Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Am J Physiol Regul Integr Comp Physiol ; 326(3): R254-R265, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38252513

ABSTRACT

Cachexia is a muscle-wasting syndrome commonly observed in patients with cancer, which can significantly worsen clinical outcomes. Because of a global rise in obesity, the coexistence of cachexia in obese individuals poses unique challenges, with the impact of excessive adiposity on cachexia severity and underlying pathophysiology not well defined. Understanding the interplay between cachexia and obesity is crucial for improving diagnosis and treatment strategies for these patients; therefore, the present study examined differences in cachexia between lean and obese mice bearing Lewis lung carcinoma (LLC) tumors. Nine-week-old, male C57Bl6J mice were placed on either a chow or a high-fat diet (HFD) for 9 wk. After the diet intervention, mice were inoculated with LLC or vehicle. Markers of cachexia, such as body and muscle loss, were noted in both chow and HFD groups with tumors. Tumor weight of HFD animals was greater than that of chow. LLC tumors reduced gastrocnemius, plantaris, and soleus mass, regardless of diet. The tibialis anterior and plantaris mass and cross-sectional area of type IIb/x fibers in the gastrocnemius were not different between HFD-chow, HFD-tumor, and chow-tumor. Using RNA sequencing (RNA-seq) of the plantaris muscle from chow-tumor and HFD-tumor groups, we identified ∼400 differentially expressed genes. Bioinformatic analysis identified changes in lipid metabolism, mitochondria, bioenergetics, and proteasome degradation. Atrophy was not greater despite larger tumor burden in animals fed an HFD, and RNA-seq data suggests that partial protection is mediated through differences in mitochondrial function and protein degradation, which may serve as future mechanistic targets.NEW & NOTEWORTHY This study provides timely information on the interaction between obesity and cancer cachexia. Lean and obese animals show signs of cachexia with reduced body weight, adipose tissue, and gastrocnemius muscle mass. There was not significant wasting in the tibialis anterior, plantaris, or fast twitch fibers in the gastrocnemius muscle of obese animals with tumors. RNA-seq analysis reveals that obese tumor bearing animals had differential expression of mitochondria- and degradation-related genes, which may direct future studies in mechanistic research.


Subject(s)
Carcinoma, Lewis Lung , Humans , Male , Animals , Mice , Carcinoma, Lewis Lung/complications , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Cachexia/etiology , Cachexia/metabolism , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Obesity/metabolism , Diet, High-Fat , Lung/pathology
2.
Am J Physiol Cell Physiol ; 325(1): C324-C331, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37335024

ABSTRACT

A gradual decline in skeletal muscle mass and function is closely tied to increased mortality and disease risk during organismal aging. Exercise training is the most effective way to enhance muscle health, but the adaptive response to exercise as well as muscle repair potential is blunted in older individuals. Numerous mechanisms contribute to the loss of muscle mass and plasticity as aging progresses. An emerging body of recent evidence implicates an accumulation of senescent ("zombie") cells in muscle as a contributing factor to the aging phenotype. Senescent cells cannot divide but can release inflammatory factors and create an unfavorable environment for homeostasis and adaptation. On balance, some evidence indicates that cells with senescent characteristics can be beneficial for the muscle adaptive process, specifically at younger ages. Emerging evidence also suggests that multinuclear muscle fibers could become senescent. In this review, we summarize current literature on the prevalence of senescent cells in skeletal muscle and highlight the consequences of senescent cell removal on muscle mass, function, and adaptability. We examine key limitations in the field of senescence specifically in skeletal muscle and identify areas of research that require future investigation.NEW & NOTEWORTHY There is evidence to suggest that senescent "zombie" cells may or may not accrue in aging skeletal muscle. When muscle is perturbed regardless of age, senescent-like cells do appear, and the benefits of removing them could be age-dependent. More work is needed to determine the magnitude of accumulation and source of senescent cells in muscle. Regardless, pharmacological senolytic treatment of aged muscle is beneficial for adaptation.


Subject(s)
Cellular Senescence , Muscle, Skeletal , Cellular Senescence/genetics , Muscle, Skeletal/physiology , Phenotype , Homeostasis
3.
J Physiol ; 601(4): 763-782, 2023 02.
Article in English | MEDLINE | ID: mdl-36533424

ABSTRACT

Exercise promotes functional improvements in aged tissues, but the extent to which it simulates partial molecular reprogramming is unknown. Using transcriptome profiling from (1) a skeletal muscle-specific in vivo Oct3/4, Klf4, Sox2 and Myc (OKSM) reprogramming-factor expression murine model; (2) an in vivo inducible muscle-specific Myc induction murine model; (3) a translatable high-volume hypertrophic exercise training approach in aged mice; and (4) human exercise muscle biopsies, we collectively defined exercise-induced genes that are common to partial reprogramming. Late-life exercise training lowered murine DNA methylation age according to several contemporary muscle-specific clocks. A comparison of the murine soleus transcriptome after late-life exercise training to the soleus transcriptome after OKSM induction revealed an overlapping signature that included higher JunB and Sun1. Also, within this signature, downregulation of specific mitochondrial and muscle-enriched genes was conserved in skeletal muscle of long-term exercise-trained humans; among these was muscle-specific Abra/Stars. Myc is the OKSM factor most induced by exercise in muscle and was elevated following exercise training in aged mice. A pulse of MYC rewired the global soleus muscle methylome, and the transcriptome after a MYC pulse partially recapitulated OKSM induction. A common signature also emerged in the murine MYC-controlled and exercise adaptation transcriptomes, including lower muscle-specific Melusin and reactive oxygen species-associated Romo1. With Myc, OKSM and exercise training in mice, as well habitual exercise in humans, the complex I accessory subunit Ndufb11 was lower; low Ndufb11 is linked to longevity in rodents. Collectively, exercise shares similarities with genetic in vivo partial reprogramming. KEY POINTS: Advances in the last decade related to cellular epigenetic reprogramming (e.g. DNA methylome remodelling) toward a pluripotent state via the Yamanaka transcription factors Oct3/4, Klf4, Sox2 and Myc (OKSM) provide a window into potential mechanisms for combatting the deleterious effects of cellular ageing. Using global gene expression analysis, we compared the effects of in vivo OKSM-mediated partial reprogramming in skeletal muscle fibres of mice to the effects of late-life murine exercise training in muscle. Myc is the Yamanaka factor most induced by exercise in skeletal muscle, and so we compared the MYC-controlled transcriptome in muscle to Yamanaka factor-mediated and exercise adaptation mRNA landscapes in mice and humans. A single pulse of MYC is sufficient to remodel the muscle methylome. We identify partial reprogramming-associated genes that are innately altered by exercise training and conserved in humans, and propose that MYC contributes to some of these responses.


Subject(s)
Aging , Cellular Reprogramming , Exercise , Muscle, Skeletal , Animals , Humans , Mice , Cellular Reprogramming/genetics , Disease Models, Animal , DNA Methylation , Exercise/physiology , Gene Expression Profiling , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Aging/genetics , Aging/physiology
4.
FASEB J ; 36(2): e22155, 2022 02.
Article in English | MEDLINE | ID: mdl-35044708

ABSTRACT

The extracellular matrix (ECM) in skeletal muscle plays an integral role in tissue development, structural support, and force transmission. For successful adaptation to mechanical loading, remodeling processes must occur. In a large cohort of older adults, transcriptomics revealed that genes involved in ECM remodeling, including matrix metalloproteinase 14 (MMP14), were the most upregulated following 14 weeks of progressive resistance exercise training (PRT). Using single-cell RNA-seq, we identified macrophages as a source of Mmp14 in muscle following a hypertrophic exercise stimulus in mice. In vitro contractile activity in myotubes revealed that the gene encoding cytokine leukemia inhibitory factor (LIF) is robustly upregulated and can stimulate Mmp14 expression in macrophages. Functional experiments confirmed that modulation of this muscle cell-macrophage axis facilitated Type I collagen turnover. Finally, changes in LIF expression were significantly correlated with MMP14 expression in humans following 14 weeks of PRT. Our experiments reveal a mechanism whereby muscle fibers influence macrophage behavior to promote ECM remodeling in response to mechanical loading.


Subject(s)
Extracellular Matrix/metabolism , Leukocytes, Mononuclear/metabolism , Matrix Metalloproteinase 14/metabolism , Muscle Fibers, Skeletal/metabolism , Adult , Aged , Animals , Cells, Cultured , Collagen Type I/metabolism , Female , Humans , Leukemia Inhibitory Factor/metabolism , Macrophages/metabolism , Male , Mice , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Resistance Training/methods
5.
Am J Physiol Cell Physiol ; 322(1): C86-C93, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34817266

ABSTRACT

Muscle fibers are syncytial postmitotic cells that can acquire exogenous nuclei from resident muscle stem cells, called satellite cells. Myonuclei are added to muscle fibers by satellite cells during conditions such as load-induced hypertrophy. It is difficult to dissect the molecular contributions of resident versus satellite cell-derived myonuclei during adaptation due to the complexity of labeling distinct nuclear populations in multinuclear cells without label transference between nuclei. To sidestep this barrier, we used a genetic mouse model where myonuclear DNA can be specifically and stably labeled via nonconstitutive H2B-GFP at any point in the lifespan. Resident myonuclei (Mn) were GFP-tagged in vivo before 8 wk of progressive weighted wheel running (PoWeR) in adult mice (>4-mo-old). Resident + satellite cell-derived myonuclei (Mn+SC Mn) were labeled at the end of PoWeR in a separate cohort. Following myonuclear isolation, promoter DNA methylation profiles acquired with low-input reduced representation bisulfite sequencing (RRBS) were compared to deduce epigenetic contributions of satellite cell-derived myonuclei during adaptation. Resident myonuclear DNA has hypomethylated promoters in genes related to protein turnover, whereas the addition of satellite cell-derived myonuclei shifts myonuclear methylation profiles to favor transcription factor regulation and cell-cell signaling. By comparing myonucleus-specific methylation profiling to previously published single-nucleus transcriptional analysis in the absence (Mn) versus the presence of satellite cells (Mn+SC Mn) with PoWeR, we provide evidence that satellite cell-derived myonuclei may preferentially supply specific ribosomal proteins to growing myofibers and retain an epigenetic "memory" of prior stem cell identity. These data offer insights on distinct epigenetic myonuclear characteristics and contributions during adult muscle growth.


Subject(s)
Adaptation, Physiological/physiology , Cell Nucleus/metabolism , Epigenesis, Genetic/physiology , Muscle Fibers, Skeletal/metabolism , Physical Conditioning, Animal/physiology , Staining and Labeling/methods , Animals , Cell Nucleus/chemistry , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/chemistry , Physical Conditioning, Animal/methods , Satellite Cells, Skeletal Muscle/chemistry , Satellite Cells, Skeletal Muscle/metabolism , Time Factors
6.
FASEB J ; 35(6): e21644, 2021 06.
Article in English | MEDLINE | ID: mdl-34033143

ABSTRACT

How regular physical activity is able to improve health remains poorly understood. The release of factors from skeletal muscle following exercise has been proposed as a possible mechanism mediating such systemic benefits. We describe a mechanism wherein skeletal muscle, in response to a hypertrophic stimulus induced by mechanical overload (MOV), released extracellular vesicles (EVs) containing muscle-specific miR-1 that were preferentially taken up by epidydimal white adipose tissue (eWAT). In eWAT, miR-1 promoted adrenergic signaling and lipolysis by targeting Tfap2α, a known repressor of Adrß3 expression. Inhibiting EV release prevented the MOV-induced increase in eWAT miR-1 abundance and expression of lipolytic genes. Resistance exercise decreased skeletal muscle miR-1 expression with a concomitant increase in plasma EV miR-1 abundance, suggesting a similar mechanism may be operative in humans. Altogether, these findings demonstrate that skeletal muscle promotes metabolic adaptations in adipose tissue in response to MOV via EV-mediated delivery of miR-1.


Subject(s)
Adipose Tissue, White/physiopathology , Exercise , Extracellular Vesicles/physiology , Lipolysis , MicroRNAs/genetics , Muscle, Skeletal/physiopathology , Stress, Mechanical , Transcription Factor AP-2/metabolism , Adolescent , Adult , Animals , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Transcription Factor AP-2/genetics , Young Adult
7.
J Physiol ; 599(21): 4845-4863, 2021 11.
Article in English | MEDLINE | ID: mdl-34569067

ABSTRACT

There is emerging evidence of a gut microbiome-skeletal muscle axis. The purpose of this study was to determine if an intact gut microbiome was necessary for skeletal muscle adaptation to exercise. Forty-two 4-month-old female C57BL/6J mice were randomly assigned to untreated (U) or antibiotic-treated (T) non-running controls (CU or CT, respectively) or progressive weighted wheel running (PoWeR, P) untreated (PU) or antibiotic-treated (PT) groups. Antibiotic treatment resulted in disruption of the gut microbiome as indicated by a significant depletion of gut microbiome bacterial species in both CT and PT groups. The training stimulus was the same between PU and PT groups as assessed by weekly (12.35 ± 2.06 vs. 11.09 ± 1.76 km/week, respectively) and total (778.9 ± 130.5 vs. 703.8 ± 112.9 km, respectively) running activity. In response to PoWeR, PT showed less hypertrophy of soleus type 1 and 2a fibres and plantaris type 2b/x fibres compared to PU. The higher satellite cell and myonuclei abundance of PU plantaris muscle after PoWeR was not observed in PT. The fibre-type shift of PU plantaris muscle to a more oxidative type 2a fibre composition following PoWeR was blunted in PT. There was no difference in serum cytokine levels among all groups suggesting disruption of the gut microbiome did not induce systemic inflammation. The results of this study provide the first evidence that an intact gut microbiome is necessary for skeletal muscle adaptation to exercise. KEY POINTS: Dysbiosis of the gut microbiome caused by continuous antibiotic treatment did not affect running activity. Continuous treatment with antibiotics did not result in systemic inflammation as indicated by serum cytokine levels. Gut microbiome dysbiosis was associated with blunted fibre type-specific hypertrophy in the soleus and plantaris muscles in response to progressive weighted wheel running (PoWeR). Gut microbiome dysbiosis was associated with impaired PoWeR-induced fibre-type shift in the plantaris muscle. Gut microbiome dysbiosis was associated with a loss of PoWeR-induced myonuclei accretion in the plantaris muscle.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Animals , Female , Mice , Mice, Inbred C57BL , Motor Activity , Muscle, Skeletal
8.
FASEB J ; 34(5): 7018-7035, 2020 05.
Article in English | MEDLINE | ID: mdl-32246795

ABSTRACT

Over the past 20 years, various identifiers of cellular senescence have been used to quantify the abundance of these cells in different tissues. These include classic markers such as p16, senescence-associated ß-gal, and γH2AX, in addition to more recent markers (Sudan Black B and HMGB1). In vivo data on the usefulness of these markers in skeletal muscle are very limited and inconsistent. In the present study, we attempted to identify senescent cells in frozen human skeletal muscle biopsies using these markers to determine the effects of age and obesity on senescent cell burden; however, we were only able to assess the abundance of DNA-damaged nuclei using γH2AX immunohistochemistry. The abundance of γH2AX+ cells, including satellite cells, was not higher in muscle from old compared to young individuals; however, γH2AX+ cells were higher with obesity. Additionally, terminally differentiated, postmitotic myofiber nuclei from obese individuals had elevated γH2AX abundance compared to muscle from lean individuals. Analyses of gene expression support the conclusion that the elevated DNA damage and the senescence-associated secretory phenotype are preferentially associated with obesity in skeletal muscle. These data implicate obesity as a larger contributor to DNA damage in skeletal muscle than aging; however, more sensitive senescence markers for human skeletal muscle are needed to determine if these cells are in fact senescent.


Subject(s)
Aging/metabolism , Histones/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Adult , Aged , Aged, 80 and over , Aging/pathology , Biomarkers/metabolism , Cell Differentiation , Cellular Senescence , DNA Damage , DNA Repair/genetics , Female , Humans , Immunohistochemistry , Male , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Obesity/pathology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Young Adult
9.
Am J Physiol Cell Physiol ; 318(6): C1178-C1188, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32320286

ABSTRACT

To date, studies that have aimed to investigate the role of satellite cells during adult skeletal muscle adaptation and hypertrophy have utilized a nontranslational stimulus and/or have been performed over a relatively short time frame. Although it has been shown that satellite cell depletion throughout adulthood does not drive skeletal muscle loss in sedentary mice, it remains unknown how satellite cells participate in skeletal muscle adaptation to long-term physical activity. The current study was designed to determine whether reduced satellite cell content throughout adulthood would influence the transcriptome-wide response to physical activity and diminish the adaptive response of skeletal muscle. We administered vehicle or tamoxifen to adult Pax7-diphtheria toxin A (DTA) mice to deplete satellite cells and assigned them to sedentary or wheel-running conditions for 13 mo. Satellite cell depletion throughout adulthood reduced balance and coordination, overall running volume, and the size of muscle proprioceptors (spindle fibers). Furthermore, satellite cell participation was necessary for optimal muscle fiber hypertrophy but not adaptations in fiber type distribution in response to lifelong physical activity. Transcriptome-wide analysis of the plantaris and soleus revealed that satellite cell function is muscle type specific; satellite cell-dependent myonuclear accretion was apparent in oxidative muscles, whereas initiation of G protein-coupled receptor (GPCR) signaling in the glycolytic plantaris may require satellite cells to induce optimal adaptations to long-term physical activity. These findings suggest that satellite cells play a role in preserving physical function during aging and influence muscle adaptation during sustained periods of physical activity.


Subject(s)
Muscle Fibers, Skeletal/pathology , Physical Conditioning, Animal , Running , Satellite Cells, Skeletal Muscle/pathology , Sedentary Behavior , Adaptation, Physiological , Animals , Diphtheria Toxin/genetics , Female , Gene Expression Regulation , Glycolysis , Hypertrophy , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Oxidation-Reduction , PAX7 Transcription Factor/genetics , Peptide Fragments/genetics , RNA, Untranslated/genetics , Satellite Cells, Skeletal Muscle/metabolism , Time Factors
10.
J Neurophysiol ; 124(6): 1571-1577, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33052800

ABSTRACT

Changes to cerebral miRNA expression have been implicated in the progression of Alzheimer's disease (AD), as miRNAs that regulate the expression of gene products involved in amyloid beta (Aß) processing, such as BACE1, are dysregulated in those that suffer from AD. Exercise training improves cognition and reduces BACE1 and Aß-plaque burden; however, the mechanisms are not fully understood. Using our progressive weighted wheel running (PoWeR) exercise program, we assessed the effect of 20 wk of exercise training on changes in hippocampal miRNA expression in female 3xTg-AD (3xTg) mice. PoWeR was sufficient to promote muscle hypertrophy and increase myonuclear abundance. Furthermore, PoWeR elevated hippocampal Dicer gene expression in 3xTg mice, while altering miRNA expression toward a more wild-type profile. Specifically, miR-29, which is validated to target BACE1, was significantly lower in sedentary 3xTg mice when compared with wild-type but was elevated following PoWeR. Accordingly, BACE1 gene expression, along with detergent-soluble Aß1-42, was lower in PoWeR-trained 3xTg mice. Our data suggest that PoWeR training upregulates Dicer gene expression to alter cerebral miRNA expression, which may contribute to reduced Aß accumulation and delay AD progression.NEW & NOTEWORTHY Previous studies have outlined the beneficial effects of exercise on lowering BACE1 expression and reducing Aß plaques. This study extends upon the work of others by outlining a new potential mechanism by which exercise elicits beneficial effects on Alzheimer's disease pathology, specifically through modulation of Dicer and miRNA expression. This is the first study to examine Dicer and miRNA expression in the hippocampus of the 3xTg model within the context of exercise.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/metabolism , DEAD-box RNA Helicases/metabolism , Hippocampus/metabolism , MicroRNAs/metabolism , Peptide Fragments/metabolism , Physical Conditioning, Animal , Ribonuclease III/metabolism , Animals , Disease Models, Animal , Female , Mice, Transgenic , Motor Activity , RNA, Messenger/metabolism
11.
Am J Physiol Cell Physiol ; 316(5): C649-C654, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30840493

ABSTRACT

Myonuclei gained during exercise-induced skeletal muscle hypertrophy may be long-lasting and could facilitate future muscle adaptability after deconditioning, a concept colloquially termed "muscle memory." The evidence for this is limited, mostly due to the lack of a murine exercise-training paradigm that is nonsurgical and reversible. To address this limitation, we developed a novel progressive weighted-wheel-running (PoWeR) model of murine exercise training to test whether myonuclei gained during exercise persist after detraining. We hypothesized that myonuclei acquired during training-induced hypertrophy would remain following loss of muscle mass with detraining. Singly housed female C57BL/6J mice performed 8 wk of PoWeR, while another group performed 8 wk of PoWeR followed by 12 wk of detraining. Age-matched sedentary cage-dwelling mice served as untrained controls. Eight weeks of PoWeR yielded significant plantaris muscle fiber hypertrophy, a shift to a more oxidative phenotype, and greater myonuclear density than untrained mice. After 12 wk of detraining, the plantaris muscle returned to an untrained phenotype with fewer myonuclei. A finding of fewer myonuclei simultaneously with plantaris deconditioning argues against a muscle memory mechanism mediated by elevated myonuclear density in primarily fast-twitch muscle. PoWeR is a novel, practical, and easy-to-deploy approach for eliciting robust hypertrophy in mice, and our findings can inform future research on the mechanisms underlying skeletal muscle adaptive potential and muscle memory.


Subject(s)
Muscle Fibers, Skeletal/physiology , Physical Conditioning, Animal/methods , Physical Conditioning, Animal/physiology , Weight-Bearing/physiology , Animals , Female , Hypertrophy/pathology , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/pathology
12.
Exerc Sport Sci Rev ; 47(2): 108-115, 2019 04.
Article in English | MEDLINE | ID: mdl-30640746

ABSTRACT

Skeletal muscle fiber branching and splitting typically is associated with damage and regeneration and is considered pathological when observed during loading-induced hypertrophy. We hypothesize that fiber splitting is a nonpathological component of extreme loading and hypertrophy, which is primarily supported by evidence in animals, and propose that the mechanisms and consequences of fiber splitting deserve further exploration.


Subject(s)
Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/growth & development , Stress, Physiological , Animals , Humans , Models, Animal , Resistance Training
13.
14.
Biochem Biophys Res Commun ; 482(4): 1067-1072, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27913296

ABSTRACT

Recent findings in adipocytes suggest that mitogen-activated protein kinase (MAPK)/extracellular-regulated signaling kinase (ERK) kinase 1/2 (MEK1/2) signaling regulates regulated in development and DNA damage 1 (REDD1) protein expression. Similarly, our previous work show that a lack of REDD1 protein expression, and associated hyperactive basal mechanistic target of rapamycin (mTOR) signaling, limits skeletal muscle's response to insulin. Therefore, we sought to determine: 1) if MEK1/2 inhibition is sufficient to reduce REDD1 protein expression and subsequently insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation via negative feedback of hyperactive mTOR in REDD1 wild-type (WT) mice and 2) if rapamycin-mediated mTOR inhibition is sufficient to improve IRS-1 tyrosine phosphorylation in REDD1 knockout (KO) mice. REDD1 WT mice were injected with 10 mg/kg BW of the MEK1/2 non-competitive inhibitor, PD184352, 3 h prior to acute insulin treatment. In separate studies, REDD1 KO mice were injected with 5 mg/kg BW of the mTOR inhibitor, rapamycin, 3 h prior to acute insulin treatment. Following the inhibitor treatment period, markers of insulin signaling activation (IRS-1 Y1222, MEK1/2 S217/221, ERK1/2 T202/Y204), REDD1, and mTOR signaling activation (S6K1 T389, rpS6 S240/244) were examined in skeletal muscle collected before and after a 10 min insulin treatment. PD184352 treatment reduced MEK/ERK phosphorylation and REDD1 protein expression, independent of insulin. This reduction in REDD1 protein expression was associated with elevated basal S6K1 and rpS6 phosphorylation and reduced insulin stimulated IRS-1 phosphorylation. Conversely, rapamycin inhibited S6K1 and rpS6 activation, and significantly improved insulin -stimulated activation of IRS-1 and MEK1/2 in KO mice. These data support that REDD1 is required for normal insulin-stimulated signaling, and that a subtle balance exists between MEK1/2, REDD1, and mTOR for the proper regulation of insulin signaling.


Subject(s)
Insulin/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Muscle, Skeletal/metabolism , Transcription Factors/metabolism , Animals , Benzamides/chemistry , Mice , Mice, Knockout , Phosphorylation , Signal Transduction , Sirolimus/chemistry , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics
15.
Muscle Nerve ; 53(1): 107-17, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25926238

ABSTRACT

INTRODUCTION: Appropriate activation of growth signaling pathways, specifically mammalian target of rapamycin complex 1 (mTORC1), is central to muscle mass and metabolism. The goal of these studies was to examine the effects of metformin on mTORC1 signaling in aged skeletal muscle in an attempt to normalize growth signaling. METHODS: Aged (23m) and young (3m) male mice were fed a low fat diet without or with 0.5% metformin for up to 8 weeks, then mTORC1-related signaling was examined in the plantar flexor complex. RESULTS: Metformin had no significant effect on lowering body weight or muscle mass in aged animals, nor altered p70 S6 Kinase 1 (S6K1) and 4E-binding protein 1 (4E-BP1) phosphorylation. However, it significantly (P < 0.05) reduced body weight and lowered S6K1 and rpS6 phosphorylation in the young. CONCLUSIONS: Collectively, these data suggest metformin is ineffective at normalizing growth signaling in aged skeletal muscle.


Subject(s)
Aging/drug effects , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Multiprotein Complexes/metabolism , Muscle, Skeletal/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Age Factors , Animals , Exercise Test , Histocompatibility Antigens Class I/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism
16.
Am J Physiol Regul Integr Comp Physiol ; 309(8): R855-63, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26269521

ABSTRACT

The objective of this study was to establish whether alterations in the REDD1-mTOR axis underlie skeletal muscle insensitivity to insulin in Type 2 diabetic (T2D), obese individuals. Vastus lateralis muscle biopsies were obtained from lean, control and obese, T2D subjects under basal and after a 2-h hyperinsulinemic (40 mU·m(-2)·min(-1))-euglycemic (5 mM) clamp. Muscle lysates were examined for total REDD1, and phosphorylated Akt, S6 kinase 1 (S6K1), 4E-BP1, ERK1/2, and MEK1/2 via Western blot analysis. Under basal conditions [(-) insulin], T2D muscle exhibited higher S6K1 and ERK1/2 and lower 4E-BP1 phosphorylation (P < 0.05), as well as elevations in blood cortisol, glucose, insulin, glycosylated hemoglobin (P < 0.05) vs. lean controls. Following insulin infusion, whole body glucose disposal rates (GDR; mg/kg/min) were lower (P < 0.05) in the T2D vs. the control group. The basal-to-insulin percent change in REDD1 expression was higher (P < 0.05) in muscle from the T2D vs. the control group. Whereas, the basal-to-insulin percent change in muscle Akt, S6K1, ERK1/2, and MEK1/2 phosphorylation was significantly lower (P < 0.05) in the T2D vs. the control group. Findings from this study propose a REDD1-regulated mechanism in T2D skeletal muscle that may contribute to whole body insulin resistance and may be a target to improve insulin action in insulin-resistant individuals.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin/pharmacology , Multiprotein Complexes/metabolism , Muscle, Skeletal/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Adult , Case-Control Studies , Female , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Middle Aged , Multiprotein Complexes/genetics , Muscle, Skeletal/drug effects , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics
17.
Biochem Biophys Res Commun ; 453(4): 778-83, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25445588

ABSTRACT

A lack of the REDD1 promotes dysregulated growth signaling, though little has been established with respect to the metabolic role of REDD1. Therefore, the goal of this study was to determine the role of REDD1 on glucose and insulin tolerance, as well as insulin stimulated growth signaling pathway activation in skeletal muscle. First, intraperitoneal (IP) injection of glucose or insulin were administered to REDD1 wildtype (WT) versus knockout (KO) mice to examine changes in blood glucose over time. Next, alterations in skeletal muscle insulin (IRS-1, Akt, ERK 1/2) and growth (4E-BP1, S6K1, REDD1) signaling intermediates were determined before and after IP insulin treatment (10min). REDD1 KO mice were both glucose and insulin intolerant when compared to WT mice, evident by higher circulating blood glucose concentrations and a greater area under the curve following IP injections of glucose or insulin. While the REDD1 KO exhibited significant though blunted insulin-stimulated increases (p<0.05) in Akt S473 and T308 phosphorylation versus the WT mice, acute insulin treatment has no effect (p<0.05) on REDD1 KO skeletal muscle 4E-BP1 T37/46, S6K1 T389, IRS-1 Y1222, and ERK 1/2 T202/Y204 phosphorylation versus the WT mice. Collectively, these novel data suggest that REDD1 has a more distinct role in whole body and skeletal muscle metabolism and insulin action than previously thought.


Subject(s)
Glucose/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Muscle, Skeletal/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Blood Glucose/metabolism , Glucose Tolerance Test , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism
18.
Exerc Sport Sci Rev ; 47(4): 260, 2019 10.
Article in English | MEDLINE | ID: mdl-31524788
19.
bioRxiv ; 2024 Jun 02.
Article in English | MEDLINE | ID: mdl-38854012

ABSTRACT

Regular exercise yields a multitude of systemic benefits, many of which may be mediated through the gut microbiome. Here, we report that cecal microbial transplants (CMTs) from exercise-trained vs. sedentary mice have modest benefits in reducing skeletal muscle atrophy using a mouse model of unilaterally hindlimb-immobilization. Direct administration of top microbial-derived exerkines from an exercise-trained gut microbiome preserved muscle function and prevented skeletal muscle atrophy.

20.
Geroscience ; 46(2): 2153-2176, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37872294

ABSTRACT

Skeletal muscle adaptation to external stimuli, such as regeneration following injury and hypertrophy in response to resistance exercise, are blunted with advanced age. The accumulation of senescent cells, along with defects in myogenic progenitor cell (MPC) proliferation, have been strongly linked as contributing factors to age-associated impairment in muscle adaptation. p53 plays an integral role in all these processes, as upregulation of p53 causes apoptosis in senescent cells and prevents mitotic catastrophe in MPCs from old mice. The goal of this study was to determine if a novel pharmaceutical agent (BI01), which functions by upregulating p53 through inhibition of binding to MDM2, the primary p53 regulatory protein, improves muscle regeneration and hypertrophy in old mice. BI01 effectively reduced the number of senescent cells in vitro but had no effect on MPC survival or proliferation at a comparable dose. Following repeated oral gavage with 2 mg/kg of BI01 (OS) or vehicle (OV), old mice (24 months) underwent unilateral BaCl2 injury in the tibialis anterior (TA) muscle, with PBS injections serving as controls. After 7 days, satellite cell number was higher in the TA of OS compared to OV mice, as was the expression of genes involved in ATP production. By 35 days, old mice treated with BI01 displayed reduced senescent cell burden, enhanced regeneration (higher muscle mass and fiber cross-sectional area) and restoration of muscle function relative to OV mice. To examine the impact of 2 mg/kg BI01 on muscle hypertrophy, the plantaris muscle was subjected to 28 days of mechanical overload (MOV) in OS and OV mice. In response to MOV, OS mice had larger plantaris muscles and muscle fibers than OV mice, particularly type 2b + x fibers, associated with reduced senescent cells. Together our data show that BI01 is an effective senolytic agent that may also augment muscle metabolism to enhance muscle regeneration and hypertrophy in old mice.


Subject(s)
Muscle, Skeletal , Tumor Suppressor Protein p53 , Animals , Mice , Cellular Senescence , Hypertrophy , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL