Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Publication year range
1.
J Neuroeng Rehabil ; 20(1): 84, 2023 06 29.
Article in English | MEDLINE | ID: mdl-37386493

ABSTRACT

BACKGROUND: Skeletal muscle is negatively impacted by conditions such as spaceflight or prolonged bed rest, resulting in a dramatic decline in muscle mass, maximum contractile force, and muscular endurance. Electrical stimulation (ES) is an essential tool in neurophysiotherapy and an effective means of preventing skeletal muscle atrophy and dysfunction. Historically, ES treatment protocols have used either low or high frequency electrical stimulation (LFES/HFES). However, our study tests the use of a combination of different frequencies in a single electrical stimulation intervention in order to determine a more effective protocol for improving both skeletal muscle strength and endurance. METHODS: An adult male SD rat model of muscle atrophy was established through 4 weeks of tail suspension (TS). To investigate the effects of different frequency combinations, the experimental animals were treated with low (20 Hz) or high (100 Hz) frequency before TS for 6 weeks, and during TS for 4weeks. The maximum contraction force and fatigue resistance of skeletal muscle were then assessed before the animals were sacrificed. The muscle mass, fiber cross-sectional area (CSA), fiber type and related protein expression were examined and analyzed to gain insights into the mechanisms by which the ES intervention protocol used in this study regulates muscle strength and endurance. RESULTS: After 4 weeks of unloading, the soleus muscle mass and fiber CSA decreased by 39% and 58% respectively, while the number of glycolytic muscle fibers increased by 21%. The gastrocnemius muscle fibers showed a 51% decrease in CSA, with a 44% decrease in single contractility and a 39% decrease in fatigue resistance. The number of glycolytic muscle fibers in the gastrocnemius also increased by 29%. However, the application of HFES either prior to or during unloading showed an improvement in muscle mass, fiber CSA, and oxidative muscle fibers. In the pre-unloading group, the soleus muscle mass increased by 62%, while the number of oxidative muscle fibers increased by 18%. In the during unloading group, the soleus muscle mass increased by 29% and the number of oxidative muscle fibers increased by 15%. In the gastrocnemius, the pre-unloading group showed a 38% increase in single contractile force and a 19% increase in fatigue resistance, while in the during unloading group, a 21% increase in single contractile force and a 29% increase in fatigue resistance was observed, along with a 37% and 26% increase in the number of oxidative muscle fibers, respectively. The combination of HFES before unloading and LFES during unloading resulted in a significant elevation of the soleus mass by 49% and CSA by 90%, with a 40% increase in the number of oxidative muscle fibers in the gastrocnemius. This combination also resulted in a 66% increase in single contractility and a 38% increase in fatigue resistance. CONCLUSION: Our results indicated that using HFES before unloading can reduce the harmful effects of muscle unloading on the soleus and gastrocnemius muscles. Furthermore, we found that combining HFES before unloading with LFES during unloading was more effective in preventing muscle atrophy in the soleus and preserving the contractile function of the gastrocnemius muscle.


Subject(s)
Muscular Disorders, Atrophic , Male , Animals , Rats , Rats, Sprague-Dawley , Muscular Disorders, Atrophic/prevention & control , Muscle Strength , Muscular Atrophy/prevention & control , Muscle, Skeletal , Electric Stimulation
2.
Am J Physiol Endocrinol Metab ; 312(5): E394-E406, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28270443

ABSTRACT

Peroxisome proliferator-activated receptor-γ coactivator (PGC)-1α and -1ß serve as master transcriptional regulators of muscle mitochondrial functional capacity and are capable of enhancing muscle endurance when overexpressed in mice. We sought to determine whether muscle-specific transgenic overexpression of PGC-1ß affects the detraining response following endurance training. First, we established and validated a mouse exercise-training-detraining protocol. Second, using multiple physiological and gene expression end points, we found that PGC-1ß overexpression in skeletal muscle of sedentary mice fully recapitulated the training response. Lastly, PGC-1ß overexpression during the detraining period resulted in partial prevention of the detraining response. Specifically, an increase in the plateau at which O2 uptake (V̇o2) did not change from baseline with increasing treadmill speed [peak V̇o2 (ΔV̇o2max)] was maintained in trained mice with PGC-1ß overexpression in muscle 6 wk after cessation of training. However, other detraining responses, including changes in running performance and in situ half relaxation time (a measure of contractility), were not affected by PGC-1ß overexpression. We conclude that while activation of muscle PGC-1ß is sufficient to drive the complete endurance phenotype in sedentary mice, it only partially prevents the detraining response following exercise training, suggesting that the process of endurance detraining involves mechanisms beyond the reversal of muscle autonomous mechanisms involved in endurance fitness. In addition, the protocol described here should be useful for assessing early-stage proof-of-concept interventions in preclinical models of muscle disuse atrophy.


Subject(s)
Muscle, Skeletal/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal/methods , Physical Endurance/physiology , Physical Fitness/physiology , Running/physiology , Animals , Male , Mice , Mice, Transgenic , Muscular Disorders, Atrophic/physiopathology , Muscular Disorders, Atrophic/prevention & control , Phenotype
3.
FASEB J ; 25(12): 4312-25, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21885651

ABSTRACT

Protein calcium sensors of the Homer family have been proposed to modulate the activity of various ion channels and nuclear factor of activated T cells (NFAT), the transcription factor modulating skeletal muscle differentiation. We monitored Homer expression and subcellular localization in human skeletal muscle biopsies following 60 d of bedrest [Second Berlin Bedrest Study (BBR2-2)]. Soleus (SOL) and vastus lateralis (VL) biopsies were taken at start (pre) and at end (end) of bedrest from healthy male volunteers of a control group without exercise (CTR; n=9), a resistive-only exercise group (RE; n=7), and a combined resistive/vibration exercise group (RVE; n=7). Confocal analysis showed Homer immunoreactivity at the postsynaptic microdomain of the neuromuscular junction (NMJ) at bedrest start. After bedrest, Homer immunoreactivity decreased (CTR), remained unchanged (RE), or increased (RVE) at the NMJ. Homer2 mRNA and protein were differently regulated in a muscle-specific way. Activated NFATc1 translocates from cytoplasm to nucleus; increased amounts of NFATc1-immunopositive slow-type myonuclei were found in RVE myofibers of both muscles. Pulldown assays identified NFATc1 and Homer as molecular partners in skeletal muscle. A direct motor nerve control of Homer2 was confirmed in rat NMJs by in vivo denervation. Homer2 is localized at the NMJ and is part of the calcineurin-NFATc1 signaling pathway. RVE has additional benefit over RE as countermeasure preventing disuse-induced neuromuscular maladaptation during bedrest.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Adaptation, Physiological , Animals , Base Sequence , Bed Rest/adverse effects , DNA Primers/genetics , Exercise/physiology , Gene Expression Regulation , Homer Scaffolding Proteins , Humans , Male , Models, Biological , Muscle Denervation , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/metabolism , Muscular Disorders, Atrophic/prevention & control , NFATC Transcription Factors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Resistance Training , Signal Transduction , Vibration/therapeutic use
4.
Crit Rev Food Sci Nutr ; 51(9): 828-34, 2011.
Article in English | MEDLINE | ID: mdl-21888533

ABSTRACT

Recently there has been much interest in the use of dietary amino acids supplements to decrease the losses in muscle mass and strength observed after space flight or during aging using bed rest analogs. This interest persists even if the results have been mixed. Of the six published amino acid supplementation studies, three showed benefit, three did not. A recent study re-evaluating protein requirements in humans suggests that the official RDA is 41% underestimated. Interestingly, the three studies that showed benefits fed their test subjects a baseline protein level around the old RDA for protein. The three that did not show benefit gave the subjects a baseline protein intake higher than the new RDA. We suggest that the positive effects observed on protein metabolism might just reflect the benefits of adequate protein intake achieved during bed rest rather than a protective effect against bed-rest induced disuse. In conclusion, the efficiency of amino acid countermeasures for preventing the loss in protein mass during space flight or bed rest needs to be seriously questioned. These results extend to clinical situations such as serious illness and progress of aging in which physical inactivity is a significant component of the loss in muscle function.


Subject(s)
Amino Acids/administration & dosage , Dietary Proteins/administration & dosage , Dietary Supplements , Muscle Strength/drug effects , Muscular Disorders, Atrophic/prevention & control , Bed Rest , Diet , Humans , Space Flight
5.
Med Sci Sports Exerc ; 53(5): 1033-1040, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33105390

ABSTRACT

PURPOSE: This study aimed to investigate the effects of blood flow restriction (BFR) combined with electrical muscle stimulation (EMS) on skeletal muscle mass and strength during a period of limb disuse. METHODS: Thirty healthy participants (22 ± 3 yr; 23 ± 3 kg·m-2) were randomly assigned to control (CON; n = 10), BFR alone (BFR; n = 10), or BFR combined with EMS (BFR + EMS; n = 10). All participants completed unloading of a single leg for 14 d, with no treatment (CON), or while treated with either BFR or BFR + EMS (twice daily, 5 d·wk-1). BFR treatment involved arterial three cycles of 5-min occlusion using suprasystolic pressure, each separated by 5 min of reperfusion. EMS (6 s on, 15 s off; 200 µs; 60 Hz; 15% maximal voluntary contraction [MVC]) was applied continuously throughout the three BFR cycles. Quadriceps muscle mass (whole-thigh lean mass via dual-energy x-ray absorptiometry and vastus lateralis [VL] muscle thickness via ultrasound) and strength (via knee extension MVC) were assessed before and after the 14-d unloading period. RESULTS: After limb unloading, whole-thigh lean mass decreased in the control group (-4% ± 1%, P < 0.001) and BFR group (-3% ± 2%, P = 0.001), but not in the BFR + EMS group (-0.3% ± 3%, P = 0.8). VL muscle thickness decreased in the control group (-4% ± 4%, P = 0.005) and was trending toward a decrease in the BFR group (-8% ± 11%, P = 0.07) and increase in the BFR + EMS group (+5% ± 10%, P = 0.07). Knee extension MVC decreased over time (P < 0.005) in the control group (-18% ± 15%), BFR group (-10% ± 13%), and BFR + EMS group (-18% ± 15%), with no difference between groups (P > 0.5). CONCLUSION: Unlike BFR performed in isolation, BFR + EMS represents an effective interventional strategy to attenuate the loss of muscle mass during limb disuse, but it does not demonstrate preservation of strength.


Subject(s)
Electric Stimulation/methods , Muscle Strength , Muscular Disorders, Atrophic/prevention & control , Quadriceps Muscle/blood supply , Quadriceps Muscle/pathology , Regional Blood Flow , Absorptiometry, Photon , Female , Humans , Immobilization/adverse effects , Male , Muscular Disorders, Atrophic/diagnostic imaging , Quadriceps Muscle/diagnostic imaging , Quadriceps Muscle/physiopathology , Thigh , Tourniquets , Ultrasonography , Young Adult
6.
Clin Transl Sci ; 14(4): 1512-1523, 2021 07.
Article in English | MEDLINE | ID: mdl-33742769

ABSTRACT

Mechanical ventilation (MV) is a life-saving instrument used to provide ventilatory support for critically ill patients and patients undergoing surgery. Unfortunately, an unintended consequence of prolonged MV is the development of inspiratory weakness due to both diaphragmatic atrophy and contractile dysfunction; this syndrome is labeled ventilator-induced diaphragm dysfunction (VIDD). VIDD is clinically important because diaphragmatic weakness is an important contributor to problems in weaning patients from MV. Investigations into the pathogenesis of VIDD reveal that oxidative stress is essential for the rapid development of VIDD as redox disturbances in diaphragm fibers promote accelerated proteolysis. Currently, no standard treatment exists to prevent VIDD and, therefore, developing a strategy to avert VIDD is vital. Guided by evidence indicating that activation of the classical axis of the renin-angiotensin system (RAS) in diaphragm fibers promotes oxidative stress and VIDD, we hypothesized that activation of the nonclassical RAS signaling pathway via angiotensin 1-7 (Ang1-7) will protect against VIDD. Using an established animal model of prolonged MV, our results disclose that infusion of Ang1-7 protects the diaphragm against MV-induced contractile dysfunction and fiber atrophy in both fast and slow muscle fibers. Further, Ang1-7 shielded diaphragm fibers against MV-induced mitochondrial damage, oxidative stress, and protease activation. Collectively, these results reveal that treatment with Ang1-7 protects against VIDD, in part, due to diminishing oxidative stress and protease activation. These important findings provide robust evidence that Ang1-7 has the therapeutic potential to protect against VIDD by preventing MV-induced contractile dysfunction and atrophy of both slow and fast muscle fibers.


Subject(s)
Angiotensin I/administration & dosage , Diaphragm/drug effects , Muscle Weakness/prevention & control , Muscular Disorders, Atrophic/prevention & control , Peptide Fragments/administration & dosage , Respiration, Artificial/adverse effects , Animals , Diaphragm/physiopathology , Disease Models, Animal , Female , Humans , Infusions, Intravenous , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Weakness/etiology , Muscle Weakness/physiopathology , Muscular Disorders, Atrophic/etiology , Muscular Disorders, Atrophic/physiopathology , Oxidative Stress/drug effects , Rats
7.
J Musculoskelet Neuronal Interact ; 10(1): 12-24, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20190376

ABSTRACT

To identify mechanotransductive signals for combating musculoskeletal deterioration, it is essential to determine the components and mechanisms critical to the anabolic processes of musculoskeletal tissues. It is hypothesized that the interaction between bone and muscle may depend on fluid exchange in these tissues by mechanical loading. It has been shown that intramedullary pressure (ImP) and low-level bone strain induced by muscle stimulation (MS) has the potential to mitigate bone loss induced by disuse osteopenia. Optimized MS signals, i.e., low-intensity and high frequency, may be critical in maintaining bone mass and mitigating muscle atrophy. The objectives for this review are to discuss the potential for MS to induce ImP and strains on bone, to regulate bone adaptation, and to identify optimized stimulation frequency in the loading regimen. The potential for MS to regulate blood and fluid flow will also be discussed. The results suggest that oscillatory MS regulates fluid dynamics with minimal mechanical strain in bone. The response was shown to be dependent on loading frequency, serving as a critical mediator in mitigating bone loss. A specific regimen of dynamic MS may be optimized in vivo to attenuate disuse osteopenia and serve as a biomechanical intervention in the clinical setting.


Subject(s)
Bone Development/physiology , Bone Diseases, Metabolic/therapy , Muscular Disorders, Atrophic/therapy , Musculoskeletal Development/physiology , Osteogenesis/physiology , Adaptation, Physiological/physiology , Animals , Bone Diseases, Metabolic/physiopathology , Electric Stimulation Therapy/methods , Humans , Mechanotransduction, Cellular/physiology , Muscle Contraction/physiology , Muscular Disorders, Atrophic/physiopathology , Muscular Disorders, Atrophic/prevention & control , Rats , Stress, Mechanical
8.
Nutrients ; 12(5)2020 May 25.
Article in English | MEDLINE | ID: mdl-32466126

ABSTRACT

Preserving skeletal muscle mass and functional capacity is essential for healthy ageing. Transient periods of disuse and/or inactivity in combination with sub-optimal dietary intake have been shown to accelerate the age-related loss of muscle mass and strength, predisposing to disability and metabolic disease. Mechanisms underlying disuse and/or inactivity-related muscle deterioration in the older adults, whilst multifaceted, ultimately manifest in an imbalance between rates of muscle protein synthesis and breakdown, resulting in net muscle loss. To date, the most potent intervention to mitigate disuse-induced muscle deterioration is mechanical loading in the form of resistance exercise. However, the feasibility of older individuals performing resistance exercise during disuse and inactivity has been questioned, particularly as illness and injury may affect adherence and safety, as well as accessibility to appropriate equipment and physical therapists. Therefore, optimising nutritional intake during disuse events, through the introduction of protein-rich whole-foods, isolated proteins and nutrient compounds with purported pro-anabolic and anti-catabolic properties could offset impairments in muscle protein turnover and, ultimately, the degree of muscle atrophy and recovery upon re-ambulation. The current review therefore aims to provide an overview of nutritional countermeasures to disuse atrophy and anabolic resistance in older individuals.


Subject(s)
Aging , Muscle Proteins/metabolism , Muscular Atrophy/diet therapy , Muscular Atrophy/prevention & control , Muscular Disorders, Atrophic/diet therapy , Muscular Disorders, Atrophic/prevention & control , Aged , Creatine/administration & dosage , Diet, High-Protein , Dietary Proteins/administration & dosage , Dietary Supplements , Exercise , Fatty Acids, Omega-3/administration & dosage , Humans , Male , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Muscular Disorders, Atrophic/complications , Nutritional Status , Valerates/administration & dosage
9.
Histochem Cell Biol ; 132(4): 383-94, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19644701

ABSTRACT

We monitored changes in SERCA isoform specific expression and S-nitrosylation in myofibers of lower limb soleus (SOL) and vastus lateralis (VL) muscle biopsies before and after 60 days of voluntary long term bed rest (BR) without (BR-CTRL group, n = 8) and with exercise countermeasure (BR-EX group, n = 8). Before BR, a typical myofiber type-specific distribution of fast and slow SERCA1/2a isoforms was seen. After BR, a subpopulation (approx. 15%) of slow myofibers in BR-CTRL additionally expressed the fast SERCA1a isoform which was not seen in BR-EX. After BR, SERCA1a S-nitrosylation patterns analyzed by the biotin-switch assay decreased in disused SOL only but increased in both muscles following exercise. Differential SERCA1a S-nitrosylation and SERCA1a/2a co-expression in subsets of slow myofibers should be considered as signs of an altered cytosolic Ca(2+) homeostasis following chronic muscle disuse. Exercise preserved myofiber type-specific SERCA1a expression and S-nitrosylation in VL and SOL in a different way, suggesting muscle-specific responses to the countermeasure protocol applied during bed rest.


Subject(s)
Muscle Fibers, Slow-Twitch/enzymology , Muscular Disorders, Atrophic/enzymology , Myofibrils/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Bed Rest/adverse effects , Exercise/physiology , Female , Humans , Lower Extremity , Muscular Disorders, Atrophic/pathology , Muscular Disorders, Atrophic/prevention & control , Protein Isoforms/metabolism , Quadriceps Muscle/enzymology
10.
J Appl Physiol (1985) ; 107(1): 34-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19390002

ABSTRACT

We sought to determine the effects of longitudinal loading (artificial gravity) on skeletal muscle protein kinetics in 15 healthy young males after 21 days of 6 degrees head-down tilt bed rest [experimental treatment (Exp) group: n = 8, 31 +/- 1 yr; control (Con) group; n = 7, 28 +/- 1 yr, means +/- SE]. On days 1 and 21 of bed rest, postabsorptive venous blood samples and muscle biopsies (vastus lateralis and soleus) were obtained during a 1-h pulse bolus infusion protocol (0 min, l-[ring-(13)C(6)]phenylalanine, 35 mumol/kg; 30 min, l-[ring-(15)N]phenylalanine, 35 mumol/kg). Outcome measures included mixed muscle fractional synthesis (FSR) and breakdown rates (FBR). The Exp group experienced 1 h of longitudinal loading (2.5G at the feet) via a short-radius centrifuge during each day of bed rest. Mixed muscle FSR in the Con group was reduced by 48.5% (day 1, 0.081 +/- 0.000%/h vs. day 21, 0.042 +/- 0.000%/h; P = 0.001) in vastus lateralis after 21 days of bed rest, whereas the Exp group maintained their rate of protein synthesis. A similar but nonsignificant change in FSR was noted for the soleus muscle (Exp, -7%; Con, -22%). No changes in muscle protein breakdown were observed. In conclusion, 1 h of daily exposure to artificial gravity maintained the rate of protein synthesis of the vastus lateralis and may represent an effective adjunct countermeasure to combat the loss of muscle mass and functional during extended spaceflight.


Subject(s)
Gravity, Altered , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Muscular Atrophy/prevention & control , Muscular Disorders, Atrophic/prevention & control , Weightlessness Countermeasures , Adult , Bed Rest/adverse effects , Head-Down Tilt/adverse effects , Humans , Male , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Disorders, Atrophic/etiology , Muscular Disorders, Atrophic/metabolism , Weight-Bearing/physiology , Weightlessness/adverse effects
11.
J Appl Physiol (1985) ; 107(1): 39-46, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19286573

ABSTRACT

The goal of this project was to examine the effects of artificial gravity (AG) on skeletal muscle strength and key anabolic/catabolic markers known to regulate muscle mass. Two groups of subjects were selected for study: 1) a 21 day-bed rest (BR) group (n = 7) and 2) an AG group (n = 8), which was subjected to 21 days of 6 degrees head-down tilt bed rest plus daily 1-h exposures to AG (2.5 G at the feet). Centrifugation was produced using a short-arm centrifuge with the foot plate approximately 220 cm from the center of rotation. The torque-velocity relationships of the knee extensors and plantar flexors of the ankle were determined pre- and posttreatment. Muscle biopsy samples obtained from the vastus lateralis and soleus muscles were used for a series of gene expression analyses (mRNA abundance) of key factors implicated in the anabolic vs. catabolic state of the muscle. Post/pre torque-velocity determinations revealed greater decrements in knee extensor performance in the BR vs. AG group (P < 0.04). The plantar flexors of the AG subjects actually demonstrated a net gain in the torque-velocity relationship, whereas in the BR group, the responses declined (AG vs. BR, P < 0.001). Muscle fiber cross-sectional area decreased by approximately 20% in the BR group, whereas no losses were evident in the AG group. RT-PCR analyses of muscle biopsy specimens demonstrated that markers of growth and cytoskeletal integrity were higher in the AG group, whereas catabolic markers were elevated in the BR group. Importantly, these patterns were seen in both muscles. We conclude that paradigms of AG have the potential to maintain the functional, biochemical, and structural homeostasis of skeletal muscle in the face of chronic unloading.


Subject(s)
Gravity, Altered , Muscle, Skeletal/physiology , Muscular Atrophy/prevention & control , Muscular Disorders, Atrophic/prevention & control , Weightlessness Countermeasures , Weightlessness/adverse effects , Adult , Bed Rest/adverse effects , Exercise Test , Gene Expression , Head-Down Tilt , Humans , Knee/anatomy & histology , Knee/physiology , Leg/anatomy & histology , Leg/physiology , Male , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Strength , Muscle, Skeletal/anatomy & histology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Disorders, Atrophic/etiology , Muscular Disorders, Atrophic/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Pilot Projects , RNA, Messenger/metabolism , Range of Motion, Articular , Weightlessness Simulation
12.
PLoS One ; 14(4): e0214982, 2019.
Article in English | MEDLINE | ID: mdl-30964931

ABSTRACT

Disuse induces adaptations in skeletal muscle, which lead to muscle deterioration. Hindlimb-unloading (HU) is a well-established model to investigate cellular mechanisms responsible for disuse-induced skeletal muscle dysfunction. In myosin heavy chain (MHC) type IIB fibers HU induces a reduction in contraction speed (Vo) and a reduction in the relative myosin light chain 3f (MLC3f) protein content compared with myosin light chain 1f (MLC1f) protein. This study tested the hypothesis that increasing the relative MLC3f protein content via rAd-MLC3f vector delivery would attenuate the HU-induced decline in Vo in single MHC type IIB fibers. Fischer-344 rats were randomly assigned to one of three groups: control, HU for 7 days, and HU for 7 days plus rAd-MLC3f. The semimembranosus muscles were injected with rAd-MLC3f (3.75 x 1011-5 x 1011 ifu/ml) at four days after the initiation of HU. In single MHC type IIB fibers the relative MLC3f content decreased by 25% (12.00±0.60% to 9.06±0.66%) and Vo was reduced by 29% (3.22±0.14fl/s vs. 2.27±0.08fl/s) with HU compared to the control group. The rAd-MLC3f injection resulted in an increase in the relative MLC3f content (12.26±1.19%) and a concomitant increase in Vo (2.90±0.15fl/s) of MHC type IIB fibers. A positive relationship was observed between the percent of MLC3f content and Vo. Maximal isometric force and specific tension were reduced with HU by 49% (741.45±44.24µN to 379.09±23.77µN) and 33% (97.58±4.25kN/m2 to 65.05±2.71kN/m2), respectively compared to the control group. The rAd-MLC3f injection did not change the HU-induced decline in force or specific tension. Collectively, these results indicate that rAd-MLC3f injection rescues hindlimb unloading-induced decline in Vo in MHC type IIB single muscle fibers.


Subject(s)
Adaptation, Physiological , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Muscular Disorders, Atrophic/prevention & control , Myosin Light Chains/biosynthesis , Adenoviridae , Animals , Genetic Vectors , Hindlimb Suspension , Male , Muscle Fibers, Skeletal/pathology , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/metabolism , Myosin Light Chains/genetics , Rats , Rats, Inbred F344 , Transduction, Genetic
13.
Med Sci Sports Exerc ; 40(3): 529-34, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18379217

ABSTRACT

PURPOSE: The aim of the present study was to compare the effects of periodic restriction of blood flow to lower extremities with those of isometric exercise on disuse muscular atrophy and weakness induced by immobilization and unloading. METHODS: The left ankle of each of 15 healthy males was immobilized for 2 wk using cast, and subjects were instructed to walk using crutches with non-weight bearing during this period. Subjects were divided into three groups: a restriction of blood flow (RBF) group (application of external compressive force of 200 mm Hg for 5 min followed by 3 min of rest, repeated five times in a single session, two sessions per day for 14 d); an isometric training (IMT) group (20 "exercises" of 5-s isometric contraction of the knee extensor, flexor, and ankle plantar flexor muscles followed by rest, twice a day, daily for 2 wk); and a control (CON) group (no intervention). We measured changes in muscle strength, thigh/leg circumferences, and serum growth hormone levels. RESULTS: Immobilization/unloading resulted in significant decreases in muscle strength of knee extensor and flexor muscles (P < 0.01 and < 0.05, respectively) and thigh and leg circumferences (P < 0.05, each) in the CON group, and significant decreases in muscle strength of the knee flexor muscles, ankle plantar flexor muscles, and leg circumference (P < 0.05) in the IMT group. RBF protected against these changes in muscle strength and thigh/leg circumference (P < 0.01 and < 0.05, respectively). No changes in serum growth hormone levels were noted. CONCLUSION: Our results indicate that repetitive restriction of blood flow to the lower extremity prevents disuse muscular weakness.


Subject(s)
Exercise Therapy , Immobilization , Muscle Weakness , Muscle, Skeletal/blood supply , Muscular Disorders, Atrophic/prevention & control , Adult , Ankle Joint , Growth Hormone , Humans , Lower Extremity/physiology , Male , Pressure
14.
J Appl Physiol (1985) ; 125(3): 850-861, 2018 09 01.
Article in English | MEDLINE | ID: mdl-28970205

ABSTRACT

Numerous situations, such as the recovery from illness or rehabilitation after injury, necessitate a period of muscle disuse in otherwise healthy individuals. Even a few days of immobilization or bed rest can lead to substantial loss of skeletal muscle tissue and compromise metabolic health. The decline in muscle mass is attributed largely to a decline in postabsorptive and postprandial muscle protein synthesis rates. Reintroduction of some level of muscle contraction by the application of neuromuscular electrical stimulation (NMES) can augment both postabsorptive and postprandial muscle protein synthesis rates and, as such, prevent or attenuate muscle loss during short-term disuse in various clinical populations. Whereas maintenance of habitual dietary protein consumption is a prerequisite for muscle mass maintenance, supplementing dietary protein above habitual intake levels does not prevent muscle loss during disuse in otherwise healthy humans. Combining the anabolic properties of physical activity (or surrogates) with appropriate nutritional support likely further increases the capacity to preserve skeletal muscle mass during a period of disuse. Therefore, effective interventional strategies to prevent or alleviate muscle disuse atrophy should include both exercise (mimetics) and appropriate nutritional support.


Subject(s)
Dietary Proteins/therapeutic use , Electric Stimulation , Muscular Disorders, Atrophic/prevention & control , Animals , Humans , Muscle, Skeletal/physiopathology , Muscular Atrophy , Muscular Disorders, Atrophic/diet therapy , Nutritional Support
15.
Appl Physiol Nutr Metab ; 43(11): 1131-1139, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29800529

ABSTRACT

The contralateral effects of unilateral strength training, known as cross-education of strength, date back well over a century. In the last decade, a limited number of studies have emerged demonstrating the preservation or "sparing" effects of cross-education during immobilization. Recently published evidence reveals that the sparing effects of cross-education show muscle site specificity and involve preservation of muscle cross-sectional area. The new research also demonstrates utility of training with eccentric contractions as a potent stimulus to preserve immobilized limb strength across multiple modes of contraction. The cumulative data in nonclinical settings suggest that cross-education can completely abolish expected declines in strength and muscle size in the range of ∼13% and ∼4%, respectively, after 3-4 weeks of immobilization of a healthy arm. The evidence hints towards the possibility that unique mechanisms may be involved in preservation effects of cross-education, as compared with those that lead to functional improvements under normal conditions. Cross-education effects after strength training appear to be larger in clinical settings, but there is still only 1 randomized clinical trial demonstrating the potential utility of cross-education in addition to standard treatment. More work is necessary in both controlled and clinical settings to understand the potential interaction of neural and muscle adaptations involved in the observed sparing effects, but there is growing evidence to advocate for the clinical utility of cross-education.


Subject(s)
Muscle Strength/physiology , Muscular Disorders, Atrophic , Resistance Training , Restraint, Physical/adverse effects , Arm/physiopathology , Arm Injuries/rehabilitation , Arm Injuries/therapy , Humans , Muscular Disorders, Atrophic/etiology , Muscular Disorders, Atrophic/physiopathology , Muscular Disorders, Atrophic/prevention & control
16.
Ageing Res Rev ; 47: 80-88, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30031068

ABSTRACT

Disuse situations can have serious adverse health consequences in the elderly, including mainly functional impairment with subsequent increase in the risk of falls or morbimortality. The present review provides clinicians and care givers with detailed and practical information on the feasibility and effectiveness of physical strategies that are currently available to prevent or attenuate the functional decline that occurs secondarily to disuse situations in the elderly, notably in the hospital setting. In this context, active approaches such as resistance exercises and maximal voluntary contractions, which can be performed both isometrically and dynamically, are feasible during most immobilization situations including in hospitalized old people and represent powerful tools for the prevention of muscle atrophy. Aerobic exercise should also be prescribed whenever possible to reduce the loss of cardiovascular capacity associated with disuse periods. Other feasible strategies for patients who are unwilling or unable to perform volitional exercise comprise neuromuscular electrical stimulation, vibration, and blood flow restriction. However, they should ideally be applied synchronously with voluntary exercise to obtain synergistic benefits.


Subject(s)
Aging/physiology , Exercise/physiology , Muscle, Skeletal/physiology , Muscular Disorders, Atrophic/prevention & control , Muscular Disorders, Atrophic/physiopathology , Aged , Animals , Humans , Muscle Contraction/physiology , Muscle, Skeletal/pathology , Muscular Atrophy/diagnosis , Muscular Atrophy/physiopathology , Muscular Atrophy/prevention & control , Muscular Disorders, Atrophic/diagnosis
17.
Sci Rep ; 8(1): 3311, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29459641

ABSTRACT

The aim of this study was to clarify the mechanism of disuse-induced muscle hyperalgesia through the evaluation of the pharmacological behaviour of muscle hyperalgesia profiles in chronic post-cast pain (CPCP) rats with acute and chronic-phase mirror-image muscle hyperalgesia treated with diclofenac (NSAID), pregabalin (an inhibitor of Ca2+ channel α2δ), and duloxetine (SNRI). After 2 weeks of cast immobilization, the peak cross-sectional area and muscle wet weight of the ipsilateral soleus and gastrocnemius muscles decreased more significantly in CPCP rats than in untreated rats. Histological findings revealed disuse-induced muscle atrophy in CPCP rats. The blood biochemical parameters of CPCP rats in acute and chronic phases did not differ significantly from those of untreated rats. The diclofenac and pregabalin-treated groups exhibited no improvement in acute or chronic muscle hyperalgesia. In contrast, the duloxetine-treated group exhibited an improvement in acute muscle hyperalgesia, but showed no apparent effect on chronic muscle hyperalgesia on ipsilateral or contralateral sides. However, the chronic muscle hyperalgesia was reversed by intrathecal administration of DAMGO (a µ-opioid receptor agonist). The results suggest that chronic muscle hyperalgesia in CPCP rats did not result from an inflammatory mechanism, and there is only a low probability that it's caused by a neuropathic mechanism.


Subject(s)
Chronic Pain/drug therapy , Diclofenac/administration & dosage , Duloxetine Hydrochloride/administration & dosage , Musculoskeletal Pain/drug therapy , Pregabalin/administration & dosage , Animals , Chronic Pain/physiopathology , Disease Models, Animal , Humans , Muscle, Skeletal/drug effects , Muscular Disorders, Atrophic/physiopathology , Muscular Disorders, Atrophic/prevention & control , Musculoskeletal Pain/physiopathology , Pain Measurement , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley
18.
Mol Nutr Food Res ; 62(9): e1700941, 2018 05.
Article in English | MEDLINE | ID: mdl-29578301

ABSTRACT

SCOPE: In this study, we aim to determine the effects of resveratrol (RSV) on muscle atrophy in streptozocin-induced diabetic mice and to explore mitochondrial quality control (MQC) as a possible mechanism. METHODS AND RESULTS: The experimental mice were fed either a control diet or an identical diet containing 0.04% RSV for 8 weeks. Examinations were subsequently carried out, including the effects of RSV on muscle atrophy and muscle function, as well as on the signaling pathways related to protein degradation and MQC processes. The results show that RSV supplementation improves muscle atrophy and muscle function, attenuates the increase in ubiquitin and muscle RING-finger protein-1 (MuRF-1), and simultaneously attenuates LC3-II and cleaved caspase-3 in the skeletal muscle of diabetic mice. Moreover, RSV treatment of diabetic mice results in an increase in mitochondrial biogenesis and inhibition of the activation of mitophagy in skeletal muscle. RSV also protects skeletal muscle against excess mitochondrial fusion and fission in the diabetic mice. CONCLUSION: The results suggest that RSV ameliorates diabetes-induced skeletal muscle atrophy by modulating MQC.


Subject(s)
Antioxidants/therapeutic use , Diabetes Mellitus, Experimental/complications , Dietary Supplements , Mitochondria, Muscle/metabolism , Mitochondrial Dynamics , Muscular Disorders, Atrophic/prevention & control , Resveratrol/therapeutic use , Animals , Apoptosis , Autophagy , Biomarkers/metabolism , Diabetes Mellitus, Experimental/physiopathology , Gene Expression Regulation , Male , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondria, Muscle/pathology , Mitochondria, Muscle/ultrastructure , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle Strength , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscle, Skeletal/ultrastructure , Muscular Atrophy/complications , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Muscular Disorders, Atrophic/complications , Muscular Disorders, Atrophic/metabolism , Muscular Disorders, Atrophic/pathology , Signal Transduction , Streptozocin , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin/antagonists & inhibitors , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
19.
Physiol Int ; 104(4): 316-328, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29278025

ABSTRACT

This study examined the aging effect on disuse muscle atrophy prevention using heat stress. Wistar rats aged 7 and 60 weeks were divided into three groups as follows: control, immobilized (Im), and immobilized and heat stressed (ImH). Heat stress was given by immersing the hindlimbs in hot water (42 °C) for 60 min, once in every 3 days and the gastrocnemius (GAS) and soleus (SOL) muscles were extracted after 14 days. Muscle-fiber types were classified using ATPase staining. Heat shock protein 70 (HSP70) was assessed through Western blotting. In GAS muscle of both groups and SOL muscle of 7-week-old rats, the fiber diameter of each muscle type in the ImH group significantly increased compared with that in the Im group. However, this could not be observed in the SOL muscle of the 60-week-old rats. The increased percentage of type-I fibers and variability of types I and II muscle-fiber diameter were evident in the SOL muscle of the 60-week rats. HSP70 was significantly elevated in the ImH group compared with in the Im group in both muscle types of both age groups. Thus, effectiveness of heat stress in the prevention of disuse muscle atrophy appears unsatisfactory in aging muscle fibers.


Subject(s)
Aging , HSP70 Heat-Shock Proteins/metabolism , Hyperthermia, Induced/methods , Muscle, Skeletal/physiopathology , Muscular Disorders, Atrophic/prevention & control , Muscular Disorders, Atrophic/physiopathology , Animals , Heat-Shock Response , Male , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Disorders, Atrophic/diagnosis , Rats , Rats, Wistar , Treatment Outcome
20.
Int J Nanomedicine ; 12: 1985-1999, 2017.
Article in English | MEDLINE | ID: mdl-28331320

ABSTRACT

Angiotensin (1-7) (Ang-(1-7)) is a bioactive heptapeptide with a short half-life and has beneficial effects in several tissues - among them, skeletal muscle - by preventing muscle atrophy. Dendrimers are promising vehicles for the protection and transport of numerous bioactive molecules. This work explored the use of a neutral, non-cytotoxic hydroxyl-terminated poly(amidoamine) (PAMAM-OH) dendrimer as an Ang-(1-7) carrier. Bioinformatics analysis showed that the Ang-(1-7)-binding capacity of the dendrimer presented a 2:1 molar ratio. Molecular dynamics simulation analysis revealed the capacity of neutral PAMAM-OH to protect Ang-(1-7) and form stable complexes. The peptide coverage ability of the dendrimer was between ~50% and 65%. Furthermore, an electrophoretic mobility shift assay demonstrated that neutral PAMAM-OH effectively bonded peptides. Experimental results showed that the Ang-(1-7)/PAMAM-OH complex, but not Ang-(1-7) alone, had an anti-atrophic effect when administered intraperitoneally, as evaluated by muscle strength, fiber diameter, myofibrillar protein levels, and atrogin-1 and MuRF-1 expressions. The results of the Ang-(1-7)/PAMAM-OH complex being intraperitoneally injected were similar to the results obtained when Ang-(1-7) was systemically administered through mini-osmotic pumps. Together, the results suggest that Ang-(1-7) can be protected for PAMAM-OH when this complex is intraperitoneally injected. Therefore, the Ang-(1-7)/PAMAM-OH complex is an efficient delivery method for Ang-(1-7), since it improves the anti-atrophic activity of this peptide in skeletal muscle.


Subject(s)
Angiotensin I/therapeutic use , Dendrimers/chemistry , Muscle, Skeletal/pathology , Muscular Atrophy/drug therapy , Muscular Atrophy/prevention & control , Muscular Disorders, Atrophic/drug therapy , Muscular Disorders, Atrophic/prevention & control , Peptide Fragments/therapeutic use , Angiotensin I/pharmacology , Animals , Electrophoretic Mobility Shift Assay , Immobilization , Injections, Intraperitoneal , Male , Mice, Inbred C57BL , Molecular Dynamics Simulation , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscular Disorders, Atrophic/pathology , Myosin Heavy Chains/metabolism , Peptide Fragments/pharmacology , Peptides/therapeutic use , SKP Cullin F-Box Protein Ligases/metabolism , Static Electricity , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL