyzed.


Sujet(s)
Clenbutérol , Modèles animaux de maladie humaine , Fibromyalgie , Hyperalgésie , Amyotrophie , Système nerveux sympathique , Animaux , Femelle , Fibromyalgie/anatomopathologie , Fibromyalgie/physiopathologie , Amyotrophie/anatomopathologie , Amyotrophie/physiopathologie , Hyperalgésie/physiopathologie , Hyperalgésie/anatomopathologie , Système nerveux sympathique/physiopathologie , Système nerveux sympathique/effets des médicaments et des substances chimiques , Système nerveux sympathique/anatomopathologie , Clenbutérol/pharmacologie , Rats , Carragénane/toxicité , Rat Sprague-Dawley , Douleur/anatomopathologie , Douleur/physiopathologie , Épinéphrine , Muscles squelettiques/anatomopathologie , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/physiopathologie , Catécholamines/métabolisme , Agonistes bêta-adrénergiques/pharmacologie
4.
J Appl Physiol (1985) ; 136(5): 1015-1039, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38328821

RÉSUMÉ

The efficacy of the NASA SPRINT exercise countermeasures program for quadriceps (vastus lateralis) and triceps surae (soleus) skeletal muscle health was investigated during 70 days of simulated microgravity. Individuals completed 6° head-down-tilt bedrest (BR, n = 9), bedrest with resistance and aerobic exercise (BRE, n = 9), or bedrest with resistance and aerobic exercise and low-dose testosterone (BRE + T, n = 8). All groups were periodically tested for muscle (n = 9 times) and aerobic (n = 4 times) power during bedrest. In BR, surprisingly, the typical bedrest-induced decrements in vastus lateralis myofiber size and power were either blunted (myosin heavy chain, MHC I) or eliminated (MHC IIa), along with no change (P > 0.05) in %MHC distribution and blunted quadriceps atrophy. In BRE, MHC I (vastus lateralis and soleus) and IIa (vastus lateralis) contractile performance was maintained (P > 0.05) or increased (P < 0.05). Vastus lateralis hybrid fiber percentage was reduced (P < 0.05) and energy metabolism enzymes and capillarization were generally maintained (P > 0.05), while not all of these positive responses were observed in the soleus. Exercise offsets 100% of quadriceps and approximately two-thirds of soleus whole muscle mass loss. Testosterone (BRE + T) did not provide any benefit over exercise alone for either muscle and for some myocellular parameters appeared detrimental. In summary, the periodic testing likely provided a partial exercise countermeasure for the quadriceps in the bedrest group, which is a novel finding given the extremely low exercise dose. The SPRINT exercise program appears to be viable for the quadriceps; however, refinement is needed to completely protect triceps surae myocellular and whole muscle health for astronauts on long-duration spaceflights.NEW & NOTEWORTHY This study provides unique exercise countermeasures development information for astronauts on long-duration spaceflights. The NASA SPRINT program was protective for quadriceps myocellular and whole muscle health, whereas the triceps surae (soleus) was only partially protected as has been shown with other programs. The bedrest control group data may provide beneficial information for overall exercise dose and targeting fast-twitch muscle fibers. Other unique approaches for the triceps surae are needed to supplement existing exercise programs.


Sujet(s)
Exercice physique , Muscles squelettiques , Chaînes lourdes de myosine , Muscle quadriceps fémoral , Simulation d'apesanteur , Humains , Mâle , Muscle quadriceps fémoral/physiologie , Muscle quadriceps fémoral/métabolisme , Simulation d'apesanteur/méthodes , Adulte , Exercice physique/physiologie , Chaînes lourdes de myosine/métabolisme , Muscles squelettiques/physiologie , Muscles squelettiques/métabolisme , NASA (USA) , États-Unis , Alitement/effets indésirables , Testostérone/métabolisme , Testostérone/sang , Vol spatial/méthodes , Amyotrophie/prévention et contrôle , Amyotrophie/physiopathologie , Entraînement en résistance/méthodes , Impesanteur/effets indésirables , Force musculaire/physiologie
5.
Age Ageing ; 51(10)2022 10 06.
Article de Anglais | MEDLINE | ID: mdl-36315433

RÉSUMÉ

INTRODUCTION: Significant losses of muscle mass and function occur after major abdominal surgery. Neuromuscular electrical stimulation (NMES) has been shown to reduce muscle atrophy in some patient groups, but evidence in post-operative patients is limited. This study assesses the efficacy of NMES for attenuating muscle atrophy and functional declines following major abdominal surgery in older adults. METHODS: Fifteen patients undergoing open colorectal resection completed a split body randomised control trial. Patients' lower limbs were randomised to control (CON) or NMES (STIM). The STIM limb underwent 15 minutes of quadriceps NMES twice daily on post-operative days (PODs) 1-4. Ultrasound measurements of Vastus Lateralis cross-sectional area (CSA) and muscle thickness (MT) were made preoperatively and on POD 5, as was dynamometry to determine knee extensor strength (KES). Change in CSA was the primary outcome. All outcomes were statistically analysed using linear mixed models. RESULTS: NMES significantly reduced the loss of CSA (-2.52 versus -9.16%, P < 0.001), MT (-2.76 versus -8.145, P = 0.001) and KES (-10.35 versus -19.69%, P = 0.03) compared to CON. No adverse events occurred, and patients reported that NMES caused minimal or no discomfort and felt that ~90-minutes of NMES daily would be tolerable. DISCUSSION: NMES reduces losses of muscle mass and function following major abdominal surgery, and as such, may be the promising tool for post-operative recovery. This is important in preventing long-term post-operative dependency, especially in the increasingly frail older patients undergoing major abdominal surgery. Further studies should establish the efficacy of bilateral NMES for improving patient-centred outcomes.


Sujet(s)
Électrothérapie , Force musculaire , Amyotrophie , Complications postopératoires , Muscle quadriceps fémoral , Sujet âgé , Humains , Stimulation électrique , Électrothérapie/effets indésirables , Électrothérapie/méthodes , Articulation du genou , Force musculaire/physiologie , Amyotrophie/étiologie , Amyotrophie/physiopathologie , Amyotrophie/prévention et contrôle , Muscle quadriceps fémoral/imagerie diagnostique , Muscle quadriceps fémoral/physiologie , Soins postopératoires , Complications postopératoires/prévention et contrôle , Colectomie/effets indésirables
6.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article de Anglais | MEDLINE | ID: mdl-35217605

RÉSUMÉ

The mechanoenzyme dynamin 2 (DNM2) is crucial for intracellular organization and trafficking. DNM2 is mutated in dominant centronuclear myopathy (DNM2-CNM), a muscle disease characterized by defects in organelle positioning in myofibers. It remains unclear how the in vivo functions of DNM2 are regulated in muscle. Moreover, there is no therapy for DNM2-CNM to date. Here, we overexpressed human amphiphysin 2 (BIN1), a membrane remodeling protein mutated in other CNM forms, in Dnm2RW/+ and Dnm2RW/RW mice modeling mild and severe DNM2-CNM, through transgenesis or with adeno-associated virus (AAV). Increasing BIN1 improved muscle atrophy and main histopathological features of Dnm2RW/+ mice and rescued the perinatal lethality and survival of Dnm2RW/RW mice. In vitro experiments showed that BIN1 binds and recruits DNM2 to membrane tubules, and that the BIN1-DNM2 complex regulates tubules fission. Overall, BIN1 is a potential therapeutic target for dominant centronuclear myopathy linked to DNM2 mutations.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Dynamine-II/physiologie , Amyotrophie/physiopathologie , Maladies musculaires/anatomopathologie , Protéines nucléaires/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Animaux , Dynamine-II/génétique , Dynamine-II/métabolisme , Humains , Souris , Souris knockout , Liaison aux protéines
7.
BMC Nephrol ; 23(1): 72, 2022 02 21.
Article de Anglais | MEDLINE | ID: mdl-35189838

RÉSUMÉ

BACKGROUND: Instrumental gait analysis in nephrology is widely neglected, although patients with chronic kidney disease (CKD) show brain changes due to cerebrovascular disease and metabolic disorders that can potentially influence gait quality. Our study assesses the association between CKD stages and gait parameters, to understand the prevalent status of brain related gait parameters (i.e. variability) and of performance related parameters (i.e. gait speed, stride length). We hypothesize that gait changes are detectable already in early stages of CKD. METHODS: Forty-five participants distributed in 5 CKD severity groups underwent an instrumental gait analysis via a triaxial accelerometer affixed to the lower trunk under single- and dual-task conditions. In addition to spatio-temporal parameters, variability and dual-task cost of gait were extracted. A battery of clinical assessments was conducted with the aim of helping to better explain the findings of the gait analysis. A correlation analysis was made to investigate a linear relation between gait parameters and CKD severity. RESULTS: Statistically significant correlations (Pearson correlation coefficient) with CKD severity were found for gait speed (p < 0.01, r = -0.55, 95% CI [-0.73;-0.30]), stride length ( p < 0.01, r = -0.40, 95% CI [-0.62;-0.12]), step length (p < 0.01, r = -0.41, 95% CI [-0.63;-0.13], coefficient of variance (CV) of step length (p = 0.01, r = 0.36, 95% CI [0.08;0.59]), gait regularity (p < 0.01, r = -0.38, 95% CI [-0.61;-0.10]), dual-task cost of gait speed (p < 0.01, r = 0.40, 95% CI [0.13;0.62]) and dual-task cost of stride time (p = 0.03, r = 0.32, 95% CI [0.03;0.57]). Adjustment for age and gender confirmed all results except for gait regularity. With increasing severity of renal failure, Handgrip strength, Time for the Expanded Timed Get Up and Go test, executive functions, haemoglobin, and haematocrit, worsen. CONCLUSIONS: The correlation of CKD severity with spatio-temporal parameters (performance indices mainly relatable to peripheral functionality) and with variability of gait (related to central factors) supported by the results of the clinical assessments, suggests that gait disturbance in CKD patients is not only due to metabolic factors that lead to muscle wasting, but also to brain changes that affect motor control. This suggests that the treatment of renal disease should include cognitive aspects in addition to metabolic and functional factors.


Sujet(s)
Dysfonctionnement cognitif/complications , Troubles neurologiques de la marche/étiologie , Démarche/physiologie , Amyotrophie/physiopathologie , Insuffisance rénale chronique/physiopathologie , Insuffisance rénale chronique/psychologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Études transversales , Analyse de démarche , Humains , Adulte d'âge moyen , Indice de gravité de la maladie
8.
Am J Physiol Cell Physiol ; 322(3): C382-C394, 2022 03 01.
Article de Anglais | MEDLINE | ID: mdl-35044855

RÉSUMÉ

Sarcolipin (SLN) is a small regulatory protein that inhibits the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pump. When bound to SERCA, SLN reduces the apparent Ca2+ affinity of SERCA and uncouples SERCA Ca2+ transport from its ATP consumption. As such, SLN plays a direct role in altering skeletal muscle relaxation and energy expenditure. Interestingly, the expression of SLN is dynamic during times of muscle adaptation, in that large increases in SLN content are found in response to development, atrophy, overload, and disease. Several groups have suggested that increases in SLN, especially in dystrophic muscle, are deleterious as it may reduce muscle function and exacerbate already abhorrent intracellular Ca2+ levels. However, there is also significant evidence to show that increased SLN content is a beneficial adaptive mechanism that protects the SERCA pump and activates Ca2+ signaling and adaptive remodeling during times of cell stress. In this review, we first discuss the role for SLN in healthy muscle during both development and overload, where SLN has been shown to activate Ca2+ signaling to promote mitochondrial biogenesis, fiber-type shifts, and muscle hypertrophy. Then, with respect to muscle disease, we summarize the discrepancies in the literature as to whether SLN upregulation is adaptive or maladaptive in nature. This review is the first to offer the concept of SLN hormesis in muscle disease, wherein both too much and too little SLN are detrimental to muscle health. Finally, the underlying mechanisms which activate SLN upregulation are discussed, specifically acknowledging a potential positive feedback loop between SLN and Ca2+ signaling molecules.


Sujet(s)
Développement musculaire , Protéines du muscle/métabolisme , Muscles squelettiques/enzymologie , Amyotrophie/enzymologie , Dystrophies musculaires/enzymologie , Protéolipides/métabolisme , Sarcoplasmic Reticulum Calcium-Transporting ATPases/métabolisme , Animaux , Signalisation calcique , Humains , Mitochondries du muscle/métabolisme , Mitochondries du muscle/anatomopathologie , Muscles squelettiques/anatomopathologie , Muscles squelettiques/physiopathologie , Amyotrophie/anatomopathologie , Amyotrophie/physiopathologie , Dystrophies musculaires/anatomopathologie , Dystrophies musculaires/physiopathologie
10.
Nat Commun ; 13(1): 149, 2022 01 10.
Article de Anglais | MEDLINE | ID: mdl-35013221

RÉSUMÉ

Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.


Sujet(s)
Cachexie/génétique , Fibrose endomyocardique/génétique , Défaillance cardiaque/génétique , Protéines du muscle/génétique , Muscles squelettiques/métabolisme , Amyotrophie/génétique , Facteurs de transcription/génétique , 2',3'-Cyclic nucleotide 3'-phosphodiesterase/génétique , 2',3'-Cyclic nucleotide 3'-phosphodiesterase/métabolisme , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Cachexie/métabolisme , Cachexie/physiopathologie , Cachexie/prévention et contrôle , Études cas-témoins , Cyclic AMP-Dependent Protein Kinases/génétique , Cyclic AMP-Dependent Protein Kinases/métabolisme , Cyclic GMP-Dependent Protein Kinases/génétique , Cyclic GMP-Dependent Protein Kinases/métabolisme , Modèles animaux de maladie humaine , Fibrose endomyocardique/métabolisme , Fibrose endomyocardique/physiopathologie , Fibrose endomyocardique/prévention et contrôle , Femelle , Régulation de l'expression des gènes , Défaillance cardiaque/métabolisme , Défaillance cardiaque/physiopathologie , Défaillance cardiaque/prévention et contrôle , Tests de la fonction cardiaque , Humains , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Protéines du muscle/agonistes , Protéines du muscle/antagonistes et inhibiteurs , Protéines du muscle/déficit , Amyotrophie/métabolisme , Amyotrophie/physiopathologie , Amyotrophie/prévention et contrôle , Myocarde/métabolisme , Myocarde/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction du signal , Facteurs de transcription/agonistes , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/déficit
11.
Nutrients ; 13(12)2021 Dec 08.
Article de Anglais | MEDLINE | ID: mdl-34959937

RÉSUMÉ

Sarcopenia, also known as skeletal muscle atrophy, is characterized by significant loss of muscle mass and strength. Oyster (Crassostrea gigas) hydrolysates have anti-cancer, antioxidant, and anti-inflammation properties. However, the anti-sarcopenic effect of oyster hydrolysates remains uninvestigated. Therefore, we prepared two different oyster hydrolysates, namely TGPN and PNY. This study aimed to determine the anti-muscle atrophy efficacy and molecular mechanisms of TGPN and PNY on both C2C12 cell lines and mice. In vitro, the TGPN and PNY recovered the dexamethasone-induced reduction in the myotube diameters. In vivo, TGPN and PNY administration not only improved grip strength and exercise endurance, but also attenuated the loss of muscle mass and muscle fiber cross-sectional area. Mechanistically, TGPN and PNY increased the expression of protein synthesis-related protein levels via phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of the rapamycin pathway, and reduced the expression of protein degradation-related protein levels via the PI3K/Akt/forkhead box O pathway. Also, TGPN and PNY stimulated NAD-dependent deacetylase sirtuin-1(SIRT1), peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), nuclear respiratory factor 1,2, mitochondrial transcription factor A, along with mitochondrial DNA content via SIRT1/PGC-1α signaling. These findings suggest oyster hydrolysates could be used as a valuable natural material that inhibits skeletal muscle atrophy via regulating protein turnover and mitochondrial biogenesis.


Sujet(s)
Mitochondries/effets des médicaments et des substances chimiques , Protéines du muscle/métabolisme , Amyotrophie/traitement médicamenteux , Amyotrophie/métabolisme , Biogenèse des organelles , Ostreidae/composition chimique , Hydrolysats de protéines/pharmacologie , Hydrolysats de protéines/usage thérapeutique , Sarcopénie/traitement médicamenteux , Sarcopénie/métabolisme , Animaux , Cellules cultivées , Tolérance à l'effort/effets des médicaments et des substances chimiques , Force de la main , Souris , Amyotrophie/étiologie , Amyotrophie/physiopathologie , Myoblastes squelettiques , Hydrolysats de protéines/isolement et purification , Sarcopénie/étiologie , Sarcopénie/physiopathologie
12.
Sci Rep ; 11(1): 21861, 2021 11 08.
Article de Anglais | MEDLINE | ID: mdl-34750405

RÉSUMÉ

We examined in a rat model of Gulf War illness (GWI), the potential of (-)-epicatechin (Epi) to reverse skeletal muscle (SkM) atrophy and dysfunction, decrease mediators of inflammation and normalize metabolic perturbations. Male Wistar rats (n = 15) were provided orally with pyridostigmine bromide (PB) 1.3 mg/kg/day, permethrin (PM) 0.13 mg/kg/day (skin), DEET 40 mg/kg/day (skin) and were physically restrained for 5 min/day for 3 weeks. A one-week period ensued to fully develop the GWI-like profile followed by 2 weeks of either Epi treatment at 1 mg/kg/day by gavage (n = 8) or water (n = 7) for controls. A normal, control group (n = 15) was given vehicle and not restrained. At 6 weeks, animals were subjected to treadmill and limb strength testing followed by euthanasia. SkM and blood sampling was used for histological, biochemical and plasma pro-inflammatory cytokine and metabolomics assessments. GWI animals developed an intoxication profile characterized SkM atrophy and loss of function accompanied by increases in modulators of muscle atrophy, degradation markers and plasma pro-inflammatory cytokine levels. Treatment of GWI animals with Epi yielded either a significant partial or full normalization of the above stated indicators relative to normal controls. Plasma metabolomics revealed that metabolites linked to inflammation and SkM waste pathways were dysregulated in the GWI group whereas Epi, attenuated such changes. In conclusion, in a rat model of GWI, Epi partially reverses detrimental changes in SkM structure including modulators of atrophy, inflammation and select plasma metabolites yielding improved function.


Sujet(s)
Catéchine/usage thérapeutique , Syndrome de la guerre du Golfe/traitement médicamenteux , Animaux , Compléments alimentaires , Modèles animaux de maladie humaine , Fatigue/traitement médicamenteux , Fatigue/physiopathologie , Humains , Mâle , Métabolome/effets des médicaments et des substances chimiques , Développement musculaire/effets des médicaments et des substances chimiques , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/anatomopathologie , Muscles squelettiques/physiopathologie , Amyotrophie/traitement médicamenteux , Amyotrophie/anatomopathologie , Amyotrophie/physiopathologie , Syndrome de la guerre du Golfe/anatomopathologie , Syndrome de la guerre du Golfe/physiopathologie , Rats , Rat Wistar
13.
Cells ; 10(11)2021 10 25.
Article de Anglais | MEDLINE | ID: mdl-34831102

RÉSUMÉ

Loss of myofibers during muscle atrophy affects functional capacity and quality of life. Dexamethasone, an inducer of rapid atrophy of skeletal myofibers, has been studied as a glucocorticoid receptor in muscle atrophy or motor neurodegeneration. In this study, we examined dexamethasone-induced muscle atrophy using zebrafish (Danio rerio), a vertebrate model, and assessed whether administration of Lepidium meyenii (maca) as a dietary supplement can prevent muscle atrophy. Changes in skeletal myofibers in zebrafish were evaluated after exposure to dexamethasone for different periods and at different concentrations. Under optimized conditions, zebrafish pre-fed with maca for 3 days were exposed to 0.01% dexamethasone for 1 h/day for 7 days. Thereafter, myofiber loss, damaged muscle contractile proteins, and abnormal exploratory behavior due to the structural and functional impairment of skeletal muscle associated with muscle atrophy were investigated using hematoxylin-eosin, immunofluorescence staining, and behavioral analyses. Our findings suggest that dexamethasone induces muscle atrophy in zebrafish, inhibiting exploratory behavior by inducing myofiber loss, inhibiting muscle contraction, and causing changes in endurance and velocity. Thus, the zebrafish model can be used to screen pharmaceutical agents and to study muscle atrophy. Furthermore, maca is a potential dietary supplement to prevent muscle atrophy, as it protects muscle fibers.


Sujet(s)
Dexaméthasone/effets indésirables , Lepidium/composition chimique , Amyotrophie/induit chimiquement , Amyotrophie/prévention et contrôle , Extraits de plantes/usage thérapeutique , Animaux , Comportement animal/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Comportement d'exploration/effets des médicaments et des substances chimiques , Contraction musculaire/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/effets des médicaments et des substances chimiques , Fibres musculaires squelettiques/anatomopathologie , Protéines du muscle/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/physiopathologie , Extraits de plantes/pharmacologie , Natation/physiologie , Danio zébré
14.
Sci Rep ; 11(1): 20899, 2021 10 22.
Article de Anglais | MEDLINE | ID: mdl-34686697

RÉSUMÉ

We tested the hypothesis that swim training reverses the impairment of Akt/FOXO3a signaling, ameliorating muscle atrophy in ALS mice. Transgenic male mice B6SJL-Tg (SOD1G93A) 1Gur/J were used as the ALS model (n = 35), with wild-type B6SJL (WT) mice as controls (n = 7). ALS mice were analyzed before ALS onset, at ALS onset, and at terminal ALS. Levels of insulin/Akt signaling pathway proteins were determined, and the body and tibialis anterior muscle mass and plasma creatine kinase. Significantly increased levels of FOXO3a in ALS groups (from about 13 to 21-fold) compared to WT mice were observed. MuRF1 levels in the ONSET untrained group (12.0 ± 1.7 AU) were significantly higher than in WT mice (1.12 ± 0.2 AU) and in the BEFORE ALS group (3.7 ± 0.9 AU). This was associated with body mass and skeletal muscle mass reduction. Swim training significantly ameliorated the reduction of skeletal muscle mass in both TERMINAL groups (p < 0.001) and partially reversed changes in the levels of Akt signaling pathway proteins. These findings shed light on the swimming-induced attenuation of skeletal muscle atrophy in ALS with possible practical implications for anti-cachexia approaches.


Sujet(s)
Sclérose latérale amyotrophique/métabolisme , Sclérose latérale amyotrophique/physiopathologie , Muscles squelettiques/physiologie , Amyotrophie/physiopathologie , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/physiologie , Natation/physiologie , Animaux , Modèles animaux de maladie humaine , Protéine O3 à motif en tête de fourche/métabolisme , Humains , Mâle , Souris , Souris transgéniques , Muscles squelettiques/métabolisme , Amyotrophie/métabolisme , Superoxide dismutase-1/métabolisme , Protéines à motif tripartite/métabolisme
15.
BMC Pulm Med ; 21(1): 314, 2021 Oct 08.
Article de Anglais | MEDLINE | ID: mdl-34625059

RÉSUMÉ

BACKGROUND: Diaphragm atrophy and dysfunction is a major problem among critically ill patients on mechanical ventilation. Ventilator-induced diaphragmatic dysfunction is thought to play a major role, resulting in a failure of weaning. Stimulation of the phrenic nerves and resulting diaphragm contraction could potentially prevent or treat this atrophy. The subject of this study is to determine the effectiveness of diaphragm stimulation in preventing atrophy by measuring changes in its thickness. METHODS: A total of 12 patients in the intervention group and 10 patients in the control group were enrolled. Diaphragm thickness was measured by ultrasound in both groups at the beginning of study enrollment (hour 0), after 24 hours, and at study completion (hour 48). The obtained data were then statistically analyzed and both groups were compared. RESULTS: The results showed that the baseline diaphragm thickness in the interventional group was (1.98 ± 0.52) mm and after 48 hours of phrenic nerve stimulation increased to (2.20 ± 0.45) mm (p=0.001). The baseline diaphragm thickness of (2.00 ± 0.33) mm decreased in the control group after 48 hours of mechanical ventilation to (1.72 ± 0.20) mm (p<0.001). CONCLUSIONS: Our study demonstrates that induced contraction of the diaphragm by pacing the phrenic nerve not only reduces the rate of its atrophy during mechanical ventilation but also leads to an increase in its thickness - the main determinant of the muscle strength required for spontaneous ventilation and successful ventilator weaning. TRIAL REGISTRATION: The study was registered with ClinicalTrials.gov (18/06/2018, NCT03559933, https://clinicaltrials.gov/ct2/show/NCT03559933 ).


Sujet(s)
Muscle diaphragme/physiopathologie , Électrothérapie/méthodes , Nerf phrénique/physiologie , Ventilation artificielle/effets indésirables , Insuffisance respiratoire/thérapie , Sujet âgé , Maladie grave , Muscle diaphragme/imagerie diagnostique , Électrothérapie/instrumentation , Femelle , Humains , Mâle , Adulte d'âge moyen , Amyotrophie/étiologie , Amyotrophie/physiopathologie , Études prospectives , Ventilation artificielle/méthodes , Échographie
16.
Physiol Rep ; 9(19): e15042, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34612585

RÉSUMÉ

Ground-based animal models have been used extensively to understand the effects of microgravity on various physiological systems. Among them, hindlimb suspension (HLS), developed in 1979 in rats, remains the gold-standard and allows researchers to study the consequences of total unloading of the hind limbs while inducing a cephalic fluid shift. While this model has already brought valuable insights to space biology, few studies have directly compared functional decrements in the muscles of males and females during HLS. We exposed 28 adult Wistar rats (14 males and 14 females) to 14 days of HLS or normal loading (NL) to better assess how sex impacts disuse-induced muscle deconditioning. Females better maintained muscle function during HLS than males, as shown by a more moderate reduction in grip strength at 7 days (males: -37.5 ± 3.1%, females: -22.4 ± 6.5%, compared to baseline), that remains stable during the second week of unloading (males: -53.3 ± 5.7%, females: -22.4 ± 5.5%, compared to day 0) while the males exhibit a steady decrease over time (effect of sex × loading p = 0.0002, effect of sex × time × loading p = 0.0099). This was further supported by analyzing the force production in response to a tetanic stimulus. Further functional analyses using force production were also shown to correspond to sex differences in relative loss of muscle mass and CSA. Moreover, our functional data were supported by histomorphometric analyzes, and we highlighted differences in relative muscle loss and CSA. Specifically, female rats seem to experience a lesser muscle deconditioning during disuse than males thus emphasizing the need for more studies that will assess male and female animals concomitantly to develop tailored, effective countermeasures for all astronauts.


Sujet(s)
Force de la main/physiologie , Suspension des membres postérieurs , Force musculaire/physiologie , Muscles squelettiques/physiopathologie , Amyotrophie/physiopathologie , Caractères sexuels , Animaux , Femelle , Mâle , Rats , Rat Wistar
17.
Sci Rep ; 11(1): 19116, 2021 09 27.
Article de Anglais | MEDLINE | ID: mdl-34580406

RÉSUMÉ

Obesity induces skeletal muscle dysfunction. The pathogenesis of which appears to substantially involve mitochondrial dysfunction, arising from impaired quality control. Exercise is a major therapeutic strategy against muscle dysfunction. Trimetazidine, a partial inhibitor of lipid oxidation, has been proposed as a metabolic modulator for several cardiovascular pathologies. However, the effects of Trimetazidine on regulating skeletal muscle function are largely unknown. Our present study used cell culture and obese mice models to test a novel hypothesis that Trimetazidine could improve muscle atrophy with similar results to exercise. In C2C12 cells, high palmitic acid-induced atrophy and mitochondrial dysfunction, which could be reversed by the treatment of Trimetazidine. In our animal models, with high-fat diet-induced obesity associated with skeletal muscle atrophy, Trimetazidine prevented muscle dysfunction, corrected metabolic abnormalities, and improved mitochondrial quality control and mitochondrial functions similarly to exercise. Thus, our study suggests that Trimetazidine successfully mimics exercise to enhance mitochondrial quality control leading to improved high-fat diet-induced muscle dysfunction.


Sujet(s)
Mitochondries/effets des médicaments et des substances chimiques , Amyotrophie/thérapie , Obésité/thérapie , Conditionnement physique d'animal , Trimétazidine/pharmacologie , Animaux , Lignée cellulaire , Association thérapeutique , Alimentation riche en graisse/effets indésirables , Modèles animaux de maladie humaine , Humains , Métabolisme lipidique/effets des médicaments et des substances chimiques , Mâle , Souris , Mitochondries/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/physiopathologie , Amyotrophie/étiologie , Amyotrophie/métabolisme , Amyotrophie/physiopathologie , Myoblastes , Obésité/étiologie , Obésité/métabolisme , Oxydoréduction , Acide palmitique/toxicité , Trimétazidine/usage thérapeutique
18.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-34502376

RÉSUMÉ

Inflammation induces a wide response of the neuroendocrine system, which leads to modifications in all the endocrine axes. The hypothalamic-growth hormone (GH)-insulin-like growth factor-1 (IGF-1) axis is deeply affected by inflammation, its response being characterized by GH resistance and a decrease in circulating levels of IGF-1. The endocrine and metabolic responses to inflammation allow the organism to survive. However, in chronic inflammatory conditions, the inhibition of the hypothalamic-GH-IGF-1 axis contributes to the catabolic process, with skeletal muscle atrophy and cachexia. Here, we review the changes in pituitary GH secretion, IGF-1, and IGF-1 binding protein-3 (IGFBP-3), as well as the mechanism that mediated those responses. The contribution of GH and IGF-1 to muscle wasting during inflammation has also been analyzed.


Sujet(s)
Cachexie/métabolisme , Protéine-3 de liaison aux IGF/métabolisme , Facteur de croissance IGF-I/métabolisme , Cachexie/physiopathologie , Hormone de croissance/métabolisme , Hormone de croissance humaine/métabolisme , Humains , Hypothalamus/métabolisme , Inflammation/physiopathologie , Insuline/métabolisme , Protéine-3 de liaison aux IGF/physiologie , Facteur de croissance IGF-I/physiologie , Amyotrophie/métabolisme , Amyotrophie/physiopathologie
19.
Pharmacol Res ; 171: 105798, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34352400

RÉSUMÉ

Skeletal muscle atrophy occurs in response to various pathophysiological stimuli, including disuse, aging, and neuromuscular disorders, mainly due to an imbalance of anabolic/catabolic signaling. Branched Chain Amino Acids (BCAAs: leucine, isoleucine, valine) supplements can be beneficial for counteracting muscle atrophy, in virtue of their reported anabolic properties. Here, we carried out a proof-of-concept study to assess the in vivo/ex vivo effects of a 4-week treatment with BCAAs on disuse-induced atrophy, in a murine model of hind limb unloading (HU). BCAAs were formulated in drinking water, alone, or plus two equivalents of L-Alanine (2 ALA) or the dipeptide L-Alanyl-L-Alanine (Di-ALA), to boost BCAAs bioavailability. HU mice were characterized by reduction of body mass, decrease of soleus - SOL - muscle mass and total protein, alteration of postural muscles architecture and fiber size, dysregulation of atrophy-related genes (Atrogin-1, MuRF-1, mTOR, Mstn). In parallel, we provided new robust readouts in the HU murine model, such as impaired in vivo isometric torque and ex vivo SOL muscle contractility and elasticity, as well as altered immune response. An acute pharmacokinetic study confirmed that L-ALA, also as dipeptide, enhanced plasma exposure of BCAAs. Globally, the most sensitive parameters to BCAAs action were muscle atrophy and myofiber cross-sectional area, muscle force and compliance to stress, protein synthesis via mTOR and innate immunity, with the new BCAAs + Di-ALA formulation being the most effective treatment. Our results support the working hypothesis and highlight the importance of developing innovative formulations to optimize BCAAs biodistribution.


Sujet(s)
Alanine/usage thérapeutique , Acides aminés à chaine ramifiée/usage thérapeutique , Dipeptides/usage thérapeutique , Amyotrophie/traitement médicamenteux , Alanine/pharmacocinétique , Acides aminés à chaine ramifiée/pharmacocinétique , Animaux , Dipeptides/pharmacocinétique , Modèles animaux de maladie humaine , Suspension des membres postérieurs , Mâle , Souris de lignée C57BL , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/anatomopathologie , Muscles squelettiques/physiologie , Amyotrophie/génétique , Amyotrophie/anatomopathologie , Amyotrophie/physiopathologie , Protéome/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques
20.
Sci Rep ; 11(1): 16317, 2021 08 11.
Article de Anglais | MEDLINE | ID: mdl-34381086

RÉSUMÉ

Diaphragm weakness affects up to 60% of ventilated patients leading to muscle atrophy, reduction of muscle fiber force via muscle fiber injuries and prolonged weaning from mechanical ventilation. Electromagnetic stimulation of the phrenic nerve can induce contractions of the diaphragm and potentially prevent and treat loss of muscular function. Recommended safety distance of electromagnetic coils is 1 m. The aim of this study was to investigate the magnetic flux density in a typical intensive care unit (ICU) setting. Simulation of magnetic flux density generated by a butterfly coil was performed in a Berlin ICU training center with testing of potential disturbance and heating of medical equipment. Approximate safety distances to surrounding medical ICU equipment were additionally measured in an ICU training center in Bern. Magnetic flux density declined exponentially with advancing distance from the stimulation coil. Above a coil distance of 300 mm with stimulation of 100% power the signal could not be distinguished from the surrounding magnetic background noise. Electromagnetic stimulation of the phrenic nerve for diaphragm contraction in an intensive care unit setting seems to be safe and feasible from a technical point of view with a distance above 300 mm to ICU equipment from the stimulation coil.


Sujet(s)
Nerf phrénique/physiopathologie , Gestion de la sécurité/méthodes , Berlin , Muscle diaphragme/physiopathologie , Phénomènes électromagnétiques , Humains , Unités de soins intensifs , Contraction musculaire/physiologie , Faiblesse musculaire/physiopathologie , Amyotrophie/physiopathologie , Ventilation artificielle/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...