Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Neuroendocrinology ; 112(7): 673-701, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34348331

RESUMEN

Sleep plays an essential role in human life. While sleep is a state elicited by the brain, its vital role reaches beyond maintaining brain health. Unhealthy sleeping habits have been associated with increased risk for inflammation, obesity, or diabetes. Evidence is emerging that sleep guides processes playing an important role in promoting the regulation of endocrine function involved in tissue regeneration and tissue remodelling. Thereby, sleep presumably is a critical factor contributing to the balance of core body tissues: bone, fat, and muscle mass. Given the increasing prevalence of various chronic diseases and comorbidities due to unhealthy lifestyle choices, sleep could be a key target to promote a healthy body composition up until old age. Here, we review the potential role of sleep and its underlying brain oscillations in body core tissues turnover. Specifically, we discuss potential underlying mechanisms linking sleep to body composition, both during rest and under challenging conditions. Among other described pathways, we highlight the possible role of the growth hormone that was found to be involved in the homeostasis of all core body tissues and has been strongly linked to brain activity dominating deep sleep, the so-called slow waves. Finally, we formulate important questions to be addressed in future research on the effect of sleep on body composition and specifically emphasize the importance of intervention studies to move from correlative to causal evidence.


Asunto(s)
Composición Corporal , Sueño , Densidad Ósea , Humanos , Músculos/metabolismo , Obesidad/metabolismo
2.
Eur J Appl Physiol ; 118(4): 847-862, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29423544

RESUMEN

PURPOSE: The myocellular response to hypoxia is primarily regulated by hypoxia-inducible factors (HIFs). HIFs thus conceivably are implicated in muscular adaptation to altitude training. Therefore, we investigated the effect of hypoxic versus normoxic training during a period of prolonged hypoxia ('living high') on muscle HIF activation during acute ischaemia. METHODS: Ten young male volunteers lived in normobaric hypoxia for 5 weeks (5 days per week, ~ 15.5 h per day, FiO2: 16.4-14.0%). One leg was trained in hypoxia (TRHYP, 12.3% FiO2) whilst the other leg was trained in normoxia (TRNOR, 20.9% FiO2). Training sessions (3 per week) consisted of intermittent unilateral knee extensions at 20-25% of the 1-repetition maximum. Before and after the intervention, a 10-min arterial occlusion and reperfusion of the leg was performed. Muscle oxygenation status was continuously measured by near-infrared spectroscopy. Biopsies were taken from m. vastus lateralis before and at the end of the occlusion. RESULTS: Irrespective of training, occlusion elevated the fraction of HIF-1α expressing myonuclei from ~ 54 to ~ 64% (P < 0.05). However, neither muscle HIF-1α or HIF-2α protein abundance, nor the expression of HIF-1α or downstream targets selected increased in any experimental condition. Training in both TRNOR and TRHYP raised muscular oxygen extraction rate upon occlusion by ~ 30%, whilst muscle hyperperfusion immediately following the occlusion increased by ~ 25% in either group (P < 0.05). CONCLUSION: Ten minutes of arterial occlusion increased HIF-1α-expressing myonuclei. However, neither normoxic nor hypoxic training during 'living high' altered muscle HIF translocation, stabilisation, or transcription in response to acute hypoxia induced by arterial occlusion.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Músculo Esquelético/metabolismo , Consumo de Oxígeno/fisiología , Adulto , Altitud , Entrenamiento de Intervalos de Alta Intensidad/métodos , Humanos , Masculino , ARN Mensajero/metabolismo
3.
FASEB J ; 28(2): 1022-34, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24200883

RESUMEN

Hypoxia-induced muscle wasting is a phenomenon often described with prolonged stays at high altitude, which has been attributed to altered protein metabolism. We hypothesized that acute normobaric hypoxia would induce a negative net protein balance by repressing anabolic and activating proteolytic signaling pathways at rest and postexercise and that those changes could be partially genetically determined. Eleven monozygotic twins participated in an experimental trial in normoxia and hypoxia (10.7% O2). Muscle biopsy samples were obtained before and after a 20-min moderate cycling exercise. In hypoxia at rest, autophagic flux was increased, as indicated by an increased microtubule-associated protein 1 light chain 3 type II/I (LC3-II/I) ratio (+25%) and LC3-II expression (+60%) and decreased p62/SQSTM1 expression (-25%; P<0.05), whereas exercise reversed those changes to a level similar to that with normoxia except for p62/SQSTM1, which was further decreased (P<0.05). Hypoxia also increased Bnip3 (+34%) and MAFbx (+18%) mRNA levels as well as REDD1 expression (+439%) and AMP-activated protein kinase phosphorylation (+22%; P<0.05). Among the molecular responses to hypoxia and/or exercise, high monozygotic similarity was found for REDD1, LC3-II, and LC3-II/I (P<0.05). Our results indicate that environmental hypoxia modulates protein metabolism at rest and after moderate exercise by primarily increasing markers of protein breakdown and, more specifically, markers of the autophagy-lysosomal system, with a modest genetic contribution.


Asunto(s)
Autofagia/fisiología , Hipoxia/fisiopatología , Proteínas Asociadas a Microtúbulos/metabolismo , Factores de Transcripción/metabolismo , Adulto , Western Blotting , Ejercicio Físico/fisiología , Genotipo , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Ligasas SKP Cullina F-box/genética , Proteínas Ligasas SKP Cullina F-box/metabolismo , Factores de Transcripción/genética , Adulto Joven
4.
Eur J Appl Physiol ; 115(6): 1219-31, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25577409

RESUMEN

PURPOSE: To investigate how acute environmental hypoxia regulates blood glucose and downstream intramuscular insulin signaling after a meal in healthy humans. METHODS: Fifteen subjects were exposed for 4 h to normoxia (NOR) or to normobaric hypoxia (HYP, FiO2 = 0.11) in a randomized order 40 min after consumption of a high glycemic meal. A muscle biopsy from m. vastus lateralis and a blood sample were taken before (T0), after 1 h (T60) and 4 h (T240) in NOR or HYP and blood glucose levels were measured before exposure and every 30 min. RESULTS: In HYP, blood glucose was reduced 100 min (110.1 ± 5.4 in NOR vs 89.5 ± 4.7 mg dl(-1) in HYP) and 130 min (98.7 ± 3.8 in NOR vs 85.6 ± 4.9 mg dl(-1) in HYP) after completion of a meal, which resulted in an 83 % lower AUC in HYP compared to NOR (p = 0.006). This coincided with 40 % lower GLUT4 protein in the cytosolic fraction (p = 0.013) and a tendency to increase in the crude membrane fraction (p = 0.070) in HYP compared to NOR. At T240, blood glucose concentration was similar between HYP and NOR, whereas plasma insulin as well as phosphorylation of muscle Akt and GSK-3 was ~2-fold higher in HYP compared to NOR (p < 0.05). In contrast, Rac1 protein was less abundant in the membrane fraction in HYP compared to NOR (p = 0.003), reflecting lower activation. CONCLUSION: Acute environmental hypoxia initially reduced blood glucose response to a meal, possibly via an increase in GLUT4 abundance at the sarcolemmal membrane. Later on, whole body insulin intolerance developed independently of defects in conventional insulin signaling in skeletal muscle.


Asunto(s)
Glucemia/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Hipoxia/metabolismo , Insulina/sangre , Músculo Esquelético/metabolismo , Oxígeno/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Carbohidratos de la Dieta/metabolismo , Grasas de la Dieta/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Resistencia a la Insulina , Masculino , Transducción de Señal , Adulto Joven
5.
Int J Sports Physiol Perform ; : 1-12, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849121

RESUMEN

OBJECTIVE: To (1) establish extensive physiological profiles of highly trained CrossFit® athletes using gold-standard tests and (2) investigate which physiological markers best correlate with CrossFit Open performance. METHODS: This study encompassed 60 participants (30 men and 30 women), all within the top 5% of the CrossFit Open, including 7 CrossFit semifinalists and 3 CrossFit Games finalists. Isokinetic dynamometers were employed to measure maximum isometric and isokinetic leg and trunk strength. Countermovement-jump height and maximum isometric midthigh-pull strength were assessed on a force plate. Peak oxygen uptake (VO2peak) was measured by a cardiopulmonary exercise test, and critical power and W' were evaluated during a 3-minute all-out test, both on a cycle ergometer. RESULTS: Male and female athletes' median (interquartile range) VO2peak was 4.64 (4.43, 4.80) and 3.21 (3.10, 3.29) L·min-1, critical power 314.5 (285.9, 343.6) and 221.3 (200.9, 238.9) W, and midthigh pull 3158 (2690, 3462) and 2035 (1728, 2347) N. Linear-regression analysis showed strong evidence for associations between different anthropometric variables and CrossFit Open performance in men and women, whereas for markers of cardiorespiratory fitness such as VO2peak, this was only true for women but not men. Conventional laboratory evaluations of strength, however, manifested minimal evidence for associations with CrossFit Open performance across both sexes. CONCLUSIONS: This study provides the first detailed insights into the physiology of high-performing CrossFit athletes and informs training optimization. Furthermore, the results emphasize the advantage of athletes with shorter limbs and suggest potential modifications to CrossFit Open workout designs to level the playing field for athletes across different anthropometric characteristics.

6.
Mol Metab ; 66: 101615, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36252815

RESUMEN

OBJECTIVE: Exercise enhances the sensitivity of mammalian target of rapamycin complex 1 (mTORC1) to amino acids, in particular leucine. How long this enhanced sensitivity lasts, and which mechanisms control enhanced leucine-mediated mTORC1 activation following exercise is currently unknown. METHODS: C57BL/6J mice were exercised for one night in a resistance-braked running wheel after a 12-day acclimatization period. Mice were gavaged with a submaximal dose of l-leucine or saline acutely or 48 h after exercise cessation, following 3 h food withdrawal. Muscles were excised 30 min after leucine administration. To study the contribution of mTORC1, we repeated those experiments but blocked mTORC1 activation using rapamycin immediately before the overnight running bout and one hour before the first dose of leucine. mTORC1 signaling, muscle protein synthesis and amino acid sensing machinery were assessed using immunoblot and qPCR. Leucine uptake was measured using L-[14C(U)]-leucine tracer labeling. RESULTS: When compared to sedentary conditions, leucine supplementation more potently activated mTORC1 and protein synthesis in acutely exercised muscle. This effect was observed in m. soleus but not in m. tibialis anterior nor m. plantaris. The synergistic effect in m. soleus was long-lasting as key downstream markers of mTORC1 as well as protein synthesis remained higher when leucine was administered 48 h after exercise. We found that exercise enhanced the expression of amino acid transporters and promoted uptake of leucine into the muscle, leading to higher free intramuscular leucine levels. This coincided with increased expression of activating transcription factor 4 (ATF4), a main transcriptional regulator of amino acid uptake and metabolism, and downstream activation of amino acid genes as well as leucyl-tRNA synthetase (LARS), a putative leucine sensor. Finally, blocking mTORC1 using rapamycin did not reduce expression and activation of ATF4, suggesting that the latter does not act downstream of mTORC1. Rather, we found a robust increase in eukaryotic initiation factor 2α (eIF2α) phosphorylation, suggesting that the integrated stress response pathway, rather than exercise-induced mTORC1 activation, drives long-term ATF4 expression in skeletal muscle after exercise. CONCLUSIONS: The enhanced sensitivity of mTORC1 to leucine is maintained at least 48 h after exercise. This shows that the anabolic window of opportunity for protein ingestion is not restricted to the first hours immediately following exercise. Increased mTORC1 sensitivity to leucine coincided with enhanced leucine influx into muscle and higher expression of genes involved in leucine sensing and amino acid metabolism. Also, exercise induced an increase in ATF4 protein expression. Altogether, these data suggest that muscular contractions switch on a coordinated program to enhance amino acid uptake as well as intramuscular sensing of key amino acids involved in mTORC1 activation and the stimulation of muscle protein synthesis.


Asunto(s)
Leucina , Diana Mecanicista del Complejo 1 de la Rapamicina , Condicionamiento Físico Animal , Animales , Ratones , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Aminoácidos/metabolismo , Leucina/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Proteínas Musculares , Sirolimus , Condicionamiento Físico Animal/fisiología
7.
Stem Cell Reports ; 17(2): 321-336, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34995499

RESUMEN

Genetic mutations in dystrophin manifest in Duchenne muscular dystrophy (DMD), the most commonly inherited muscle disease. Here, we report on reprogramming of fibroblasts from two DMD mouse models into induced myogenic progenitor cells (iMPCs) by MyoD overexpression in concert with small molecule treatment. DMD iMPCs proliferate extensively, while expressing myogenic stem cell markers including Pax7 and Myf5. Additionally, DMD iMPCs readily give rise to multinucleated myofibers that express mature skeletal muscle markers; however, they lack DYSTROPHIN expression. Utilizing an exon skipping-based approach with CRISPR/Cas9, we report on genetic correction of the dystrophin mutation in DMD iMPCs and restoration of protein expression in vitro. Furthermore, engraftment of corrected DMD iMPCs into the muscles of dystrophic mice restored DYSTROPHIN expression and contributed to the muscle stem cell reservoir. Collectively, our findings report on a novel in vitro cellular model for DMD and utilize it in conjunction with gene editing to restore DYSTROPHIN expression in vivo.


Asunto(s)
Reprogramación Celular/genética , Distrofina/metabolismo , Edición Génica/métodos , Distrofia Muscular de Duchenne/patología , Animales , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Modelos Animales de Enfermedad , Distrofina/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Desarrollo de Músculos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Mutación , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Células Madre/citología , Células Madre/metabolismo
8.
Nutrients ; 13(5)2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33919313

RESUMEN

Increased amino acid availability acutely stimulates protein synthesis partially via activation of mechanistic target of rapamycin complex 1 (mTORC1). Plant-and insect-based protein sources matched for total protein and/or leucine to animal proteins induce a lower postprandial rise in amino acids, but their effects on mTOR activation in muscle are unknown. C57BL/6J mice were gavaged with different protein solutions: whey, a pea-rice protein mix matched for total protein or leucine content to whey, worm protein matched for total protein, or saline. Blood was drawn 30, 60, 105 and 150 min after gavage and muscle samples were harvested 60 min and 150 min after gavage to measure key components of the mTORC1 pathway. Ingestion of plant-based proteins induced a lower rise in blood leucine compared to whey, which coincided with a dampened mTORC1 activation, both acutely and 150 min after administration. Matching total leucine content to whey did not rescue the reduced rise in plasma amino acids, nor the lower increase in mTORC1 compared to whey. Insect protein elicits a similar activation of downstream mTORC1 kinases as plant-based proteins, despite lower postprandial aminoacidemia. The mTORC1 response following ingestion of high-quality plant-based and insect proteins is dampened compared to whey in mouse skeletal muscle.


Asunto(s)
Ingestión de Alimentos , Proteínas de Insectos/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Músculo Esquelético/metabolismo , Proteínas de Plantas/farmacología , Proteína de Suero de Leche/farmacología , Aminoácidos/administración & dosificación , Aminoácidos/sangre , Animales , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
9.
J Cachexia Sarcopenia Muscle ; 11(4): 1121-1140, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32596975

RESUMEN

BACKGROUND: Frailty is a geriatric syndrome characterized by increased susceptibility to adverse health outcomes. One major determinant thereof is the gradual weakening of the musculoskeletal system and the associated osteosarcopenia. To improve our understanding of the underlying pathophysiology and, more importantly, to test potential interventions aimed at counteracting frailty, suitable animal models are needed. METHODS: To evaluate the relevance of prematurely aged PolgA(D257A/D257A) mice as a model for frailty and osteosarcopenia, we quantified the clinical mouse frailty index in PolgA(D257A/D257A) and wild-type littermates (PolgA(+/+) , WT) with age and concertedly assessed the quantity and quality of bone and muscle tissue. Lastly, the anabolic responsiveness of skeletal muscle, muscle progenitors, and bone was assessed. RESULTS: PolgA(D257A/D257A) accumulated health deficits at a higher rate compared with WT, resulting in a higher frailty index at 40 and 46 weeks of age (+166%, +278%, P < 0.0001), respectively, with no differences between genotypes at 34 weeks. Concomitantly, PolgA(D257A/D257A) displayed progressive musculoskeletal deterioration such as reduced bone and muscle mass as well as impaired functionality thereof. In addition to lower muscle weights (-14%, P < 0.05, -23%, P < 0.0001) and fibre area (-20%, P < 0.05, -22%, P < 0.0001) at 40 and 46 weeks, respectively, PolgA(D257A/D257A) showed impairments in grip strength and concentric muscle forces (P < 0.05). PolgA(D257A/D257A) mutation altered the acute response to various anabolic stimuli in skeletal muscle and muscle progenitors. While PolgA(D257A/D257A) muscles were hypersensitive to eccentric contractions as well as leucine administration, shown by larger downstream signalling response of the mechanistic target of rapamycin complex 1, myogenic progenitors cultured in vitro showed severe anabolic resistance to leucine and robust impairments in cell proliferation. Longitudinal micro-computed tomography analysis of the sixth caudal vertebrae showed that PolgA(D257A/D257A) had lower bone morphometric parameters (e.g. bone volume fraction, trabecular, and cortical thickness, P < 0.05) as well as reduced remodelling activities (e.g. bone formation and resorption rate, P < 0.05) compared with WT. When subjected to 4 weeks of cyclic loading, young but not aged PolgA(D257A/D257A) caudal vertebrae showed load-induced bone adaptation, suggesting reduced mechanosensitivity with age. CONCLUSIONS: PolgA(D257A/D257A) mutation leads to hallmarks of age-related frailty and osteosarcopenia and provides a powerful model to better understand the relationship between frailty and the aging musculoskeletal system.


Asunto(s)
ADN Polimerasa gamma/metabolismo , Sarcopenia/genética , Envejecimiento Prematuro , Animales , Modelos Animales de Enfermedad , Femenino , Fragilidad , Humanos , Ratones , Sarcopenia/patología
10.
Skelet Muscle ; 10(1): 21, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32646489

RESUMEN

BACKGROUND: Satellite cells (SCs) are required for muscle repair following injury and are involved in muscle remodeling upon muscular contractions. Exercise stimulates SC accumulation and myonuclear accretion. To what extent exercise training at different mechanical loads drive SC contribution to myonuclei however is unknown. RESULTS: By performing SC fate tracing experiments, we show that 8 weeks of voluntary wheel running increased SC contribution to myofibers in mouse plantar flexor muscles in a load-dependent, but fiber type-independent manner. Increased SC fusion however was not exclusively linked to muscle hypertrophy as wheel running without external load substantially increased SC fusion in the absence of fiber hypertrophy. Due to nuclear propagation, nuclear fluorescent fate tracing mouse models were inadequate to quantify SC contribution to myonuclei. Ultimately, by performing fate tracing at the DNA level, we show that SC contribution mirrors myonuclear accretion during exercise. CONCLUSIONS: Collectively, mechanical load during exercise independently promotes SC contribution to existing myofibers. Also, due to propagation of nuclear fluorescent reporter proteins, our data warrant caution for the use of existing reporter mouse models for the quantitative evaluation of satellite cell contribution to myonuclei.


Asunto(s)
Fusión Celular , Fibras Musculares Esqueléticas/citología , Carrera , Células Satélite del Músculo Esquelético/citología , Animales , Núcleo Celular/fisiología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/fisiología , Células Satélite del Músculo Esquelético/fisiología
11.
Nat Commun ; 11(1): 174, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31924757

RESUMEN

mTORC1 is an important regulator of muscle mass but how it is modulated by oxygen and nutrients is not completely understood. We show that loss of the prolyl hydroxylase domain isoform 1 oxygen sensor in mice (PHD1KO) reduces muscle mass. PHD1KO muscles show impaired mTORC1 activation in response to leucine whereas mTORC1 activation by growth factors or eccentric contractions was preserved. The ability of PHD1 to promote mTORC1 activity is independent of its hydroxylation activity but is caused by decreased protein content of the leucyl tRNA synthetase (LRS) leucine sensor. Mechanistically, PHD1 interacts with and stabilizes LRS. This interaction is promoted during oxygen and amino acid depletion and protects LRS from degradation. Finally, elderly subjects have lower PHD1 levels and LRS activity in muscle from aged versus young human subjects. In conclusion, PHD1 ensures an optimal mTORC1 response to leucine after episodes of metabolic scarcity.


Asunto(s)
Leucina-ARNt Ligasa/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Músculos/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Adulto , Anciano , Envejecimiento/metabolismo , Aminoácidos/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Hidroxilación , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Leucina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos , Músculos/patología , Oxígeno/metabolismo , Procolágeno-Prolina Dioxigenasa/genética , Transducción de Señal
12.
Cell Metab ; 31(6): 1136-1153.e7, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32492393

RESUMEN

Endothelial cell (EC)-derived signals contribute to organ regeneration, but angiocrine metabolic communication is not described. We found that EC-specific loss of the glycolytic regulator pfkfb3 reduced ischemic hindlimb revascularization and impaired muscle regeneration. This was caused by the reduced ability of macrophages to adopt a proangiogenic and proregenerative M2-like phenotype. Mechanistically, loss of pfkfb3 reduced lactate secretion by ECs and lowered lactate levels in the ischemic muscle. Addition of lactate to pfkfb3-deficient ECs restored M2-like polarization in an MCT1-dependent fashion. Lactate shuttling by ECs enabled macrophages to promote proliferation and fusion of muscle progenitors. Moreover, VEGF production by lactate-polarized macrophages was increased, resulting in a positive feedback loop that further stimulated angiogenesis. Finally, increasing lactate levels during ischemia rescued macrophage polarization and improved muscle reperfusion and regeneration, whereas macrophage-specific mct1 deletion prevented M2-like polarization. In summary, ECs exploit glycolysis for angiocrine lactate shuttling to steer muscle regeneration from ischemia.


Asunto(s)
Células Endoteliales/química , Isquemia/metabolismo , Lactatos/farmacología , Macrófagos/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Animales , Células Cultivadas , Isquemia/patología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/metabolismo
13.
Front Physiol ; 10: 1271, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31636571

RESUMEN

Long-term voluntary resistance running has been shown to be a valid model to induce muscle growth in rodents. Moreover, the mammalian target of rapamycin complex 1 (mTORC1) is a key signaling complex regulating exercise/nutrient-induced alterations in muscle protein synthesis. How acute resistance running affects mTORC1 signaling in muscle and if resistance applied to the wheel can modulate mTORC1 activation has not yet been fully elucidated. Here, we show that both acute resistance running and acute free running activated mTORC1 signaling in the m. gastrocnemius, m. soleus, and m. plantaris, but not in m. tibialis anterior of mice when compared to sedentary controls. Furthermore, only the low threshold oxidative part in the m. gastrocnemius showed increased mTORC1 signaling upon running and acute heavy-load resistance running evoked higher downstream mTORC1 signaling in both m. soleus and m. plantaris than free running without resistance, pointing toward mechanical load as an important independent regulator of mTORC1. Collectively, in this study, we show that voluntary resistance running is an easy-to-use, time-efficient and low stress model to study acute alterations in mTORC1 signaling upon high-load muscular contractions in mice.

14.
Front Physiol ; 8: 347, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28620311

RESUMEN

In the setting of "living high," it is unclear whether high-intensity interval training (HIIT) should be performed "low" or "high" to stimulate muscular and performance adaptations. Therefore, 10 physically active males participated in a 5-week "live high-train low or high" program (TR), whilst eight subjects were not engaged in any altitude or training intervention (CON). Five days per week (~15.5 h per day), TR was exposed to normobaric hypoxia simulating progressively increasing altitude of ~2,000-3,250 m. Three times per week, TR performed HIIT, administered as unilateral knee-extension training, with one leg in normobaric hypoxia (~4,300 m; TRHYP) and with the other leg in normoxia (TRNOR). "Living high" elicited a consistent elevation in serum erythropoietin concentrations which adequately predicted the increase in hemoglobin mass (r = 0.78, P < 0.05; TR: +2.6%, P < 0.05; CON: -0.7%, P > 0.05). Muscle oxygenation during training was lower in TRHYP vs. TRNOR (P < 0.05). Muscle homogenate buffering capacity and pH-regulating protein abundance were similar between pretest and posttest. Oscillations in muscle blood volume during repeated sprints, as estimated by oscillations in NIRS-derived tHb, increased from pretest to posttest in TRHYP (~80%, P < 0.01) but not in TRNOR (~50%, P = 0.08). Muscle capillarity (~15%) as well as repeated-sprint ability (~8%) and 3-min maximal performance (~10-15%) increased similarly in both legs (P < 0.05). Maximal isometric strength increased in TRHYP (~8%, P < 0.05) but not in TRNOR (~4%, P > 0.05). In conclusion, muscular and performance adaptations were largely similar following normoxic vs. hypoxic HIIT. However, hypoxic HIIT stimulated adaptations in isometric strength and muscle perfusion during intermittent sprinting.

15.
Diabetes ; 66(6): 1548-1559, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28389470

RESUMEN

Exercise bypasses insulin resistance to increase glucose uptake in skeletal muscle and therefore represents an important alternative to stimulate glucose uptake in insulin-resistant muscle. Both Rac1 and AMPK have been shown to partly regulate contraction-stimulated muscle glucose uptake, but whether those two signaling pathways jointly account for the entire signal to glucose transport is unknown. We therefore studied the ability of contraction and exercise to stimulate glucose transport in isolated muscles with AMPK loss of function combined with either pharmacological inhibition or genetic deletion of Rac1.Muscle-specific knockout (mKO) of Rac1, a kinase-dead α2 AMPK (α2KD), and double knockout (KO) of ß1 and ß2 AMPK subunits (ß1ß2 KO) each partially decreased contraction-stimulated glucose transport in mouse soleus and extensor digitorum longus (EDL) muscle. Interestingly, when pharmacological Rac1 inhibition was combined with either AMPK ß1ß2 KO or α2KD, contraction-stimulated glucose transport was almost completely inhibited. Importantly, α2KD+Rac1 mKO double-transgenic mice also displayed severely impaired contraction-stimulated glucose transport, whereas exercise-stimulated glucose uptake in vivo was only partially reduced by Rac1 mKO with no additive effect of α2KD. It is concluded that Rac1 and AMPK together account for almost the entire ex vivo contraction response in muscle glucose transport, whereas only Rac1, but not α2 AMPK, regulates muscle glucose uptake during submaximal exercise in vivo.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Glucosa/metabolismo , Contracción Muscular , Músculo Esquelético/metabolismo , Neuropéptidos/genética , Condicionamiento Físico Animal , Proteína de Unión al GTP rac1/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Composición Corporal , Desoxiglucosa/metabolismo , Estimulación Eléctrica , Tolerancia al Ejercicio , Técnicas de Silenciamiento del Gen , Prueba de Tolerancia a la Glucosa , Glucosa-6-Fosfato/metabolismo , Glucógeno/metabolismo , Immunoblotting , Imagen por Resonancia Magnética , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/fisiología , Neuropéptidos/metabolismo , Tritio , Proteína de Unión al GTP rac1/metabolismo
16.
Front Physiol ; 7: 676, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28149279

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) is a master regulator of myocellular adaptation to exercise and hypoxia. However, the role of genetic factors in regulation of HIF-1 responses to exercise and hypoxia is unknown. We hypothesized that hypoxia at rest and during exercise stimulates the HIF-1 pathway and its downstream targets in energy metabolism regulation in a genotype-dependent manner. Eleven monozygotic twin (MZ) pairs performed an experimental trial in both normoxia and hypoxia (FiO2 10.7%). Biopsies were taken from m. vastus lateralis before and after a 20-min submaximal cycling bout @~30% of sea-level VO2max. Key-markers of the HIF-1 pathway and glycolytic and oxidative metabolism were analyzed using real-time PCR and Western Blot. Hypoxia increased HIF-1α protein expression by ~120% at rest vs. +150% during exercise (p < 0.05). Furthermore, hypoxia but not exercise increased muscle mRNA content of HIF-1α (+50%), PHD2 (+45%), pVHL (+45%; p < 0.05), PDK4 (+1200%), as well as PFK-M (+20%) and PPAR-γ1 (+60%; p < 0.05). Neither hypoxia nor exercise altered PHD1, LDH-A, PDH-A1, COX-4, and CS mRNA expressions. The hypoxic, but not normoxic exercise-induced increment of muscle HIF-1α mRNA content was about 10-fold more similar within MZ twins than between the twins (p < 0.05). Furthermore, in resting muscle the hypoxia-induced increments of muscle HIF-1α protein content, and HIF-1α and PDK4 mRNA content were about 3-4-fold more homogeneous within than between the twins pairs (p < 0.05). The present observations in monozygotic twins for the first time clearly indicate that the HIF-1α protein as well as mRNA responses to submaximal exercise in acute hypoxia are at least partly regulated by genetic factors.

17.
Endocrinology ; 157(8): 3009-15, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27285860

RESUMEN

Exercise has a potent insulin-sensitivity enhancing effect on skeletal muscle, but the intracellular mechanisms that mediate this effect are not well understood. In muscle, Ras-related C3 botulinum toxin substrate 1 (Rac1) regulates both insulin- and contraction-stimulated glucose transport and is dysregulated in insulin resistant muscle. However, whether Rac1 is involved in mediating enhanced insulin sensitivity after an acute bout of exercise is unresolved. To address this question, we investigated after exercise whole-body (insulin tolerance test) as well as muscle (insulin-stimulated 2-deoxyglucose transport in isolated soleus muscle) insulin sensitivity in inducible muscle-specific Rac1 knockout (mKO) and wild-type (WT) littermate mice. Previous exercise enhanced whole-body insulin sensitivity by 40% in WT mice and rescued the insulin intolerance in Rac1 mKO mice by improving whole-body insulin sensitivity by 230%. In agreement, previous exercise significantly improved insulin sensitivity by 20% in WT and by 40% in Rac1 mKO soleus muscles. These findings suggest that muscle Rac1 is dispensable for the insulin sensitizing effect of exercise. Moreover, insulin resistance in Rac1 mKO mice can be completely normalized by previous exercise explaining why insulin resistant patients can increase insulin action with exercise despite dysfunctional Rac1 activity in muscle.


Asunto(s)
Insulina/farmacología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Neuropéptidos/fisiología , Condicionamiento Físico Animal/fisiología , Proteína de Unión al GTP rac1/fisiología , Animales , Transporte Biológico/genética , Femenino , Glucosa/metabolismo , Resistencia a la Insulina/genética , Ratones , Ratones Noqueados , Contracción Muscular/efectos de los fármacos , Contracción Muscular/genética , Neuropéptidos/genética , Proteína de Unión al GTP rac1/genética
18.
Hypoxia (Auckl) ; 4: 1-14, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27800505

RESUMEN

Chronic hypoxia leads to muscle atrophy. The molecular mechanisms responsible for this phenomenon are not well defined in vivo. We sought to determine how chronic hypoxia regulates molecular markers of protein synthesis and degradation in human skeletal muscle and whether these regulations were related to the regulation of the hypoxia-inducible factor (HIF) pathway. Eight young male subjects lived in a normobaric hypoxic hotel (FiO2 14.1%, 3,200 m) for 15 days in well-controlled conditions for nutrition and physical activity. Skeletal muscle biopsies were obtained in the musculus vastus lateralis before (PRE) and immediately after (POST) hypoxic exposure. Intramuscular hypoxia-inducible factor-1 alpha (HIF-1α) protein expression decreased (-49%, P=0.03), whereas hypoxia-inducible factor-2 alpha (HIF-2α) remained unaffected from PRE to POST hypoxic exposure. Also, downstream HIF-1α target genes VEGF-A (-66%, P=0.006) and BNIP3 (-24%, P=0.002) were downregulated, and a tendency was measured for neural precursor cell expressed, developmentally Nedd4 (-47%, P=0.07), suggesting lowered HIF-1α transcriptional activity after 15 days of exposure to environmental hypoxia. No difference was found on microtubule-associated protein 1 light chain 3 type II/I (LC3b-II/I) ratio, and P62 protein expression tended to increase (+45%, P=0.07) compared to PRE exposure levels, suggesting that autophagy was not modulated after chronic hypoxia. The mammalian target of rapamycin complex 1 pathway was not altered as Akt, mammalian target of rapamycin, S6 kinase 1, and 4E-binding protein 1 phosphorylation did not change between PRE and POST. Finally, myofiber cross-sectional area was unchanged between PRE and POST. In summary, our data indicate that moderate chronic hypoxia differentially regulates HIF-1α and HIF-2α, marginally affects markers of protein degradation, and does not modify markers of protein synthesis or myofiber cross-sectional area in human skeletal muscle.

19.
Physiol Rep ; 2(5): e00286, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24819751

RESUMEN

Needle biopsies are being extensively used in clinical trials addressing muscular adaptation to exercise and diet. Still, the potential artifacts due to biopsy sampling are often overlooked. Healthy volunteers (n = 9) underwent two biopsies through a single skin incision in a pretest. Two days later (posttest) another biopsy was taken 3 cm proximally and 3 cm distally to the pretest incision. Muscle oxygenation status (tissue oxygenation index [TOI]) was measured by near-infrared spectroscopy. Biopsy samples were analyzed for 40 key markers (mRNA and protein contents) of myocellular O2 sensing, inflammation, cell proliferation, mitochondrial biogenesis, protein synthesis and breakdown, oxidative stress, and energy metabolism. In the pretest, all measurements were identical between proximal and distal biopsies. However, compared with the pretest, TOI in the posttest was reduced in the proximal (-10%, P < 0.05), but not in the distal area. Conversely, most inflammatory markers were upregulated at the distal (100-500%, P < 0.05), but not at the proximal site. Overall, 29 of the 40 markers measured, equally distributed over all pathways studied, were either up- or downregulated by 50-500% (P < 0.05). In addition, 19 markers yielded conflicting results between the proximal and distal measurements (P < 0.05). This study clearly documents that prior muscle biopsies can cause major disturbances in myocellular signaling pathways in needle biopsies specimens sampled 48 h later. In addition, different biopsy sites within identical experimental conditions yielded conflicting results.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA