Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Biophys Res Commun ; 586: 157-162, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34847441

ABSTRACT

Sarcopenia is the age-related loss of muscle mass and function and no pharmacological medication has been approved for its treatment. We established an atrogin-1/MAFbx promoter assay to find drug candidates that inhibit myotube atrophy. Alverine citrate (AC) was identified using high-throughput screening of an existing drug library. AC is an established medicine for stomach and intestinal spasms. AC treatment increased myotube diameter and inhibited atrophy signals induced by either C26-conditioned medium or dexamethasone in cultured C2C12 myoblasts. AC also enhanced myoblast fusion through the upregulation of fusion-related genes during C2C12 myoblast differentiation. Oral administration of AC improves muscle mass and physical performance in aged mice, as well as hindlimb-disused mice. Taken together, our data suggest that AC may be a novel therapeutic candidate for improving muscle weakness, including sarcopenia.


Subject(s)
Aging/genetics , Cell Differentiation/drug effects , Muscular Atrophy/prevention & control , Parasympatholytics/pharmacology , Propylamines/pharmacology , Sarcopenia/prevention & control , Aging/metabolism , Animals , Biomarkers/metabolism , Cadherins/genetics , Cadherins/metabolism , Caveolin 3/genetics , Caveolin 3/metabolism , Cell Line , Dexamethasone/pharmacology , Disease Models, Animal , Gene Expression , High-Throughput Screening Assays , Immobilization , Integrin beta1/genetics , Integrin beta1/metabolism , Mice , Mice, Inbred C57BL , Muscle Development/genetics , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , Sarcopenia/genetics , Sarcopenia/metabolism , Sarcopenia/pathology
2.
Nat Commun ; 11(1): 5661, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33168829

ABSTRACT

Sarcopenia is characterized by decreased skeletal muscle mass and function with age. Aged muscles have altered lipid compositions; however, the role and regulation of lipids are unknown. Here we report that FABP3 is upregulated in aged skeletal muscles, disrupting homeostasis via lipid remodeling. Lipidomic analyses reveal that FABP3 overexpression in young muscles alters the membrane lipid composition to that of aged muscle by decreasing polyunsaturated phospholipid acyl chains, while increasing sphingomyelin and lysophosphatidylcholine. FABP3-dependent membrane lipid remodeling causes ER stress via the PERK-eIF2α pathway and inhibits protein synthesis, limiting muscle recovery after immobilization. FABP3 knockdown induces a young-like lipid composition in aged muscles, reduces ER stress, and improves protein synthesis and muscle recovery. Further, FABP3 reduces membrane fluidity and knockdown increases fluidity in vitro, potentially causing ER stress. Therefore, FABP3 drives membrane lipid composition-mediated ER stress to regulate muscle homeostasis during aging and is a valuable target for sarcopenia.


Subject(s)
Aging/physiology , Endoplasmic Reticulum Stress/physiology , Fatty Acid Binding Protein 3/metabolism , Membrane Lipids/metabolism , Muscle, Skeletal/metabolism , Animals , Cell Line , Eukaryotic Initiation Factor-2/metabolism , Fatty Acid Binding Protein 3/genetics , Female , Gene Knockdown Techniques , Lipidomics , Membrane Fluidity , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Myoblasts/pathology , Myoblasts/physiology , Phospholipids/metabolism , Protein Serine-Threonine Kinases , Sarcopenia , Up-Regulation
3.
J Cachexia Sarcopenia Muscle ; 11(5): 1336-1350, 2020 10.
Article in English | MEDLINE | ID: mdl-32495509

ABSTRACT

BACKGROUND: The microRNAs (miRNAs) down-regulated in aged mouse skeletal muscle were mainly clustered within the delta-like homologue 1 and the type III iodothyronine deiodinase (Dlk1-Dio3) genomic region. Although clustered miRNAs are coexpressed and regulate multiple targets in a specific signalling pathway, the function of miRNAs in the Dlk1-Dio3 cluster in muscle aging is largely unknown. We aimed to ascertain whether these miRNAs play a common role to regulate age-related muscle atrophy. METHODS: To examine anti-atrophic effect of miRNAs, we individually transfected 42 miRNA mimics in fully differentiated myotubes and analysed their diameters. The luciferase reporter assay using target 3' untranslated region (UTR) and RNA pull-down assay were employed to ascertain the target predicted by the TargetScan algorithm. To investigate the therapeutic potential of the miRNAs in vivo, we generated adeno-associated virus (AAV) serotype 9 expressing green fluorescent protein (GFP) (AAV9-GFP) bearing miR-376c-3p and infected it into the tibialis anterior muscle of old mice. We performed morphometric analysis and measured ex vivo isometric force using a force transducer. Human gluteus maximus muscle tissues (ages ranging from 25 to 80 years) were used to investigate expression levels of the conserved miRNAs in the Dlk1-Dio3 cluster. RESULTS: We found that the majority of miRNAs (33 out of 42 tested) in the cluster induced anti-atrophic phenotypes in fully differentiated myotubes with increasing their diameters. Eighteen of these miRNAs, eight of which are conserved in humans, harboured predicted binding sites in the 3' UTR of muscle atrophy gene-1 (Atrogin-1) encoding a muscle-specific E3 ligase. Direct interactions were identified between these miRNAs and the 3' UTR of Atrogin-1, leading to repression of Atrogin-1 and thereby induction of eIF3f protein content, in both human and mouse skeletal muscle cells. Intramuscular delivery of AAV9 expressing miR-376c-3p, one of the most effective miRNAs in myotube thickening, dramatically ameliorated skeletal muscle atrophy and improved muscle function, including isometric force, twitch force, and fatigue resistance in old mice. Consistent with our findings in mice, the expression of miRNAs in the cluster was significantly down-regulated in human muscle from individuals > 50 years old. CONCLUSIONS: Our study suggests that genetic intervention using a muscle-directed miRNA delivery system has therapeutic efficacy in preventing Atrogin-1-mediated muscle atrophy in sarcopenia.


Subject(s)
MicroRNAs , Animals , Calcium-Binding Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins , Iodide Peroxidase , Membrane Proteins , Mice , MicroRNAs/genetics , Muscle Fibers, Skeletal , Muscular Atrophy/genetics , Muscular Atrophy/therapy
4.
Sci Rep ; 8(1): 8574, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29872072

ABSTRACT

Sarcopenia is a gradual loss of skeletal muscle mass and function with aging. Given that sarcopenia has been recognized as a disease entity, effective molecular biomarkers for early diagnosis are required. We recruited 46 normal subjects and 50 patients with moderate sarcopenia aged 60 years and older. Sarcopenia was clinically identified on the basis of the appendicular skeletal muscle index by applying cutoff values derived from the Asian Working Group for Sarcopenia. The serum levels of 21 potential biomarkers were analyzed and statistically examined. Interleukin 6, secreted protein acidic and rich in cysteine, macrophage migration inhibitory factor, and insulin-like growth factor 1 levels differed significantly between the normal and sarcopenia groups. However, in each case, the area under the receiver operating characteristics curve (AUC) was <0.7. Subsequent combination of the measurements of these biomarkers into a single risk score based on logistic regression coefficients enhanced the accuracy of diagnosis, yielding an AUC value of 0.763. The best cutoff value of 1.529 had 70.0% sensitivity and 78.3% specificity (95% CI = 2.80-21.69, p < 0.0001). Combined use of the selected biomarkers provides higher diagnostic accuracy than individual biomarkers, and may be effectively utilized for early diagnosis and prognosis of sarcopenia.


Subject(s)
Biomarkers/blood , Early Diagnosis , Sarcopenia/blood , Sarcopenia/diagnosis , Aged , Aged, 80 and over , Female , Humans , Insulin-Like Growth Factor I/metabolism , Interleukin-6/blood , Logistic Models , Macrophage Migration-Inhibitory Factors/blood , Male , Osteonectin/blood , Sensitivity and Specificity
5.
Aging (Albany NY) ; 8(9): 2062-2080, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27658230

ABSTRACT

Skeletal muscle mass and power decrease with age, leading to impairment of mobility and metabolism in the elderly. Ca2+ signaling is crucial for myoblast differentiation as well as muscle contraction through activation of transcription factors and Ca2+-dependent kinases and phosphatases. Ca2+ channels, such as dihydropyridine receptor (DHPR), two-pore channel (TPC) and inositol 1,4,5-triphosphate receptor (ITPR), function to maintain Ca2+ homeostasis in myoblasts. Here, we observed a significant decrease in expression of type 1 IP3 receptor (ITPR1), but not types 2 and 3, in aged mice skeletal muscle and isolated myoblasts, compared with those of young mice. ITPR1 knockdown using shRNA-expressing viruses in C2C12 myoblasts and tibialis anterior muscle of mice inhibited myotube formation and muscle regeneration after injury, respectively, a typical phenotype of aged muscle. This aging phenotype was associated with repression of muscle-specific genes and activation of the epidermal growth factor receptor (EGFR)-Ras-extracellular signal-regulated kinase (ERK) pathway. ERK inhibition by U0126 not only induced recovery of myotube formation in old myoblasts but also facilitated muscle regeneration after injury in aged muscle. The conserved decline in ITPR1 expression in aged human skeletal muscle suggests utility as a potential therapeutic target for sarcopenia, which can be treated using ERK inhibition strategies.


Subject(s)
Aging/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Muscle, Skeletal/physiology , Myoblasts/metabolism , Regeneration/physiology , Adult , Age Factors , Aged , Aging/genetics , Animals , Calcium/metabolism , Calcium Signaling/physiology , Cells, Cultured , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Middle Aged , Muscle Development/physiology , Muscle, Skeletal/metabolism , Phenotype , Signal Transduction
6.
Genes Dev ; 29(15): 1605-17, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26215566

ABSTRACT

The myogenic capacity of myoblasts decreases in skeletal muscle with age. In addition to environmental factors, intrinsic factors are important for maintaining the regenerative potential of muscle progenitor cells, but their identities are largely unknown. Here, comparative analysis of microRNA (miRNA) expression profiles in young and old myoblasts uncovered miR-431 as a novel miRNA showing markedly reduced abundance in aged myoblasts. Importantly, elevating miR-431 improved the myogenic capacity of old myoblasts, while inhibiting endogenous miR-431 lowered myogenesis. Bioinformatic and biochemical analyses revealed that miR-431 directly interacted with the 3' untranslated region (UTR) of Smad4 mRNA, which encodes one of the downstream effectors of TGF-ß signaling. In keeping with the low levels of miR-431 in old myoblasts, SMAD4 levels increased in this myoblast population. Interestingly, in an in vivo model of muscle regeneration following cardiotoxin injury, ectopic miR-431 injection greatly improved muscle regeneration and reduced SMAD4 levels. Consistent with the finding that the mouse miR-431 seed sequence in the Smad4 3' UTR is conserved in the human SMAD4 3' UTR, inhibition of miR-431 also repressed the myogenic capacity of human skeletal myoblasts. Taken together, our results suggest that the age-associated miR-431 plays a key role in maintaining the myogenic ability of skeletal muscle with age.


Subject(s)
Cell Differentiation , MicroRNAs/metabolism , Muscle Development/genetics , Muscle, Skeletal/physiology , Myoblasts/cytology , Regeneration/genetics , Smad4 Protein/genetics , 3' Untranslated Regions , Animals , Cell Line , Cellular Senescence , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Muscle, Skeletal/cytology , Protein Binding
7.
J Lipid Res ; 56(2): 294-303, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25548260

ABSTRACT

Nucleoredoxin (NRX) is a member of the thioredoxin family of proteins that controls redox homeostasis in cell. Redox homeostasis is a well-known regulator of cell differentiation into various tissue types. We found that NRX expression levels were higher in white adipose tissue of obese ob/ob mice and increased in the early adipogenic stage of 3T3-L1 preadipocyte differentiation. Knockdown of NRX decreased differentiation of 3T3-L1 cells, whereas overexpression increased differentiation. Adipose tissue-specific NRX transgenic mice showed increases in adipocyte size as well as number compared with WT mice. We further confirmed that the Wingless/int-1 class (Wnt)/ß-catenin pathway was also involved in NRX-promoted adipogenesis, consistent with a previous report showing NRX regulation of this pathway. Genes involved in lipid metabolism were downregulated, whereas inflammatory genes, including those encoding macrophage markers, were significantly upregulated, likely contributing to the obesity in Adipo-NRX mice. Our results therefore suggest that NRX acts as a novel proadipogenic factor and controls obesity in vivo.


Subject(s)
Cell Differentiation/physiology , Nuclear Proteins/metabolism , Oxidoreductases/metabolism , Signal Transduction , beta Catenin/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipogenesis/genetics , Adipogenesis/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Female , Male , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Oxidoreductases/genetics , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology
8.
Proteomics ; 14(1): 121-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24243720

ABSTRACT

Aging is associated with a progressive loss of skeletal muscular function that often leads to progressive disability and loss of independence. Although muscle aging is well documented, the molecular mechanisms of this condition still remain unclear. To gain greater insight into the changes associated with aging of skeletal muscle, we performed quantitative proteomic analyses on young (6 months) and aged (27 months) mouse gastrocnemius muscles using mTRAQ stable isotope mass tags. We identified and quantified a total of 4585 peptides corresponding to 236 proteins (protein probability >0.9). Among them, 33 proteins were more than 1.5-fold upregulated and 20 proteins were more than 1.5-fold downregulated in aged muscle compared with young muscle. An ontological analysis revealed that differentially expressed proteins belonged to distinct functional groups, including ion homeostasis, energy metabolism, protein turnover, and Ca(2+) signaling. Identified proteins included aralar1, ß-enolase, fatty acid-binding protein 3, 3-hydroxyacyl-CoA dehydrogenase (Hadh), F-box protein 22, F-box, and leucine-rich repeat protein 18, voltage-dependent L-type calcium channel subunit beta-1, ryanodine receptor (RyR), and calsequestrin. Ectopic expression of calsequestrin in C2C12 myoblast resulted in decreased activity of nuclear factor of activated T-cells and increased levels of atrogin-1 and MuRF1 E3 ligase, suggesting that these differentially expressed proteins are involved in muscle aging.


Subject(s)
Aging/physiology , Muscle, Skeletal/chemistry , Proteome/analysis , Proteome/physiology , Proteomics/methods , Animals , Biomarkers/analysis , Biomarkers/chemistry , Calsequestrin , Immunoblotting , Isotope Labeling , Mass Spectrometry , Mice , Muscle, Skeletal/metabolism , NFATC Transcription Factors , Proteins/analysis , Proteins/chemistry , Proteome/chemistry
9.
J Biol Chem ; 287(3): 1679-87, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22123818

ABSTRACT

Calsequestrin (CSQ), the major intrasarcoplasmic reticulum calcium storage protein, undergoes dynamic polymerization and depolymerization in a Ca(2+)-dependent manner. However, no direct evidence of CSQ depolymerization in vivo with physiological relevance has been obtained. In the present study, live cell imaging analysis facilitated characterization of the in vivo dynamics of the macromolecular CSQ structure. CSQ2 appeared as speckles in the presence of normal sarcoplasmic reticulum (SR) Ca(2+) that were decondensed upon Ca(2+) depletion. Moreover, CSQ2 decondensation occurred only in the stoichiometric presence of junctin (JNT). When expressed alone, CSQ2 speckles remained unchanged, even after Ca(2+) depletion. FRET analysis revealed constant interactions between CSQ2 and JNT, regardless of the SR Ca(2+) concentration, implying that JNT is an essential component of the CSQ scaffold. In vitro solubility assay, electron microscopy, and atomic force microscopy studies using purified recombinant proteins confirmed Ca(2+) and JNT-dependent disassembly of the CSQ2 polymer. Accordingly, we conclude that reversible polymerization and depolymerization of CSQ are critical in SR Ca(2+) homeostasis.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Calsequestrin/metabolism , Membrane Proteins/metabolism , Mixed Function Oxygenases/metabolism , Muscle Proteins/metabolism , Protein Multimerization/physiology , Sarcoplasmic Reticulum/metabolism , Animals , Calcium-Binding Proteins/genetics , Calsequestrin/genetics , Cell Line , Homeostasis/physiology , Humans , Membrane Proteins/genetics , Mice , Mixed Function Oxygenases/genetics , Muscle Proteins/genetics , Sarcoplasmic Reticulum/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...