Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 384
Filter
1.
Exp Neurol ; 376: 114772, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599366

ABSTRACT

Animals on Earth need to hold postures and execute a series of movements under gravity and atmospheric pressure. VAChT-Cre is a transgenic Cre driver mouse line that expresses Cre recombinase selectively in motor neurons of S-type (slow-twitch fatigue-resistant) and FR-type (fast-twitch fatigue-resistant). Sequential motor unit recruitment is a fundamental principle for fine and smooth locomotion; smaller-diameter motor neurons (S-type, FR-type) first contract low-intensity oxidative type I and type IIa muscle fibers, and thereafter larger-diameter motor neurons (FInt-type, FF-type) are recruited to contract high-intensity glycolytic type IIx and type IIb muscle fibers. To selectively eliminate S- and FR-type motor neurons, VAChT-Cre mice were crossbred with NSE-DTA mice in which the cytotoxic diphtheria toxin A fragment (DTA) was expressed in Cre-expressing neurons. The VAChT-Cre;NSE-DTA mice were born normally but progressively manifested various characteristics, including body weight loss, kyphosis, kinetic and postural tremor, and muscular atrophy. The progressive kinetic and postural tremor was remarkable from around 20 weeks of age and aggravated. Muscular atrophy was apparent in slow muscles, but not in fast muscles. The increase in motor unit number estimation was detected by electromyography, reflecting compensatory re-innervation by remaining FInt- and FF-type motor neurons to the orphaned slow muscle fibers. The muscle fibers gradually manifested fast/slow hybrid phenotypes, and the remaining FInt-and FF-type motor neurons gradually disappeared. These results suggest selective ablation of S- and FR-type motor neurons induces progressive muscle fiber-type transition, exhaustion of remaining FInt- and FF-type motor neurons, and late-onset kinetic and postural tremor in mice.


Subject(s)
Mice, Transgenic , Motor Neurons , Tremor , Animals , Motor Neurons/pathology , Motor Neurons/physiology , Mice , Tremor/genetics , Tremor/physiopathology , Muscle Fibers, Slow-Twitch/pathology , Muscle Fibers, Fast-Twitch/pathology , Muscular Diseases/physiopathology , Muscular Diseases/pathology , Muscular Diseases/etiology , Muscle Fatigue/physiology , Posture/physiology , Animals, Newborn , Disease Models, Animal
2.
Can J Physiol Pharmacol ; 102(5): 342-360, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38118126

ABSTRACT

Sarcopenia is a musculoskeletal disease that reduces muscle mass and strength in older individuals. The study investigates the effects of azilsartan (AZL) on skeletal muscle loss in natural sarcopenic rats. Male Sprague-Dawley rats aged 4-6 months and 18-21 months were selected as young-matched control and natural-aged (sarcopenic) rats, respectively. Rats were allocated into young and old control (YC and OC) and young and old AZL treatment (YT and OT) groups, which received vehicles and AZL (8 mg/kg, orally) for 6 weeks. Rats were then sacrificed after muscle function analysis. Serum and gastrocnemius (GN) muscles were isolated for further endpoints. AZL significantly improved muscle grip strength and antioxidant levels in sarcopenic rats. AZL also restored the levels of insulin, testosterone, and muscle biomarkers such as myostatin and creatinine kinase in sarcopenic rats. Furthermore, AZL treatment improved the cellular and ultrastructure of GN muscle and prevented the shift of type II (glycolytic) myofibers to type I (oxidative) myofibers. The results showed that AZL intervention restored protein synthesis in natural sarcopenic rats by increasing p-Akt-1 and decreasing muscle RING-finger protein-1 and tumor necrosis factor alpha immunoexpressions. In conclusion, the present findings showed that AZL could be an effective intervention in treating age-related muscle impairments.


Subject(s)
Aging , Benzimidazoles , Muscle Fibers, Fast-Twitch , Muscle Fibers, Slow-Twitch , Oxadiazoles , Rats, Sprague-Dawley , Sarcopenia , Animals , Sarcopenia/prevention & control , Sarcopenia/metabolism , Sarcopenia/drug therapy , Sarcopenia/pathology , Male , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Aging/drug effects , Rats , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Proto-Oncogene Proteins c-akt/metabolism , Myostatin/metabolism , Antioxidants/pharmacology
3.
Cells ; 10(7)2021 07 08.
Article in English | MEDLINE | ID: mdl-34359900

ABSTRACT

Store-operated Ca2+ entry (SOCE) is a ubiquitous mechanism regulating extracellular Ca2+ entry to control a multitude of Ca2+-dependent signaling pathways and cellular processes. SOCE relies on the concerted activity of the reticular Ca2+ sensor STIM1 and the plasma membrane Ca2+ channel ORAI1, and dysfunctions of these key factors result in human pathologies. STIM1 and ORAI1 gain-of-function (GoF) mutations induce excessive Ca2+ influx through SOCE over-activation, and cause tubular aggregate myopathy (TAM) and Stormorken syndrome (STRMK), two overlapping disorders characterized by muscle weakness and additional multi-systemic signs affecting growth, platelets, spleen, skin, and intellectual abilities. In order to investigate the pathophysiological effect of overactive SOCE on muscle function and structure, we combined transcriptomics with morphological and functional studies on a TAM/STRMK mouse model. Muscles from Stim1R304W/+ mice displayed aberrant expression profiles of genes implicated in Ca2+ handling and excitation-contraction coupling (ECC), and in vivo investigations evidenced delayed muscle contraction and relaxation kinetics. We also identified signs of reticular stress and abnormal mitochondrial activity, and histological and respirometric analyses on muscle samples revealed enhanced myofiber degeneration associated with reduced mitochondrial respiration. Taken together, we uncovered a molecular disease signature and deciphered the pathomechanism underlying the functional and structural muscle anomalies characterizing TAM/STRMK.


Subject(s)
Muscles/pathology , Muscles/physiopathology , Stromal Interaction Molecule 1/metabolism , Animals , Calcium Signaling , Cell Death , Endoplasmic Reticulum Stress , Excitation Contraction Coupling , Gene Expression Regulation , Gene Regulatory Networks , Mice , Mitochondria/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscles/metabolism , Mutation/genetics , Stromal Interaction Molecule 1/genetics , Transcriptome/genetics
4.
Am J Physiol Cell Physiol ; 321(4): C704-C720, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34432537

ABSTRACT

Duchenne muscular dystrophy (DMD) is the second most common fatal genetic disease in humans and is characterized by the absence of a functional copy of the protein dystrophin from skeletal muscle. In dystrophin-negative humans and rodents, regenerated skeletal muscle fibers show abnormal branching. The number of fibers with branches and the complexity of branching increases with each cycle of degeneration/regeneration. Previously, using the mdx mouse model of DMD, we have proposed that once the number and complexity of branched fibers present in dystrophic fast-twitch EDL muscle surpasses a stable level, we term the "tipping point," the branches, in and of themselves, mechanically weaken the muscle by rupturing when subjected to high forces during eccentric contractions. Here, we use the slow-twitch soleus muscle from the dystrophic mdx mouse to study prediseased "periambulatory" dystrophy at 2-3 wk, the peak regenerative "adult" phase at 6-9 wk, and "old" at 58-112 wk. Using isolated mdx soleus muscles, we examined contractile function and response to eccentric contraction correlated with the amount and complexity of regenerated branched fibers. The intact muscle was enzymatically dispersed into individual fibers in order to count fiber branching and some muscles were optically cleared to allow laser scanning confocal microscopy. We demonstrate throughout the lifespan of the mdx mouse that dystrophic slow-twitch soleus muscle is no more susceptible to eccentric contraction-induced injury than age-matched littermate controls and that this is correlated with a reduction in the number and complexity of branched fibers compared with fast-twitch dystrophic EDL muscles.


Subject(s)
Dystrophin/deficiency , Muscle Contraction , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscular Dystrophy, Duchenne/metabolism , Age Factors , Animals , Disease Models, Animal , Dystrophin/genetics , Kinetics , Male , Mice, Inbred mdx , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/pathology , Muscle Strength , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Mutation
5.
FASEB J ; 35(7): e21714, 2021 07.
Article in English | MEDLINE | ID: mdl-34118107

ABSTRACT

We tested the hypothesis that cancer cachexia progression would induce oxidative post-translational modifications (Ox-PTMs) associated with skeletal muscle wasting, with different responses in muscles with the prevalence of glycolytic and oxidative fibers. We used cysteine-specific isotopic coded affinity tags (OxICAT) and gel-free mass spectrometry analysis to investigate the cysteine Ox-PTMs profile in the proteome of both plantaris (glycolytic) and soleus (oxidative) muscles in tumor-bearing and control rats. Histological analysis revealed muscle atrophy in type II fibers in plantaris muscle, with no changes in plantaris type I fibers and no differences in both soleus type I and II fibers in tumor-bearing rats when compared to healthy controls. Tumor progression altered the Ox-PTMs profile in both plantaris and soleus. However, pathway analysis including the differentially oxidized proteins revealed tricarboxylic acid cycle and oxidative phosphorylation as main affected pathways in plantaris muscle from tumor-bearing rats, while the same analysis did not show main metabolic pathways affected in the soleus muscle. In addition, cancer progression affected several metabolic parameters such as ATP levels and markers of oxidative stress associated with muscle atrophy in plantaris muscle, but not in soleus. However, isolated soleus from tumor-bearing rats had a reduced force production capacity when compared to controls. These novel findings demonstrate that tumor-bearing rats have severe muscle atrophy exclusively in glycolytic fibers. Cancer progression is associated with cysteine Ox-PTMs in the skeletal muscle, but these modifications affect different pathways in a glycolytic muscle compared to an oxidative muscle, indicating that intrinsic muscle oxidative capacity determines the response to cancer cachectic effects.


Subject(s)
Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Neoplasms/pathology , Oxidative Stress/physiology , Animals , Cachexia/pathology , Disease Progression , Glycolysis/physiology , Male , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/pathology , Oxidation-Reduction , Oxidative Phosphorylation , Rats , Rats, Wistar
6.
Cell Tissue Res ; 384(3): 745-756, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33660050

ABSTRACT

Skeletal muscle fibers are classified as slow-twitch and fast-twitch fibers, which have different reactive oxygen species (ROS) metabolism and mitochondrial biogenesis. Recently, Attractin (Atrn), which encodes secreted (sAtrn) and transmembrane (mAtrn)-type proteins, has been shown to be involved in free radical scavenging. Although Atrn has been found in skeletal muscle, little is known about the expression levels and function of Atrn in each muscle fiber type. Therefore, we investigate sAtrn and mAtrn expression levels in the slow-twitch soleus (sol) and fast-twitch extensor digitorum longus (EDL) muscles as well as the morphology and expression levels of antioxidant enzymes and functional mitochondrial markers using Atrn-deficient muscles. Both types of Atrn were expressed in the sol and EDL. mAtrn was mainly expressed in the adult sol, whereas sAtrn expression levels did not differ between muscle types. Moreover, mAtrn in the sol was abundantly localized in the subsarcolemmal area, especially in the myoplasm near mitochondria. Atrn-deficient Zitter rats showed muscle fiber atrophy, myofibril misalignment, mitochondrial swelling and vacuolation in the sol but not EDL. Furthermore, the Atrn-deficient sol exhibited a marked reduction in antioxidant enzyme SOD1, GPx1, catalase and Prx6 and mitochondrial functional protein, UCP2, expression. Even Atrn-deficient EDL showed a significant reduction in Prx3, Prx6, UCP2 and UCP3 expression. These data indicate that Atrn-deficiency disturbs ROS metabolism in skeletal muscles. In particular, mAtrn is involved in metabolism in the slow-twitch sol muscle and mAtrn-deficiency may cause ROS imbalance, resulting in morphological abnormalities in the muscle.


Subject(s)
Membrane Proteins/deficiency , Muscle Fibers, Slow-Twitch , Muscular Diseases/metabolism , Animals , Male , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Rats , Rats, Sprague-Dawley
7.
Cell Signal ; 81: 109939, 2021 05.
Article in English | MEDLINE | ID: mdl-33529759

ABSTRACT

BACKGROUND: previous studies have shown that muscle atrophy is observed after sleep deprivation (SD) protocols; however, the mechanisms responsible are not fully understood. Muscle trophism can be modulated by several factors, including energy balance (positive or negative), nutritional status, oxidative stress, the level of physical activity, and disuse. The metabolic differences that exist in different types of muscle fiber may also be the result of different adaptive responses. To better understand these mechanisms, we evaluated markers of oxidative damage and histopathological changes in different types of muscle fibers in sleep-deprived rats. METHODS: Twenty male Wistar EPM-1 rats were randomly allocated in two groups: a control group (CTL group; n = 10) and a sleep deprived group (SD group; n = 10). The SD group was submitted to continuous paradoxical SD for 96  h; the soleus (type I fibers) and plantar (type II fiber) muscles were analyzed for histopathological changes, trophism, lysosomal activity, and oxidative damage. Oxidative damage was assessed by lipid peroxidation and nuclear labeling of 8-OHdG. RESULTS: The data demonstrated that SD increased the nuclear labeling of 8-OHdG and induced histopathological changes in both muscles, being more evident in the soleus muscle. In the type I fibers there was signs of tissue degeneration, inflammatory infiltrate and tissue edema. Muscle atrophy was observed in both muscles. The concentration of malondialdehyde, and cathepsin L activity only increased in type I fibers after SD. CONCLUSION: These data indicate that the histopathological changes observed after 96 h of SD in the skeletal muscle occur by different processes, according to the type of muscle fiber, with muscles predominantly composed of type I fibers undergoing greater oxidative damage and catabolic activity, as evidenced by a larger increase in 8-OHdG labeling, lipid peroxidation, and lysosomal activity.


Subject(s)
Muscle Fibers, Fast-Twitch , Muscle Fibers, Slow-Twitch , Oxidative Stress , Sleep Deprivation , Animals , Male , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Rats , Rats, Wistar , Sleep Deprivation/metabolism , Sleep Deprivation/pathology
8.
Biomed Pharmacother ; 133: 110977, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33249280

ABSTRACT

Puerarin is an isoflavonoid extracted from Pueraria lobate with extensive pharmacological effects in traditional Chinese medicine. The evidence implicates that puerarin mitigates hyperglycemia and various relevant complications. Here, the effect of puerarin on skeletal muscle wasting induced by type 1 diabetes (T1D) was explored. Streptozotocin (STZ)-induced T1D male Sprague Dawley (SD) rats were used in this study. Muscle strength, weight and size were measured. L6 rat skeletal muscle cells were applied for in vitro study. Our results showed that eight-week oral puerarin administration (100 mg/kg) increased muscle strengths and weights accompanied by enhanced skeletal muscle cross-sectional areas in diabetic rats. Simultaneously, puerarin also reduced expressions of several muscle wasting marker genes including F-box only protein 32 (Atrogin-1) and muscle-specific RING-finger 1 (Murf-1) in diabetic group both in vitro and in vivo. Transformation from type I fibers (slow muscle) to type II fibers (fast muscle) were also observed under puerarin administration in diabetic rats. Puerarin promoted Akt/mTOR while inhibited LC3/p62 signaling pathway in skeletal muscle cells. In conclusion, our study showed that puerarin mitigated skeletal muscle wasting in T1D rats and closely related with Akt/mTOR activation and autophagy inhibition. Whether this effect in murine applies to humans remains to be determined.


Subject(s)
Cell Differentiation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Isoflavones/pharmacology , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/drug effects , Muscular Atrophy/prevention & control , Animals , Blood Glucose/metabolism , Cell Line , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/chemically induced , Male , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle Strength/drug effects , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , Streptozocin , TOR Serine-Threonine Kinases/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
9.
Int J Mol Sci ; 21(19)2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32992783

ABSTRACT

Nearly 100 years ago, Otto Warburg investigated the metabolism of growing tissues and discovered that tumors reprogram their metabolism. It is poorly understood whether and how hypertrophying muscle, another growing tissue, reprograms its metabolism too. Here, we studied pyruvate kinase muscle (PKM), which can be spliced into two isoforms (PKM1, PKM2). This is of interest, because PKM2 redirects glycolytic flux towards biosynthetic pathways, which might contribute to muscle hypertrophy too. We first investigated whether resistance exercise changes PKM isoform expression in growing human skeletal muscle and found that PKM2 abundance increases after six weeks of resistance training, whereas PKM1 decreases. Second, we determined that Pkm2 expression is higher in fast compared to slow fiber types in rat skeletal muscle. Third, by inducing hypertrophy in differentiated C2C12 cells and by selectively silencing Pkm1 and/or Pkm2 with siRNA, we found that PKM2 limits myotube growth. We conclude that PKM2 contributes to hypertrophy in C2C12 myotubes and indicates a changed metabolic environment within hypertrophying human skeletal muscle fibers. PKM2 is preferentially expressed in fast muscle fibers and may partly contribute to the increased potential for hypertrophy in fast fibers.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Muscle Fibers, Fast-Twitch/enzymology , Muscle Fibers, Slow-Twitch/enzymology , Resistance Training , Thyroid Hormones/metabolism , Adult , Cell Line , Humans , Hypertrophy , Male , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/pathology , Thyroid Hormone-Binding Proteins
10.
J Neurol Neurosurg Psychiatry ; 91(10): 1085-1091, 2020 10.
Article in English | MEDLINE | ID: mdl-32934110

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is a hereditary neuromuscular disorder caused by CAG trinucleotide expansion in the gene encoding the androgen receptor (AR). In the central nervous system, lower motor neurons are selectively affected, whereas pathology of patients and animal models also indicates involvement of skeletal muscle including loss of fast-twitch type 2 fibres and increased slow-twitch type 1 fibres, together with a glycolytic-to-oxidative metabolic switch. Evaluation of muscle and fat using MRI, in addition to biochemical indices such as serum creatinine level, are promising biomarkers to track the disease progression. The serum level of creatinine starts to decrease before the onset of muscle weakness, followed by the emergence of hand tremor, a prodromal sign of the disease. Androgen-dependent nuclear accumulation of the polyglutamine-expanded AR is an essential step in the pathogenesis, providing therapeutic opportunities via hormonal manipulation and gene silencing with antisense oligonucleotides. Animal studies also suggest that hyperactivation of Src, alteration of autophagy and a mitochondrial deficit underlie the neuromuscular degeneration in SBMA and provide alternative therapeutic targets.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked/metabolism , Bulbo-Spinal Atrophy, X-Linked/therapy , 5-alpha Reductase Inhibitors/therapeutic use , Adipose Tissue/diagnostic imaging , Adrenergic beta-Agonists/therapeutic use , Autophagy , Biomarkers , Bulbo-Spinal Atrophy, X-Linked/diagnostic imaging , Bulbo-Spinal Atrophy, X-Linked/physiopathology , Clenbuterol/therapeutic use , Creatinine/metabolism , Dutasteride/therapeutic use , Glycolysis , Humans , Insulin-Like Growth Factor I/analogs & derivatives , Leuprolide/therapeutic use , Magnetic Resonance Imaging , Mitochondria/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Oligonucleotides, Antisense/therapeutic use , Oxidation-Reduction , RNAi Therapeutics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Trinucleotide Repeat Expansion
11.
Neurobiol Dis ; 145: 105052, 2020 11.
Article in English | MEDLINE | ID: mdl-32827689

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult-onset disease characterized by the progressive death of motoneurons and denervation of muscle fibers. To restore motor function, surviving motoneurons in partially denervated muscles typically sprout axons to reinnervate denervated endplates. However, studies on the SOD1G93A rodent models of ALS indicate that sprouting is significantly limited in fast, but not slow, twitch muscles after disease onset. This limitation hastens the rate of muscle weakness and loss of motor function. The causes of this limitation are currently unknown. Sprouting could be limited because the SOD1G93A mutation weakens motoneurons making them incapable of expanding their field of innervation. Alternatively, motoneurons may be capable of sprouting, but unable to do so due to the loss of a permissive sprouting environment. To distinguish between the two possibilities, we compared the sprouting capacity of motoneuron subtypes by partially denervating the fast twitch plantaris (composed of type IIa/IIb muscle fibers) and slow twitch soleus muscles (type I/IIa fibers) prior to disease onset and weakening in SOD1G93A and WT mice. We found that only motoneurons innervating the SOD1G93A plantaris had a limited sprouting capacity. This was correlated with the selective loss of terminal Schwann cells (TSCs) at IIb fibers and an increase in macrophage infiltration. Treating SOD1G93A mice with the tyrosine kinase inhibitor, masitinib, significantly reduced infiltration, prevented TSC loss, and increased the sprouting capacity to near normal. These results suggest that TSCs at denervated type IIb muscle fibers are aberrantly targeted by infiltrating macrophages in SOD1G93A mice, and their loss accounts, at least in part, for the compromised sprouting capacity of the largest motoneurons during early stages of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/physiology , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/pathology , Nerve Regeneration/physiology , Schwann Cells/pathology , Animals , Mice , Mice, Transgenic , Muscle Denervation , Muscle, Skeletal/innervation , Superoxide Dismutase-1/genetics
12.
Biomed Res ; 41(3): 139-148, 2020.
Article in English | MEDLINE | ID: mdl-32522931

ABSTRACT

Radix astragali is a popular traditional herbal medicine that provides significant protection against tissue injury in various models of oxidative stress-related diseases. In this study, we aimed to investigate whether administration of Radix astragali prevented atrophy in both slow- and fast-twitch muscles following cast immobilization. Twenty-seven 12-week-old male F344 rats were divided into three experimental groups: control (CON), immobilized (IM), and immobilized with Radix astragali administration (IM+AR). Rats in the IM and IM+AR groups were subjected to immobilization of both lower extremities using casting-tape for 14 days. Rats in the IM+AR group were orally administered a decoction of Radix astragali daily for 21 days beginning 7 days before cast immobilization. As expected, rats in the IM group showed significant decreases (P < 0.05) in soleus and plantaris muscle-to-body weight ratios by 74.3% and 70.5%, respectively, compared with those in the CON group. Administration of Radix astragali significantly reversed (+35.5%) the weight reduction observed in soleus muscle, but not in the plantaris muscle, compared with that in the IM group. Furthermore, administration of Radix astragali inhibited MuRF1 mRNA expression only in the soleus muscle during cast immobilization. Our results demonstrated that administration of Radix astragali suppressed the immobilization-induced reductions in skeletal muscle mass and expression of MuRF1 mRNA in slow-twitch soleus muscles, but not in fast-twitch plantaris muscles.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Slow-Twitch/drug effects , Muscular Atrophy/drug therapy , Animals , Astragalus propinquus , Gene Expression , Hindlimb , Immobilization/adverse effects , Immobilization/methods , Male , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Organ Size/drug effects , Phytotherapy/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
13.
Muscle Nerve ; 62(2): 284-288, 2020 08.
Article in English | MEDLINE | ID: mdl-32367547

ABSTRACT

INTRODUCTION: The mechanism by which weakness develops in idiopathic inflammatory myopathies (IIMs) is still unclear. In this study we investigated the maximum force of single muscle fibers from patients with IIMs. METHODS: Permeabilized single muscle fibers from patients with IIMs and healthy controls were subjected to contractility measurements. Maximum force and specific force production (maximum force normalized to fiber size) and fiber type were determined for each isolated fiber. RESULTS: A total of 178 fibers were studied from five patients with IIMs and 95 fibers from four controls. Specific force production was significantly lower in the IIM group for all fiber types. DISCUSSION: The findings from this exploratory study suggest that weakness in IIMs may, in part, be caused by dysfunction of the contractile apparatus. These findings provide a basis for further studies into the mechanisms underlying weakness in IIMs.


Subject(s)
Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Skeletal/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle Strength/physiology , Myositis/physiopathology , Adult , Biopsy , Case-Control Studies , Cell Size , Dermatomyositis/metabolism , Dermatomyositis/pathology , Dermatomyositis/physiopathology , Female , Humans , Middle Aged , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Myosin Heavy Chains/metabolism , Myositis/metabolism , Myositis/pathology , Polymyositis/metabolism , Polymyositis/pathology , Polymyositis/physiopathology , Young Adult
14.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R360-R368, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31850817

ABSTRACT

We examined molecular mechanisms that were altered during rapid soleus (type I fiber-dominant) and plantaris (type II fiber-dominant) hypertrophy in rats. Twelve Wistar rats (3.5 mo old; 6 female, 6 male) were subjected to surgical right-leg soleus and plantaris dual overload [synergist ablation (SA)], and sham surgeries were performed on left legs (CTL). At 14 days after surgery, the muscles were dissected. Plantaris mass was 27% greater in the SA than CTL leg (P < 0.001), soleus mass was 13% greater in the SA than CTL leg (P < 0.001), and plantaris mass was higher than soleus mass in the SA leg (P = 0.001). Plantaris total RNA concentrations and estimated total RNA levels (suggestive of ribosome density) were 19% and 47% greater in the SA than CTL leg (P < 0.05), protein synthesis levels were 64% greater in the SA than CTL leg (P = 0.038), and satellite cell number per fiber was 60% greater in the SA than CTL leg (P = 0.003); no differences in these metrics were observed between soleus SA and CTL legs. Plantaris, as well as soleus, 20S proteasome activity was lower in the SA than CTL leg (P < 0.05), although the degree of downregulation was greater in the plantaris than soleus muscle (-63% vs. -20%, P = 0.001). These data suggest that early-phase plantaris hypertrophy occurs more rapidly than soleus hypertrophy, which coincided with greater increases in ribosome biogenesis, protein synthesis, and satellite cell density, as well as greater decrements in 20S proteasome activity, in the plantaris muscle.


Subject(s)
Ablation Techniques , Cell Proliferation , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/surgery , Satellite Cells, Skeletal Muscle/pathology , Animals , Female , Hypertrophy , Male , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA/metabolism , Rats, Wistar , Ribosomes/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Sex Factors , Time Factors
15.
J Appl Physiol (1985) ; 128(2): 229-240, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31829804

ABSTRACT

Parkinson's disease (PD) is a common neurodegenerative disorder impacting cognition, movement, and quality of life in >10 million individuals worldwide. We recently characterized and quantified a skeletal muscle pathology in PD represented by exaggerated type I myofiber grouping presumed to result from denervation-reinnervation processes. Our previous findings indicated that impaired neuromuscular junction integrity may be involved in type I grouping, which is associated with excessive motor unit activation during weight-bearing tasks. In this study, we performed transcriptional profiling to test the hypothesis that type I grouping severity would link to distinct gene expression networks. We generated transcriptome-wide poly(A) RNA-Seq data from skeletal muscle of individuals with PD [n = 12 (9 men, 3 women); 67 ± 2 yr], age- and sex-matched older adults (n = 12; 68 ± 2 yr), and sex-matched young adults (n = 12; 30 ± 1 yr). Differentially expressed genes were evaluated across cohorts. Weighted gene correlation network analysis (WGCNA) was performed to identify gene networks most correlated with indicators of abnormal type I grouping. Among coexpression networks mapping to phenotypes pathologically increased in PD muscle, one network was highly significantly correlated to type I myofiber group size and another to percentage of type I myofibers found in groups. Annotation of coexpressed networks revealed that type I grouping is associated with altered expression of genes involved in neural development, postsynaptic signaling, cell cycle regulation and cell survival, protein and energy metabolism, inflammation/immunity, and posttranscriptional regulation (microRNAs). These transcriptomic findings suggest that skeletal muscle may play an active role in signaling to promote myofiber survival, reinnervation, and remodeling, perhaps to an extreme in PD.NEW & NOTEWORTHY Despite our awareness of the impact of Parkinson's disease (PD) on motor function for over two centuries, limited attention has focused on skeletal muscle. We previously identified type I myofiber grouping, a novel indicator of muscle dysfunction in PD, presumably a result of heightened rates of denervation/reinnervation. Using transcriptional profiling to identify networks associated with this phenotype, we provide insight into potential mechanistic roles of skeletal muscle in signaling to promote its survival in PD.


Subject(s)
Gene Regulatory Networks , Muscle Fibers, Slow-Twitch/pathology , Muscle, Skeletal/pathology , Neuromuscular Junction/physiopathology , Parkinson Disease , Adult , Aged , Female , Humans , Male , Parkinson Disease/genetics , Parkinson Disease/pathology , Quality of Life , RNA-Seq , Transcriptome
16.
J Cell Physiol ; 235(1): 526-537, 2020 01.
Article in English | MEDLINE | ID: mdl-31241186

ABSTRACT

Muscle wasting is associated with chronic diseases and cancer. Elucidation of the biological mechanism involved in the process of muscle mass loss and cachexia may help identify therapeutic targets. We hypothesized that l-carnitine treatment may differentially revert muscle fiber atrophy and other structural alterations in slow- and fast-twitch limb muscles of rats bearing the Yoshida ascites hepatoma. In soleus and gastrocnemius of tumor-bearing rats (108 AH-130 Yoshida ascites hepatoma cells inoculated intraperitoneally) with and without treatment with l-carnitine (1 g/kg body weight for 7 days, intragastric), food intake, body and muscle weights, fiber typing and morphometry, morphological features, redox balance, autophagy and proteolytic, and signaling markers were explored. Levels of carnitine palmitoyl transferase were also measured in all the study muscles. l-Carnitine treatment ameliorated the atrophy of both slow- and fast-twitch fibers (gastrocnemius particularly), muscle structural alterations (both muscles), and attenuated oxidative stress, proteolytic and signaling markers (gastrocnemius). Despite that carnitine palmitoyl transferase-1 levels increased in both muscle types in a similar fashion, l-carnitine ameliorated muscle atrophy and proteolysis in a muscle-specific manner in cancer-induced cachexia. These data reveal the need to study muscles of different fiber type composition and function to better understand whereby l-carnitine exerts its beneficial effects on the myofibers in muscle wasting processes. These findings also have potential clinical implications, since combinations of various exercise and muscle training modalities with l-carnitine should be specifically targeted for the muscle groups to be trained.


Subject(s)
Cachexia/drug therapy , Carnitine/pharmacology , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/pathology , Muscular Atrophy/drug therapy , Animals , Autophagy/drug effects , Cachexia/pathology , Carcinoma, Hepatocellular/pathology , Carnitine O-Palmitoyltransferase/metabolism , Liver Neoplasms/pathology , Male , Muscle, Skeletal/physiology , Muscular Atrophy/pathology , Oxidative Stress/drug effects , Proteolysis/drug effects , Rats , Rats, Wistar , Sarcoma, Yoshida/pathology , Signal Transduction/drug effects
17.
J Neurol Sci ; 408: 116499, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31726383

ABSTRACT

BACKGROUND: Muscle pathology usually contributes to mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episode (MELAS), even in patients without prominent muscle symptoms. We report a series of patients with MELAS without significant myopathic changes. METHODS: Twelve patients without ragged-red fibers (RRFs) on muscle pathology (RRF-negative group) and 99 patients with MELAS and RRFs and/or cytochrome c oxidase (COX)-deficient fibers (control RRF-positive group) were recruited. We analyzed clinical features, neuroimaging and pathological findings, gene mutation data, immunofluorescence assay of key respiratory chain subunits of complexes I and IV and mitochondrial DNA (mtDNA) mutation load in biopsied muscle samples. RESULTS: None of the RRF-negative patients had RRF or COX-negative fibers, but four patients had strongly succinate dehydrogenase-stained vessels (SSVs). There was a lower proportion of m.3243A>G and higher proportion of mitochondria-encoded ND gene mutations in RRF-negative than RRF-positive patients. The proportion of aphasia was relatively higher, while complex I and IV subunit abundance in muscle and mutation load were lower in RRF-negative than in RRF-positive patients. CONCLUSION: RRF-negative patients had a similar disease course, clinical symptoms, and neuroimaging results to RRF-positive patients with MELAS. SSV is a valuable diagnostic indicator for MELAS. For highly suspected MELAS yet without positive myopathological findings, combined immunofluorescence and genetic studies should be used to achieve final diagnosis.


Subject(s)
MELAS Syndrome/diagnostic imaging , Muscle Fibers, Slow-Twitch/pathology , Adolescent , Adult , Child , Child, Preschool , Female , Fluorescent Antibody Technique/methods , Humans , Infant , MELAS Syndrome/genetics , MELAS Syndrome/metabolism , Magnetic Resonance Imaging/methods , Male , Middle Aged , Muscle Fibers, Slow-Twitch/chemistry , Retrospective Studies , Young Adult
18.
Sci Rep ; 9(1): 10769, 2019 07 24.
Article in English | MEDLINE | ID: mdl-31341183

ABSTRACT

Mutations in the Des gene coding for the muscle-specific intermediate filament protein desmin lead to myopathies and cardiomyopathies. We previously generated a R349P desmin knock-in mouse strain as a patient-mimicking model for the corresponding most frequent human desmin mutation R350P. Since nothing is known about the age-dependent changes in the biomechanics of affected muscles, we investigated the passive and active biomechanics of small fiber bundles from young (17-23 wks), adult (25-45 wks) and aged (>60 wks) heterozygous and homozygous R349P desmin knock-in mice in comparison to wild-type littermates. We used a novel automated biomechatronics platform, the MyoRobot, to perform coherent quantitative recordings of passive (resting length-tension curves, visco-elasticity) and active (caffeine-induced force transients, pCa-force, 'slack-tests') parameters to determine age-dependent effects of the R349P desmin mutation in slow-twitch soleus and fast-twitch extensor digitorum longus small fiber bundles. We demonstrate that active force properties are not affected by this mutation while passive steady-state elasticity is vastly altered in R349P desmin fiber bundles compatible with a pre-aged phenotype exhibiting stiffer muscle preparations. Visco-elasticity on the other hand, was not altered. Our study represents the first systematic age-related characterization of small muscle fiber bundle preparation biomechanics in conjunction with inherited desminopathy.


Subject(s)
Cardiomyopathies/pathology , Muscle Fibers, Skeletal/pathology , Muscular Dystrophies/pathology , Age Factors , Animals , Automation, Laboratory , Biomechanical Phenomena , Biotechnology/instrumentation , Biotechnology/methods , Cardiomyopathies/physiopathology , Desmin/genetics , Female , Gene Knock-In Techniques , Male , Mice , Mice, Transgenic , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Skeletal/parasitology , Muscle Fibers, Slow-Twitch/pathology , Muscle Fibers, Slow-Twitch/physiology , Muscular Dystrophies/physiopathology , Robotics/instrumentation , Robotics/methods
19.
Eur J Orthop Surg Traumatol ; 29(7): 1539-1547, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31111314

ABSTRACT

Mangled extremities were classically managed by amputation. But over the past few decades, with the advancement in surgical techniques, an increased number of limb salvages have been possible. As muscles usually get damaged in such grievous injuries, a thorough understanding of muscle regeneration may give a better insight into muscle healing in these injuries. Muscles are composed of slow and fast fibers which can be represented by slow and fast myosin, respectively. There are some animal studies which reported differential regeneration of slow and fast muscle fibers during muscle healing. We conducted this pilot study to find out whether the same holds true for muscle healing in mangled extremities also. This pilot study is designed in 15 patients with lower limb mangled extremities presenting to trauma center of PGIMER, Chandigarh, who were operated within 24 h of injury to see whether muscle healing in mangled extremities follows the same pattern. Biopsies were taken during initial surgery conducted within 24 h of injury and on the 7th day of injury when patient was posted again for secondary wound closure procedure or revision amputation. The biopsy samples were subjected to histopathological and immunohistochemistry examination using antibodies against fast and slow myosin. We found that the regenerating muscle fibers in the biopsy sample taken on the 7th day of injury showed only slow muscle fibers with the absence of fast muscle fibers when compared with the initial biopsy results showing differential regeneration of slow muscle fibers.


Subject(s)
Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Myosins/metabolism , Regeneration , Wounds and Injuries/metabolism , Wounds and Injuries/pathology , Biomarkers/metabolism , Biopsy , Humans , Lower Extremity/injuries , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/pathology , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Pilot Projects , Prospective Studies , Wounds and Injuries/surgery
20.
Chron Respir Dis ; 16: 1479973119843650, 2019.
Article in English | MEDLINE | ID: mdl-31131626

ABSTRACT

In chronic obstructive pulmonary disease (COPD), lower limb dysfunction is associated with reduced exercise capacity, increased hospitalizations and mortality. We investigated sex differences in the prevalence of quadriceps dysfunction and fibre abnormalities in a large COPD cohort, controlling for the normal sex differences in health. We compared existing data from 76 male and 38 female COPD patients where each variable was expressed as a function of gender-specific normal values (obtained from 16 male and 14 female controls). Female COPD patients had lower quadriceps muscle strength and peak workload on a maximal incremental cycle ergometry protocol compared to male patients. Female patients had a smaller type II fibre cross-sectional area (CSA) compared to male patients, suggesting a greater female preponderance to fibre atrophy, although this result was largely driven by a few male patients with a large type II fibre CSA. Female patients had significantly higher concentrations of a number of plasma pro-inflammatory cytokines including tumour necrosis factor alpha and interleukin 8 (IL8), but not lower levels of physical activity or arterial oxygenation, compared to males. Our data confirm results from a previous small study and suggest that female COPD patients have a greater prevalence of muscle wasting and weakness. Larger studies investigating sex differences in COPD-related muscle atrophy and weakness are needed, as the results will have implications for monitoring in clinical practice and for design of clinical trials evaluating novel muscle anabolic agents.


Subject(s)
Muscle Fibers, Skeletal/pathology , Muscle Weakness/physiopathology , Muscular Atrophy/physiopathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Quadriceps Muscle/physiopathology , Aged , Blood Gas Analysis , Ergometry , Exercise , Female , Forced Expiratory Volume , Humans , Inflammation/metabolism , Interleukin-8/metabolism , Male , Middle Aged , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/pathology , Muscle Strength , Muscle Weakness/etiology , Muscle Weakness/metabolism , Muscle Weakness/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , NF-kappa B/metabolism , Organ Size , Oxygen Consumption , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/metabolism , Quadriceps Muscle/pathology , Receptors, Tumor Necrosis Factor/metabolism , Sex Factors , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Walk Test
SELECTION OF CITATIONS
SEARCH DETAIL
...