Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.866
Filter
1.
Physiol Behav ; 281: 114575, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38692384

ABSTRACT

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


Subject(s)
Clenbuterol , Disease Models, Animal , Fibromyalgia , Hyperalgesia , Muscular Atrophy , Sympathetic Nervous System , Animals , Female , Fibromyalgia/pathology , Fibromyalgia/physiopathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Hyperalgesia/physiopathology , Hyperalgesia/pathology , Sympathetic Nervous System/physiopathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/pathology , Clenbuterol/pharmacology , Rats , Carrageenan/toxicity , Rats, Sprague-Dawley , Pain/pathology , Pain/physiopathology , Epinephrine , Muscle, Skeletal/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Catecholamines/metabolism , Adrenergic beta-Agonists/pharmacology
2.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732203

ABSTRACT

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


Subject(s)
Muscle Fatigue , Muscle, Skeletal , Humans , Muscle Fatigue/physiology , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Atrophy/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology
3.
Int J Mol Sci ; 25(9)2024 May 05.
Article in English | MEDLINE | ID: mdl-38732255

ABSTRACT

This research aimed to explore the healing impacts of Melittin treatment on gastrocnemius muscle wasting caused by immobilization with a cast in rabbits. Twenty-four rabbits were randomly allocated to four groups. The procedures included different injections: 0.2 mL of normal saline to Group 1 (G1-NS); 4 µg/kg of Melittin to Group 2 (G2-4 µg/kg Melittin); 20 µg/kg of Melittin to Group 3 (G3-20 µg/kg Melittin); and 100 µg/kg of Melittin to Group 4 (G4-100 µg/kg Melittin). Ultrasound was used to guide the injections into the rabbits' atrophied calf muscles following two weeks of immobilization via casting. Clinical measurements, including the length of the calf, the compound muscle action potential (CMAP) of the tibial nerve, and the gastrocnemius muscle thickness, were assessed. Additionally, cross-sectional slices of gastrocnemius muscle fibers were examined, and immunohistochemistry and Western blot analyses were performed following two weeks of therapy. The mean regenerative changes, as indicated by clinical parameters, in Group 4 were significantly more pronounced than in the other groups (p < 0.05). Furthermore, the cross-sectional area of the gastrocnemius muscle fibers and immunohistochemical indicators in Group 4 exceeded those in the remaining groups (p < 0.05). Western blot analysis also showed a more significant presence of anti-inflammatory and angiogenic cytokines in Group 4 compared to the others (p < 0.05). Melittin therapy at a higher dosage can more efficiently activate regeneration in atrophied gastrocnemius muscle compared to lower doses of Melittin or normal saline.


Subject(s)
Melitten , Muscle, Skeletal , Muscular Atrophy , Regeneration , Animals , Rabbits , Melitten/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Regeneration/drug effects , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Male
4.
Molecules ; 29(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38792078

ABSTRACT

Disuse muscle atrophy (DMA) is a significant healthcare challenge characterized by progressive loss of muscle mass and function resulting from prolonged inactivity. The development of effective strategies for muscle recovery is essential. In this study, we established a DMA mouse model through hindlimb suspension to evaluate the therapeutic potential of lactate in alleviating the detrimental effects on the gastrocnemius muscle. Using NMR-based metabolomic analysis, we investigated the metabolic changes in DMA-injured gastrocnemius muscles compared to controls and evaluated the beneficial effects of lactate treatment. Our results show that lactate significantly reduced muscle mass loss and improved muscle function by downregulating Murf1 expression, decreasing protein ubiquitination and hydrolysis, and increasing myosin heavy chain levels. Crucially, lactate corrected perturbations in four key metabolic pathways in the DMA gastrocnemius: the biosynthesis of phenylalanine, tyrosine, and tryptophan; phenylalanine metabolism; histidine metabolism; and arginine and proline metabolism. In addition to phenylalanine-related pathways, lactate also plays a role in regulating branched-chain amino acid metabolism and energy metabolism. Notably, lactate treatment normalized the levels of eight essential metabolites in DMA mice, underscoring its potential as a therapeutic agent against the consequences of prolonged inactivity and muscle wasting. This study not only advances our understanding of the therapeutic benefits of lactate but also provides a foundation for novel treatment approaches aimed at metabolic restoration and muscle recovery in conditions of muscle wasting.


Subject(s)
Lactic Acid , Metabolomics , Muscle, Skeletal , Animals , Mice , Metabolomics/methods , Lactic Acid/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/drug effects , Muscular Atrophy/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/drug therapy , Muscular Atrophy/pathology , Disease Models, Animal , Magnetic Resonance Spectroscopy , Male , Muscle Proteins/metabolism , Muscular Disorders, Atrophic/metabolism , Muscular Disorders, Atrophic/drug therapy , Muscular Disorders, Atrophic/pathology , Ubiquitin-Protein Ligases/metabolism , Metabolome/drug effects , Hindlimb Suspension , Tripartite Motif Proteins/metabolism , Mice, Inbred C57BL , Myosin Heavy Chains/metabolism
5.
Mol Med Rep ; 30(1)2024 Jul.
Article in English | MEDLINE | ID: mdl-38757344

ABSTRACT

Muscle atrophy is a debilitating condition with various causes; while aging is one of these causes, reduced engagement in routine muscle­strengthening activities also markedly contributes to muscle loss. Although extensive research has been conducted on microRNAs (miRNAs/miRs) and their associations with muscle atrophy, the roles played by miRNA precursors remain underexplored. The present study detected the upregulation of the miR­206 precursor in cell­free (cf)RNA from the plasma of patients at risk of sarcopenia, and in cfRNAs from the muscles of mice subjected to muscle atrophy. Additionally, a decline in the levels of the miR­6516 precursor was observed in mice with muscle atrophy. The administration of mimic­miR­6516 to mice immobilized due to injury inhibited muscle atrophy by targeting and inhibiting cyclin­dependent kinase inhibitor 1b (Cdkn1b). Based on these results, the miR­206 precursor appears to be a potential biomarker of muscle atrophy, whereas miR­6516 shows promise as a therapeutic target to alleviate muscle deterioration in patients with muscle disuse and atrophy.


Subject(s)
MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Mice , Humans , Male , Female , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Disease Models, Animal , Middle Aged , Aged , Muscular Disorders, Atrophic/genetics , Muscular Disorders, Atrophic/metabolism , Muscular Disorders, Atrophic/pathology , Muscular Disorders, Atrophic/therapy , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Biomarkers , Sarcopenia/metabolism , Sarcopenia/genetics , Sarcopenia/pathology , Sarcopenia/therapy , Adult
6.
ACS Nano ; 18(22): 14427-14440, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38776414

ABSTRACT

Muscle atrophy resulting from peripheral nerve injury (PNI) poses a threat to a patient's mobility and sensitivity. However, an effective method to inhibit muscle atrophy following PNI remains elusive. Drawing inspiration from the sea cucumber, we have integrated microneedles (MNs) and microchannel technology into nerve guidance conduits (NGCs) to develop bionic microneedle NGCs (MNGCs) that emulate the structure and piezoelectric function of sea cucumbers. Morphologically, MNGCs feature an outer surface with outward-pointing needle tips capable of applying electrical stimulation to denervated muscles. Simultaneously, the interior contains microchannels designed to guide the migration of Schwann cells (SCs). Physiologically, the incorporation of conductive reduced graphene oxide and piezoelectric zinc oxide nanoparticles into the polycaprolactone scaffold enhances conductivity and piezoelectric properties, facilitating SCs' migration, myelin regeneration, axon growth, and the restoration of neuromuscular function. These combined effects ultimately lead to the inhibition of muscle atrophy and the restoration of nerve function. Consequently, the concept of the synergistic effect of inhibiting muscle atrophy and promoting nerve regeneration has the capacity to transform the traditional approach to PNI repair and find broad applications in PNI repair.


Subject(s)
Muscular Atrophy , Needles , Nerve Regeneration , Sea Cucumbers , Animals , Nerve Regeneration/drug effects , Muscular Atrophy/prevention & control , Muscular Atrophy/pathology , Sea Cucumbers/chemistry , Schwann Cells , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/therapy , Graphite/chemistry , Rats , Polyesters/chemistry , Rats, Sprague-Dawley , Mice
7.
Proc Natl Acad Sci U S A ; 121(22): e2405123121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38781208

ABSTRACT

Mitochondria play a central role in muscle metabolism and function. A unique family of iron-sulfur proteins, termed CDGSH Iron Sulfur Domain-containing (CISD/NEET) proteins, support mitochondrial function in skeletal muscles. The abundance of these proteins declines during aging leading to muscle degeneration. Although the function of the outer mitochondrial CISD/NEET proteins, CISD1/mitoNEET and CISD2/NAF-1, has been defined in skeletal muscle cells, the role of the inner mitochondrial CISD protein, CISD3/MiNT, is currently unknown. Here, we show that CISD3 deficiency in mice results in muscle atrophy that shares proteomic features with Duchenne muscular dystrophy. We further reveal that CISD3 deficiency impairs the function and structure of skeletal muscles, as well as their mitochondria, and that CISD3 interacts with, and donates its [2Fe-2S] clusters to, complex I respiratory chain subunit NADH Ubiquinone Oxidoreductase Core Subunit V2 (NDUFV2). Using coevolutionary and structural computational tools, we model a CISD3-NDUFV2 complex with proximal coevolving residue interactions conducive of [2Fe-2S] cluster transfer reactions, placing the clusters of the two proteins 10 to 16 Å apart. Taken together, our findings reveal that CISD3/MiNT is important for supporting the biogenesis and function of complex I, essential for muscle maintenance and function. Interventions that target CISD3 could therefore impact different muscle degeneration syndromes, aging, and related conditions.


Subject(s)
Electron Transport Complex I , Mitochondrial Proteins , Muscle, Skeletal , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mice , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondria/metabolism , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Mice, Knockout , Mitochondria, Muscle/metabolism , Humans , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/genetics
8.
PLoS One ; 19(5): e0304380, 2024.
Article in English | MEDLINE | ID: mdl-38820523

ABSTRACT

Skeletal muscle atrophy is characterized by a decrease in muscle mass and strength caused by an imbalance in protein synthesis and degradation. This process naturally occurs upon reduced or absent physical activity, often related to illness, forced bed rest, or unhealthy lifestyles. Currently, no treatment is available for atrophy, and it can only be prevented by overloading exercise, causing severe problems for patients who cannot exercise due to chronic diseases, disabilities, or being bedridden. The two murine models commonly used to induce muscle atrophy are hindlimb suspension and ankle joint immobilization, both of which come with criticalities. The lack of treatments and the relevance of this atrophic process require a unilateral, safe, and robust model to induce muscle atrophy. In this work, we designed and developed a 3D-printed cast to be used for the study of disuse skeletal muscle atrophy. Applying two halves of the cast is non-invasive, producing little to no swelling or skin damage. The application of the cast induces, in 2-weeks immobilized leg, the activation of atrophy-related genes, causing a muscle weight loss up to 25% in the gastrocnemius muscle, and 31% in the soleus muscle of the immobilized leg compared to the control leg. The cross-sectional area of the fibers is decreased by 31% and 34% respectively, with a peculiar effect on fiber types. In the immobilized gastrocnemius, absolute muscle force is reduced by 38%, while normalized force is reduced by 16%. The contralateral leg did not show signs of overload or hypertrophy when compared to free roaming littermates, offering a good internal control over the immobilized limb. Upon removing the cast, the mice effectively recovered mass and force in 3 weeks.


Subject(s)
Disease Models, Animal , Muscle, Skeletal , Muscular Atrophy , Printing, Three-Dimensional , Animals , Muscle, Skeletal/pathology , Mice , Muscular Atrophy/pathology , Muscular Atrophy/etiology , Muscular Atrophy/therapy , Male , Muscular Disorders, Atrophic/pathology , Muscular Disorders, Atrophic/therapy , Hindlimb Suspension/adverse effects , Mice, Inbred C57BL
9.
Sci Rep ; 14(1): 8176, 2024 04 08.
Article in English | MEDLINE | ID: mdl-38589505

ABSTRACT

Knee osteoarthritis (KOA) usually leads to quadriceps femoris atrophy, which in turn can further aggravate the progression of KOA. Curcumin (CUR) has anti-inflammatory and antioxidant effects and has been shown to be a protective agent for skeletal muscle. CUR has been shown to have a protective effect on skeletal muscle. However, there are no studies related to whether CUR improves KOA-induced quadriceps femoris muscle atrophy. We established a model of KOA in rats. Rats in the experimental group were fed CUR for 5 weeks. Changes in autophagy levels, reactive oxygen species (ROS) levels, and changes in the expression of the Sirutin3 (SIRT3)-superoxide dismutase 2 (SOD2) pathway were detected in the quadriceps femoris muscle of rats. KOA led to quadriceps femoris muscle atrophy, in which autophagy was induced and ROS levels were increased. CUR increased SIRT3 expression, decreased SOD2 acetylation and ROS levels, inhibited the over-activation of autophagy, thereby alleviating quadriceps femoris muscle atrophy and improving KOA. CUR has a protective effect against quadriceps femoris muscle atrophy, and KOA is alleviated after improvement of quadriceps femoris muscle atrophy, with the possible mechanism being the reduction of ROS-induced autophagy via the SIRT3-SOD2 pathway.


Subject(s)
Curcumin , Osteoarthritis, Knee , Sirtuin 3 , Superoxide Dismutase , Rats , Animals , Reactive Oxygen Species/metabolism , Osteoarthritis, Knee/pathology , Quadriceps Muscle/metabolism , Sirtuin 3/metabolism , Curcumin/pharmacology , Muscular Atrophy/drug therapy , Muscular Atrophy/pathology , Autophagy , Signal Transduction
10.
PLoS One ; 19(4): e0302194, 2024.
Article in English | MEDLINE | ID: mdl-38630690

ABSTRACT

Cancer cachexia causes skeletal muscle atrophy, impacting the treatment and prognosis of patients with advanced cancer, but no treatment has yet been established to control cancer cachexia. We demonstrated that transcutaneous application of carbon dioxide (CO2) could improve local blood flow and reduce skeletal muscle atrophy in a fracture model. However, the effects of transcutaneous application of CO2 in cancer-bearing conditions are not yet known. In this study, we calculated fat-free body mass (FFM), defined as the skeletal muscle mass, and evaluated the expression of muscle atrophy markers and uncoupling protein markers as well as the cross-sectional area (CSA) to investigate whether transcutaneous application of CO2 to skeletal muscle could suppress skeletal muscle atrophy in cancer-bearing mice. Human oral squamous cell carcinoma was transplanted subcutaneously into the upper dorsal region of nude mice, and 1 week later, CO2 gas was applied to the legs twice a week for 4 weeks and FFM was calculated by bioimpedance spectroscopy. After the experiment concluded, the quadriceps were extracted, and muscle atrophy markers (muscle atrophy F-box protein (MAFbx), muscle RING-finger protein 1 (MuRF-1)) and uncoupling protein markers (uncoupling protein 2 (UCP2) and uncoupling protein 3 (UCP3)) were evaluated by real-time polymerase chain reaction and immunohistochemical staining, and CSA by hematoxylin and eosin staining. The CO2-treated group exhibited significant mRNA and protein expression inhibition of the four markers. Furthermore, immunohistochemical staining showed decreased MAFbx, MuRF-1, UCP2, and UCP3 in the CO2-treated group. In fact, the CSA in hematoxylin and eosin staining and the FFM revealed significant suppression of skeletal muscle atrophy in the CO2-treated group. We suggest that transcutaneous application of CO2 to skeletal muscle suppresses skeletal muscle atrophy in a mouse model of oral squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Mice , Animals , Carbon Dioxide/metabolism , Cachexia/etiology , Carcinoma, Squamous Cell/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Mice, Nude , Eosine Yellowish-(YS) , Hematoxylin , Mouth Neoplasms/pathology , Muscular Atrophy/pathology , Muscle, Skeletal/metabolism , Head and Neck Neoplasms/pathology , Mitochondrial Uncoupling Proteins/metabolism
11.
PLoS One ; 19(4): e0301726, 2024.
Article in English | MEDLINE | ID: mdl-38574091

ABSTRACT

OBJECTIVES: Evaluation of spinal muscle morphology may be critical because of its impact on segmental stability and control of the lumbar spine in the subset of patients with clinical lumbar segmental instability (LSI). The purpose of this study was to compare lumbar muscle morphology in CNLBP patients with clinical LSI, CNLBP patients without clinical LSI. METHODS: This case-control study included 30 patients with CNLBP (15 with clinical LSI and 15 without clinical LSI) and 15 subjects without LBP. Axial magnetic resonance images from the L2 to S1 lumbar levels were used to evaluate the morphology of the lumbar muscles. RESULTS: A significant increase in the muscle-to-fat infiltration index and a significant decrease in the relative muscle cross-sectional area (rmCSA) of the multifidus muscle at the L3-L4 to L5-S1 levels were observed in both CNLBP groups compared to the control group (p<0.05). The mean erector spinae mean rmCSA was significantly greater in the clinical LSI group compared to the control group (SMD = 0.853, 95% CI = 0.105 to -1.6, P = 0.044) and also compared to the CNLBP without clinical LSI (SMD = 0.894, 95% CI = -1.645 to -0.144, P = 0.030) at the L4-L5 level. CONCLUSIONS: The atrophic changes of the multifidus muscle, in CNLBP patients with or without clinical LSI was observed. However, hypertrophic changes of the erector spinae muscle at the L4-L5 lumbar level were observed only in the clinical LSI group. Psaos major did not show significant atrophic or hypertrophic changes.


Subject(s)
Joint Instability , Low Back Pain , Spinal Diseases , Humans , Low Back Pain/diagnostic imaging , Low Back Pain/pathology , Case-Control Studies , Lumbar Vertebrae/diagnostic imaging , Lumbar Vertebrae/pathology , Spinal Diseases/pathology , Muscular Atrophy/pathology , Magnetic Resonance Imaging , Paraspinal Muscles/anatomy & histology , Joint Instability/diagnostic imaging
12.
Nature ; 629(8010): 154-164, 2024 May.
Article in English | MEDLINE | ID: mdl-38649488

ABSTRACT

Muscle atrophy and functional decline (sarcopenia) are common manifestations of frailty and are critical contributors to morbidity and mortality in older people1. Deciphering the molecular mechanisms underlying sarcopenia has major implications for understanding human ageing2. Yet, progress has been slow, partly due to the difficulties of characterizing skeletal muscle niche heterogeneity (whereby myofibres are the most abundant) and obtaining well-characterized human samples3,4. Here we generate a single-cell/single-nucleus transcriptomic and chromatin accessibility map of human limb skeletal muscles encompassing over 387,000 cells/nuclei from individuals aged 15 to 99 years with distinct fitness and frailty levels. We describe how cell populations change during ageing, including the emergence of new populations in older people, and the cell-specific and multicellular network features (at the transcriptomic and epigenetic levels) associated with these changes. On the basis of cross-comparison with genetic data, we also identify key elements of chromatin architecture that mark susceptibility to sarcopenia. Our study provides a basis for identifying targets in the skeletal muscle that are amenable to medical, pharmacological and lifestyle interventions in late life.


Subject(s)
Aging , Muscle, Skeletal , Single-Cell Analysis , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Young Adult , Aging/genetics , Aging/pathology , Aging/physiology , Cell Nucleus/metabolism , Chromatin/metabolism , Chromatin/genetics , Disease Susceptibility , Epigenesis, Genetic , Frailty/genetics , Frailty/pathology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Sarcopenia/genetics , Sarcopenia/pathology , Transcriptome
13.
Nat Commun ; 15(1): 3563, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670969

ABSTRACT

Synthetic glucocorticoids (GC), such as dexamethasone, are extensively used to treat chronic inflammation and autoimmune disorders. However, long-term treatments are limited by various side effects, including muscle atrophy. GC activities are mediated by the glucocorticoid receptor (GR), that regulates target gene expression in various tissues in association with cell-specific co-regulators. Here we show that GR and the lysine-specific demethylase 1 (LSD1) interact in myofibers of male mice, and that LSD1 connects GR-bound enhancers with NRF1-associated promoters to stimulate target gene expression. In addition, we unravel that LSD1 demethylase activity is required for triggering starvation- and dexamethasone-induced skeletal muscle proteolysis in collaboration with GR. Importantly, inhibition of LSD1 circumvents muscle wasting induced by pharmacological levels of dexamethasone, without affecting their anti-inflammatory activities. Thus, our findings provide mechanistic insights into the muscle-specific GC activities, and highlight the therapeutic potential of targeting GR co-regulators to limit corticotherapy-induced side effects.


Subject(s)
Dexamethasone , Glucocorticoids , Histone Demethylases , Muscle, Skeletal , Muscular Atrophy , Receptors, Glucocorticoid , Animals , Male , Histone Demethylases/metabolism , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/genetics , Glucocorticoids/pharmacology , Dexamethasone/pharmacology , Receptors, Glucocorticoid/metabolism , Mice , Muscular Atrophy/chemically induced , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/drug therapy , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Mice, Inbred C57BL , Gene Expression Regulation/drug effects
14.
Anticancer Res ; 44(5): 1963-1971, 2024 May.
Article in English | MEDLINE | ID: mdl-38677769

ABSTRACT

BACKGROUND/AIM: Cancer cachexia is a wasting syndrome that has a devastating impact on the prognosis of patients with cancer. It is well-documented that pro-inflammatory cytokines are involved in the progression of this disorder. Therefore, this study was conducted to investigate the protective effect of taurine, an essential nonprotein amino acid with great anti-inflammatory properties, in attenuating muscle atrophy induced by cancer. MATERIALS AND METHODS: Conditioned media (CM) derived from T24 human bladder carcinoma cells with or without 5 mM taurine were incubated with human skeletal muscle cells (HSkMCs) and their differentiation was examined. The intracellular reactive oxygen species (ROS), morphology, and the catabolic pathway were monitored. RESULTS: T24-derived CM with high levels of TNF-α and IL-6 caused aberrant ROS accumulation and formation of atrophic myotubes by HSkMCs. In T24 cancer cells, taurine significantly inhibited the production of TNF-α and IL-6. In HSkMCs, taurine increased ROS clearance during differentiation and preserved the myotube differentiation ability impaired by the inflammatory tumor microenvironment. In addition, taurine ameliorated myotube atrophy by regulating the Akt/FoxO1/MuRF1 and MAFbx signaling pathways. CONCLUSION: Taurine rescues cancer-induced atrophy in human skeletal muscle cells by ameliorating the inflammatory tumor microenvironment. Taurine supplementation may be a promising approach for intervening with the progression of cancer cachexia.


Subject(s)
Muscular Atrophy , Reactive Oxygen Species , Taurine , Tumor Microenvironment , Humans , Taurine/pharmacology , Tumor Microenvironment/drug effects , Muscular Atrophy/pathology , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscular Atrophy/etiology , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Cell Differentiation/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/drug effects , Signal Transduction/drug effects , Cachexia/drug therapy , Cachexia/pathology , Cachexia/metabolism , Cachexia/etiology , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Culture Media, Conditioned/pharmacology , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukin-6/metabolism
15.
PLoS One ; 19(4): e0301379, 2024.
Article in English | MEDLINE | ID: mdl-38648220

ABSTRACT

While chemotherapy treatment can be lifesaving, it also has adverse effects that negatively impact the quality of life. To investigate the effects of doxorubicin chemotherapy on body weight loss, strength and muscle mass loss, and physical function impairments, all key markers of cachexia, sarcopenia, and frailty. Seventeen C57/BL/6 mice were allocated into groups. 1) Control (n = 7): mice were exposed to intraperitoneal (i.p.) injections of saline solution. 2) Dox (n = 10): mice were exposed to doxorubicin chemotherapy cycles (total dose of 18 mg/kg divided over 15 days). The body weight loss and decreased food intake were monitored to assess cachexia. To assess sarcopenia, we measured muscle strength loss using a traction method and evaluated muscle atrophy through histology of the gastrocnemius muscle. To evaluate physical function impairments and assess frailty, we employed the open field test to measure exploratory capacity. Doxorubicin administration led to the development of cachexia, as evidenced by a significant body weight loss (13%) and a substantial decrease in food intake (34%) over a 15-day period. Furthermore, 90% of the mice treated with doxorubicin exhibited sarcopenia, characterized by a 20% reduction in traction strength (p<0,05), a 10% decrease in muscle mass, and a 33% reduction in locomotor activity. Importantly, all mice subjected to doxorubicin treatment were considered frail based on the evaluation of their overall condition and functional impairments. The proposed model holds significant characteristics of human chemotherapy treatment and can be useful to understand the intricate relationship between chemotherapy, cachexia, sarcopenia, and frailty.


Subject(s)
Cachexia , Doxorubicin , Frailty , Mice, Inbred C57BL , Muscle, Skeletal , Sarcopenia , Animals , Doxorubicin/adverse effects , Cachexia/chemically induced , Cachexia/etiology , Sarcopenia/chemically induced , Sarcopenia/pathology , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Male , Muscle Strength/drug effects , Muscular Atrophy/chemically induced , Muscular Atrophy/pathology , Weight Loss/drug effects , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity
16.
Apoptosis ; 29(5-6): 663-680, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38598070

ABSTRACT

Cancer cachexia-associated muscle wasting as a multifactorial wasting syndrome, is an important factor affecting the long-term survival rate of tumor patients. Photobiomodulation therapy (PBMT) has emerged as a promising tool to cure and prevent many diseases. However, the effect of PBMT on skeletal muscle atrophy during cancer progression has not been fully demonstrated yet. Here, we found PBMT alleviated the atrophy of myotube diameter induced by cancer cells in vitro, and prevented cancer-associated muscle atrophy in mice bearing tumor. Mechanistically, the alleviation of muscle wasting by PBMT was found to be involved in inhibiting E3 ubiquitin ligases MAFbx and MuRF-1. In addition, transcriptomic analysis using RNA-seq and GSEA revealed that PI3K/AKT pathway might be involved in PBMT-prevented muscle cachexia. Next, we showed the protective effect of PBMT against muscle cachexia was totally blocked by AKT inhibitor in vitro and in vivo. Moreover, PBMT-activated AKT promoted FoxO3a phosphorylation and thus inhibiting the nucleus entry of FoxO3a. Lastly, in cisplatin-treated muscle cachexia model, PBMT had also been shown to ameliorate muscle atrophy through enhancing PI3K/AKT pathway to suppress MAFbx and MuRF-1 expression. These novel findings revealed that PBMT could be a promising therapeutic approach in treating muscle cachexia induced by cancer.


Subject(s)
Cachexia , Forkhead Box Protein O3 , Low-Level Light Therapy , Muscular Atrophy , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Cachexia/etiology , Cachexia/metabolism , Cachexia/genetics , Cachexia/pathology , Forkhead Box Protein O3/metabolism , Forkhead Box Protein O3/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Mice , Muscular Atrophy/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Humans , Neoplasms/radiotherapy , Neoplasms/complications , Neoplasms/metabolism , Male , Cell Line, Tumor , Muscle Proteins/metabolism , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/radiation effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
17.
Cell Mol Biol Lett ; 29(1): 59, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654156

ABSTRACT

Skeletal muscle is the largest metabolic organ of the human body. Maintaining the best quality control and functional integrity of mitochondria is essential for the health of skeletal muscle. However, mitochondrial dysfunction characterized by mitochondrial dynamic imbalance and mitophagy disruption can lead to varying degrees of muscle atrophy, but the underlying mechanism of action is still unclear. Although mitochondrial dynamics and mitophagy are two different mitochondrial quality control mechanisms, a large amount of evidence has indicated that they are interrelated and mutually regulated. The former maintains the balance of the mitochondrial network, eliminates damaged or aged mitochondria, and enables cells to survive normally. The latter degrades damaged or aged mitochondria through the lysosomal pathway, ensuring cellular functional health and metabolic homeostasis. Skeletal muscle atrophy is considered an urgent global health issue. Understanding and gaining knowledge about muscle atrophy caused by mitochondrial dysfunction, particularly focusing on mitochondrial dynamics and mitochondrial autophagy, can greatly contribute to the prevention and treatment of muscle atrophy. In this review, we critically summarize the recent research progress on mitochondrial dynamics and mitophagy in skeletal muscle atrophy, and expound on the intrinsic molecular mechanism of skeletal muscle atrophy caused by mitochondrial dynamics and mitophagy. Importantly, we emphasize the potential of targeting mitochondrial dynamics and mitophagy as therapeutic strategies for the prevention and treatment of muscle atrophy, including pharmacological treatment and exercise therapy, and summarize effective methods for the treatment of skeletal muscle atrophy.


Subject(s)
Mitochondrial Dynamics , Mitophagy , Muscle, Skeletal , Muscular Atrophy , Humans , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Atrophy/therapy , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Animals , Mitochondria/metabolism , Mitochondria/pathology
18.
Biomed Pharmacother ; 174: 116517, 2024 May.
Article in English | MEDLINE | ID: mdl-38574619

ABSTRACT

Age-associated osteosarcopenia is an unresolved syndrome characterized by the concomitant loss of bone (osteopenia) and skeletal muscle (sarcopenia) tissues increasing falls, immobility, morbidity, and mortality. Unbalanced resorption of bone in the remodeling process and excessive protein breakdown, especially fast type II myosin heavy chain (MyHC-II) isoform and myofiber metabolic shift, are the leading causes of bone and muscle deterioration in the elderly, respectively. Equisetum arvense (EQ) is a plant traditionally recommended for many pathological conditions due to its anti-inflammatory properties. Thus, considering that a chronic low-grade inflammatory state predisposes to both osteoporosis and sarcopenia, we tested a standardized hydroalcoholic extract of EQ in in vitro models of muscle atrophy [C2C12 myotubes treated with proinflammatory cytokines (TNFα/IFNγ), excess glucocorticoids (dexamethasone), or the osteokine, receptor activator of nuclear factor kappa-B ligand (RANKL)] and osteoclastogenesis (RAW 264.7 cells treated with RANKL). We found that EQ counteracted myotube atrophy, blunting the activity of several pathways depending on the applied stimulus, and reduced osteoclast formation and activity. By in silico target fishing, IKKB-dependent nuclear factor kappa-B (NF-κB) inhibition emerges as a potential common mechanism underlying EQ's anti-atrophic effects. Consumption of EQ (500 mg/kg/day) by pre-geriatric C57BL/6 mice for 3 months translated into: i) maintenance of muscle mass and performance; ii) restrained myofiber oxidative shift; iii) slowed down age-related modifications in osteoporotic bone, significantly preserving trabecular connectivity density; iv) reduced muscle- and spleen-related inflammation. EQ can preserve muscle functionality and bone remodeling during aging, potentially valuable as a natural treatment for osteosarcopenia.


Subject(s)
Equisetum , Plant Extracts , Sarcopenia , Animals , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Mice , Sarcopenia/drug therapy , Sarcopenia/pathology , RAW 264.7 Cells , Equisetum/chemistry , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/metabolism , Aging/drug effects , Aging/pathology , Muscular Atrophy/drug therapy , Muscular Atrophy/pathology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , RANK Ligand/metabolism , NF-kappa B/metabolism , Osteogenesis/drug effects , Anti-Inflammatory Agents/pharmacology
19.
J Vet Intern Med ; 38(3): 1334-1344, 2024.
Article in English | MEDLINE | ID: mdl-38622799

ABSTRACT

BACKGROUND: Muscle atrophy and intramuscular fatty infiltration, as well as their association with prognosis, have not been quantified in dogs with spontaneous hypercortisolism (HC). OBJECTIVE: To quantitatively evaluate muscle atrophy and IM fatty infiltration in dogs with HC and determine their prognostic impact. ANIMALS: Fifty-three dogs with HC and 66 control dogs without HC. METHODS: Retrospective cohort study. Medical records and computed tomography images obtained between 2014 and 2021 were evaluated. Kaplan-Meier curves and log-rank tests were used to analyze the effect of muscle atrophy and IM fatty infiltration on the prognosis of dogs with HC. RESULTS: Dogs with HC showed lower visually measured cross-sectional area (VCSA) and cross-sectional area based on attenuation (HCSA) than control dogs (median [interquartile range {IQR}]: 50.3 mm2/mm [36.2-67.8] vs 66.7 mm2/mm [48.0-85.9]; P < .001; 30.4 mm2/mm [13.7-57.2] vs 54.8 mm2/mm [39.7-71.5]; P < .001, respectively). Dogs with HC had lower epaxial muscle attenuation (L3HU) than control dogs (median [IQR]: 21.2 Hounsfield [HU] [12.4-28.2] vs 33.2 HU [22.6-43.6]; P < .001). Dogs with HC with lower HCSA or L3HU had shorter survival (median [IQR]: 670 days [222-673] vs 949 days [788-1074], P < .01; 523 days [132-670] vs 949 days [756-1074], P < .01, respectively) but not lower VCSA (median [IQR]: 673 days [132-788] vs 949 days [523 to not applicable]; P = .30). CONCLUSION AND CLINICAL IMPORTANCE: Hypercortisolism in dogs causes muscle atrophy and IM fatty infiltration and is associated with poor prognosis.


Subject(s)
Cushing Syndrome , Dog Diseases , Muscle, Skeletal , Muscular Atrophy , Animals , Dogs , Dog Diseases/pathology , Retrospective Studies , Male , Female , Prognosis , Cushing Syndrome/veterinary , Cushing Syndrome/pathology , Muscular Atrophy/veterinary , Muscular Atrophy/pathology , Muscle, Skeletal/pathology , Adipose Tissue/pathology , Tomography, X-Ray Computed/veterinary , Cohort Studies
20.
Cell Rep Med ; 5(4): 101498, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38569555

ABSTRACT

Progressive weakness and muscle loss are associated with multiple chronic conditions, including muscular dystrophy and cancer. Cancer-associated cachexia, characterized by dramatic weight loss and fatigue, leads to reduced quality of life and poor survival. Inflammatory cytokines have been implicated in muscle atrophy; however, available anticytokine therapies failed to prevent muscle wasting in cancer patients. Here, we show that oncostatin M (OSM) is a potent inducer of muscle atrophy. OSM triggers cellular atrophy in primary myotubes using the JAK/STAT3 pathway. Identification of OSM targets by RNA sequencing reveals the induction of various muscle atrophy-related genes, including Atrogin1. OSM overexpression in mice causes muscle wasting, whereas muscle-specific deletion of the OSM receptor (OSMR) and the neutralization of circulating OSM preserves muscle mass and function in tumor-bearing mice. Our results indicate that activated OSM/OSMR signaling drives muscle atrophy, and the therapeutic targeting of this pathway may be useful in preventing muscle wasting.


Subject(s)
Neoplasms , Oncostatin M , Quality of Life , Animals , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Neoplasms/pathology , Oncostatin M/genetics , Oncostatin M/metabolism , Oncostatin M/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...