Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Aging Cell ; 23(4): e14097, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38297807

ABSTRACT

The loss of skeletal muscle mass during aging is a significant health concern linked to adverse outcomes in older individuals. Understanding the molecular basis of age-related muscle loss is crucial for developing strategies to combat this debilitating condition. Long noncoding RNAs (lncRNAs) are a largely uncharacterized class of biomolecules that have been implicated in cellular homeostasis and dysfunction across a many tissues and cell types. To identify lncRNAs that might contribute to skeletal muscle aging, we screened for lncRNAs whose expression was altered in vastus lateralis muscle from older compared to young adults. We identified FRAIL1 as an aging-induced lncRNA with high abundance in human skeletal muscle. In healthy young and older adults, skeletal muscle FRAIL1 was increased with age in conjunction with lower muscle function. Forced expression of FRAIL1 in mouse tibialis anterior muscle elicits a dose-dependent reduction in skeletal muscle fiber size that is independent of changes in muscle fiber type. Furthermore, this reduction in muscle size is dependent on an intact region of FRAIL1 that is highly conserved across non-human primates. Unbiased transcriptional and proteomic profiling of the effects of FRAIL1 expression in mouse skeletal muscle revealed widespread changes in mRNA and protein abundance that recapitulate age-related changes in pathways and processes that are known to be altered in aging skeletal muscle. Taken together, these findings shed light on the intricate molecular mechanisms underlying skeletal muscle aging and implicate FRAIL1 in age-related skeletal muscle phenotypes.


Subject(s)
RNA, Long Noncoding , Humans , Animals , Mice , Aged , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Proteomics , Muscle, Skeletal/metabolism , Muscle Fibers, Skeletal/metabolism , Aging/metabolism
2.
Animals (Basel) ; 14(2)2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38254356

ABSTRACT

Muscle atrophy and weakness are prevalent and debilitating conditions in dogs that cannot be reliably prevented or treated by current approaches. In non-canine species, the natural dietary compound ursolic acid inhibits molecular mechanisms of muscle atrophy, leading to improvements in muscle health. To begin to translate ursolic acid to canine health, we developed a novel ursolic acid dietary supplement for dogs and confirmed its safety and tolerability in dogs. We then conducted a randomized, placebo-controlled, proof-of-concept efficacy study in older beagles with age-related muscle atrophy, also known as sarcopenia. Animals received placebo or ursolic acid dietary supplements once a day for 60 days. To assess the study's primary outcome, we biopsied the quadriceps muscle and quantified atrophy-associated mRNA expression. Additionally, to determine whether the molecular effects of ursolic acid might have functional correlates consistent with improvements in muscle health, we assessed secondary outcomes of exercise participation and T-maze performance. Importantly, in canine skeletal muscle, ursolic acid inhibited numerous mRNA expression changes that are known to promote muscle atrophy and weakness. Furthermore, ursolic acid significantly improved exercise participation and T-maze performance. These findings identify ursolic acid as a natural dietary compound that inhibits molecular mechanisms of muscle atrophy and improves functional performance in dogs.

3.
JCI Insight ; 8(22)2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37815864

ABSTRACT

Aging and many illnesses and injuries impair skeletal muscle mass and function, but the molecular mechanisms are not well understood. To better understand the mechanisms, we generated and studied transgenic mice with skeletal muscle-specific expression of growth arrest and DNA damage inducible α (GADD45A), a signaling protein whose expression in skeletal muscle rises during aging and a wide range of illnesses and injuries. We found that GADD45A induced several cellular changes that are characteristic of skeletal muscle atrophy, including a reduction in skeletal muscle mitochondria and oxidative capacity, selective atrophy of glycolytic muscle fibers, and paradoxical expression of oxidative myosin heavy chains despite mitochondrial loss. These cellular changes were at least partly mediated by MAP kinase kinase kinase 4, a protein kinase that is directly activated by GADD45A. By inducing these changes, GADD45A decreased the mass of muscles that are enriched in glycolytic fibers, and it impaired strength, specific force, and endurance exercise capacity. Furthermore, as predicted by data from mouse models, we found that GADD45A expression in skeletal muscle was associated with muscle weakness in humans. Collectively, these findings identify GADD45A as a mediator of mitochondrial loss, atrophy, and weakness in mouse skeletal muscle and a potential target for muscle weakness in humans.


Subject(s)
Mitochondria, Muscle , Muscle, Skeletal , Muscular Atrophy , Animals , Humans , Mice , Aging , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Mitochondria, Muscle/metabolism , Muscle Weakness/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/pathology
4.
Geroscience ; 45(4): 2525-2543, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37014538

ABSTRACT

Aging slowly erodes skeletal muscle strength and mass, eventually leading to profound functional deficits and muscle atrophy. The molecular mechanisms of skeletal muscle aging are not well understood. To better understand mechanisms of muscle aging, we investigated the potential role of ATF4, a transcription regulatory protein that can rapidly promote skeletal muscle atrophy in young animals deprived of adequate nutrition or activity. To test the hypothesis that ATF4 may be involved in skeletal muscle aging, we studied fed and active muscle-specific ATF4 knockout mice (ATF4 mKO mice) at 6 months of age, when wild-type mice have achieved peak muscle mass and function, and at 22 months of age, when wild-type mice have begun to manifest age-related muscle atrophy and weakness. We found that 6-month-old ATF4 mKO mice develop normally and are phenotypically indistinguishable from 6-month-old littermate control mice. However, as ATF4 mKO mice become older, they exhibit significant protection from age-related declines in strength, muscle quality, exercise capacity, and muscle mass. Furthermore, ATF4 mKO muscles are protected from some of the transcriptional changes characteristic of normal muscle aging (repression of certain anabolic mRNAs and induction of certain senescence-associated mRNAs), and ATF4 mKO muscles exhibit altered turnover of several proteins with important roles in skeletal muscle structure and metabolism. Collectively, these data suggest ATF4 as an essential mediator of skeletal muscle aging and provide new insight into a degenerative process that impairs the health and quality of life of many older adults.


Subject(s)
Muscle, Skeletal , Quality of Life , Mice , Animals , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Aging/metabolism , Mice, Knockout
5.
J Proteome Res ; 22(2): 311-322, 2023 02 03.
Article in English | MEDLINE | ID: mdl-36165806

ABSTRACT

In spite of its central role in biology and disease, protein turnover is a largely understudied aspect of most proteomic studies due to the complexity of computational workflows that analyze in vivo turnover rates. To address this need, we developed a new computational tool, TurnoveR, to accurately calculate protein turnover rates from mass spectrometric analysis of metabolic labeling experiments in Skyline, a free and open-source proteomics software platform. TurnoveR is a straightforward graphical interface that enables seamless integration of protein turnover analysis into a traditional proteomics workflow in Skyline, allowing users to take advantage of the advanced and flexible data visualization and curation features built into the software. The computational pipeline of TurnoveR performs critical steps to determine protein turnover rates, including isotopologue demultiplexing, precursor-pool correction, statistical analysis, and generation of data reports and visualizations. This workflow is compatible with many mass spectrometric platforms and recapitulates turnover rates and differential changes in turnover rates between treatment groups calculated in previous studies. We expect that the addition of TurnoveR to the widely used Skyline proteomics software will facilitate wider utilization of protein turnover analysis in highly relevant biological models, including aging, neurodegeneration, and skeletal muscle atrophy.


Subject(s)
Proteomics , Software , Proteomics/methods , Proteolysis , Mass Spectrometry/methods , Workflow , Isotope Labeling/methods
6.
J Nutr ; 152(4): 926-938, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34958390

ABSTRACT

Activating transcription factor 4 (ATF4) is a multifunctional transcription regulatory protein in the basic leucine zipper superfamily. ATF4 can be expressed in most if not all mammalian cell types, and it can participate in a variety of cellular responses to specific environmental stresses, intracellular derangements, or growth factors. Because ATF4 is involved in a wide range of biological processes, its roles in human health and disease are not yet fully understood. Much of our current knowledge about ATF4 comes from investigations in cultured cell models, where ATF4 was originally characterized and where further investigations continue to provide new insights. ATF4 is also an increasingly prominent topic of in vivo investigations in fully differentiated mammalian cell types, where our current understanding of ATF4 is less complete. Here, we review some important high-level concepts and questions concerning the basic biology of ATF4. We then discuss current knowledge and emerging questions about the in vivo role of ATF4 in one fully differentiated cell type, mammalian skeletal muscle fibers.


Subject(s)
Activating Transcription Factor 4 , Muscular Atrophy , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Biology , Cell Differentiation , Humans , Mammals , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/etiology
7.
EMBO J ; 40(9): e106491, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33847380

ABSTRACT

Exercise can alter the skeletal muscle DNA methylome, yet little is known about the role of the DNA methylation machinery in exercise capacity. Here, we show that DNMT3A expression in oxidative red muscle increases greatly following a bout of endurance exercise. Muscle-specific Dnmt3a knockout mice have reduced tolerance to endurance exercise, accompanied by reduction in oxidative capacity and mitochondrial respiration. Moreover, Dnmt3a-deficient muscle overproduces reactive oxygen species (ROS), the major contributors to muscle dysfunction. Mechanistically, we show that DNMT3A suppresses the Aldh1l1 transcription by binding to its promoter region, altering its epigenetic profile. Forced expression of ALDH1L1 elevates NADPH levels, which results in overproduction of ROS by the action of NADPH oxidase complex, ultimately resulting in mitochondrial defects in myotubes. Thus, inhibition of ALDH1L1 pathway can rescue oxidative stress and mitochondrial dysfunction from Dnmt3a deficiency in myotubes. Finally, we show that in vivo knockdown of Aldh1l1 largely rescues exercise intolerance in Dnmt3a-deficient mice. Together, we establish that DNMT3A in skeletal muscle plays a pivotal role in endurance exercise by controlling intracellular oxidative stress.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Muscle, Skeletal/metabolism , Oxidoreductases Acting on CH-NH Group Donors/genetics , Physical Endurance/genetics , Animals , Cell Line , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Gene Expression Profiling , Gene Knockout Techniques , Mice , Mitochondria, Muscle/metabolism , Oxidative Stress , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Rats , Reactive Oxygen Species/metabolism , Sequence Analysis, RNA
8.
J Biol Chem ; 295(9): 2787-2803, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31953319

ABSTRACT

Skeletal muscle atrophy is a highly-prevalent and debilitating condition that remains poorly understood at the molecular level. Previous work found that aging, fasting, and immobilization promote skeletal muscle atrophy via expression of activating transcription factor 4 (ATF4) in skeletal muscle fibers. However, the direct biochemical mechanism by which ATF4 promotes muscle atrophy is unknown. ATF4 is a member of the basic leucine zipper transcription factor (bZIP) superfamily. Because bZIP transcription factors are obligate dimers, and because ATF4 is unable to form highly-stable homodimers, we hypothesized that ATF4 may promote muscle atrophy by forming a heterodimer with another bZIP family member. To test this hypothesis, we biochemically isolated skeletal muscle proteins that associate with the dimerization- and DNA-binding domain of ATF4 (the bZIP domain) in mouse skeletal muscle fibers in vivo Interestingly, we found that ATF4 forms at least five distinct heterodimeric bZIP transcription factors in skeletal muscle fibers. Furthermore, one of these heterodimers, composed of ATF4 and CCAAT enhancer-binding protein ß (C/EBPß), mediates muscle atrophy. Within skeletal muscle fibers, the ATF4-C/EBPß heterodimer interacts with a previously unrecognized and evolutionarily conserved ATF-C/EBP composite site in exon 4 of the Gadd45a gene. This three-way interaction between ATF4, C/EBPß, and the ATF-C/EBP composite site activates the Gadd45a gene, which encodes a critical mediator of muscle atrophy. Together, these results identify a biochemical mechanism by which ATF4 induces skeletal muscle atrophy, providing molecular-level insights into the etiology of skeletal muscle atrophy.


Subject(s)
Activating Transcription Factor 4/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Muscular Atrophy/etiology , Protein Multimerization , Activating Transcription Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Mice , Muscle, Skeletal/pathology
9.
J Appl Physiol (1985) ; 127(4): 1075-1084, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31465716

ABSTRACT

Age-related skeletal muscle atrophy is a very common and serious condition that remains poorly understood at the molecular level. Several lines of evidence have suggested that the tumor suppressor p53 may play a central, causative role in skeletal muscle aging, whereas other, apparently contradictory lines of evidence have suggested that p53 may be critical for normal skeletal muscle function. To help address these issues, we performed an aging study in male muscle-specific p53-knockout mice (p53 mKO mice), which have a lifelong absence of p53 expression in skeletal muscle fibers. We found that the absence of p53 expression in skeletal muscle fibers had no apparent deleterious or beneficial effects on skeletal muscle mass or function under basal conditions up to 6 mo of age, when mice are fully grown and exhibit peak muscle mass and function. Furthermore, at 22 and 25 mo of age, when age-related muscle weakness and atrophy are clearly evident in mice, p53 mKO mice demonstrated no improvement or worsening of skeletal muscle mass or function relative to littermate control mice. At advanced ages, p53 mKO mice began to die prematurely and had an increased incidence of osteosarcoma, precluding analyses of muscle mass and function in very old p53 mKO mice. In light of these results, we conclude that p53 expression in skeletal muscle fibers has minimal if any direct, cell autonomous effect on basal or age-related changes in skeletal muscle mass and function up to at least 22 mo of age.NEW & NOTEWORTHY Previous studies implicated the transcriptional regulator p53 as a potential mediator of age-related skeletal muscle weakness and atrophy. We tested this hypothesis by investigating the effect of aging in muscle-specific p53-knockout mice. Our results strongly suggest that p53 activity within skeletal muscle fibers is not required for age-related skeletal muscle atrophy or weakness.


Subject(s)
Aging/metabolism , Muscle, Skeletal/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Female , Male , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle Weakness/metabolism , Muscular Atrophy/metabolism
10.
FASEB J ; 33(11): 11735-11745, 2019 11.
Article in English | MEDLINE | ID: mdl-31361156

ABSTRACT

Basal protein turnover, which largely relies on the degradation of ubiquitinated substrates, is instrumental for maintenance of muscle mass and function. However, the regulation of ubiquitinated protein degradation in healthy, nonatrophying skeletal muscle is still evolving, and potential tissue-specific modulators remain unknown. Using an unbiased expression analysis of 34 putative autophagy genes across mouse tissues, we identified unc-51 like autophagy activating kinase (Ulk)2, a homolog of the yeast autophagy related protein 1, as particularly enriched in skeletal muscle. Subsequent experiments revealed accumulations of insoluble ubiquitinated protein aggregates associated with the adaptors sequestosome 1 (SQSTM1, also known as p62) and next to breast cancer type 1 susceptibility protein gene 1 protein (NBR1) in adult muscles with ULK2 deficiency. ULK2 deficiency also led to impaired muscle force and caused myofiber atrophy and degeneration. These features were not observed in muscles with deficiency of the ULK2 paralog, ULK1. Furthermore, short-term ULK2 deficiency did not impair autophagy initiation, autophagosome to lysosome fusion, or protease activities of the lysosome and proteasome. Altogether, our results indicate that skeletal muscle ULK2 has a unique role in basal selective protein degradation by stimulating the recognition and proteolytic sequestration of insoluble ubiquitinated protein aggregates associated with p62 and NBR1. These findings have potential implications for conditions of poor protein homeostasis in muscles as observed in several myopathies and aging.-Fuqua, J. D., Mere, C. P., Kronemberger, A., Blomme, J., Bae, D., Turner, K. D., Harris, M. P., Scudese, E., Edwards, M., Ebert, S. M., de Sousa, L. G. O., Bodine, S. C., Yang, L., Adams, C. M., Lira, V. A. ULK2 is essential for degradation of ubiquitinated protein aggregates and homeostasis in skeletal muscle.


Subject(s)
Homeostasis/physiology , Muscle, Skeletal/metabolism , Protein Aggregates/physiology , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagosomes/metabolism , Autophagy/genetics , Lysosomes/metabolism , Male , Mice, Inbred C57BL , Ubiquitination
11.
Physiology (Bethesda) ; 34(4): 232-239, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31165685

ABSTRACT

Skeletal muscle atrophy proceeds through a complex molecular signaling network that is just beginning to be understood. Here, we discuss examples of recently identified molecular mechanisms of muscle atrophy and how they highlight an immense need and opportunity for focused biochemical investigations and further unbiased discovery work.


Subject(s)
Muscle, Skeletal/physiology , Muscular Atrophy/physiopathology , Animals , Humans , Signal Transduction/physiology
12.
J Physiol ; 596(14): 2883-2900, 2018 07.
Article in English | MEDLINE | ID: mdl-29726007

ABSTRACT

KEY POINTS: Force transfer is integral for maintaining skeletal muscle structure and function. One important component is dystrophin. There is limited understanding of how force transfer is impacted by age and loading. Here, we investigate the force transfer apparatus in muscles of adult and old rats exposed to periods of disuse and reloading. Our results demonstrate an increase in dystrophin protein during the reloading phase in the adult tibialis anterior muscle that is delayed in the old muscle. The consequence of this delay is an increased susceptibility towards contraction-induced muscle injury. Central to the lack of dystrophin protein is an increase in miR-31, a microRNA that inhibits dystrophin translation. In vivo electroporation with a miR-31 sponge led to increased dystrophin protein and decreased contraction-induced muscle injury in old skeletal muscle. Overall, our results detail the importance of the force transfer apparatus and provide new mechanisms for contraction-induced injury in ageing skeletal muscle. ABSTRACT: In healthy muscle, the dystrophin-associated glycoprotein complex (DGC), the integrin/focal adhesion complex, intermediate filaments and Z-line proteins transmit force from the contractile proteins to the extracellular matrix. How loading and age affect these proteins is poorly understood. The experiments reported here sought to determine the effect of ageing on the force transfer apparatus following muscle unloading and reloading. Adult (9 months) and old (28 months) rats were subjected to 14 days of hindlimb unloading and 1, 3, 7 and 14 days of reloading. The DGC complex, intermediate filament and Z-line protein and mRNA levels, as well as dystrophin-targeting miRNAs (miR-31, -146b and -374) were examined in the tibialis anterior (TA) and medial gastrocnemius muscles at both ages. There was a significant increase in dystrophin protein levels (2.79-fold) upon 3 days of reloading in the adult TA muscle that did not occur in the old rats (P ≤ 0.05), and the rise in dystrophin protein occurred independent of dystrophin mRNA. The disconnect between dystrophin protein and mRNA levels can partially be explained by age-dependent differences in miR-31. The impaired dystrophin response in aged muscle was followed by an increase in other force transfer proteins (ß-dystroglycan, desmuslin and LIM) that was not sufficient to prevent membrane disruption and muscle injury early in the reloading period. Inserting a miR-31 sponge increased dystrophin protein and decreased contraction-induced injury in the TA (P ≤ 0.05). Collectively, these data suggest that increased miR-31 with age contributes to an impaired dystrophin response and increased muscle injury after disuse.


Subject(s)
Dystrophin/metabolism , Gene Expression Regulation , Hindlimb Suspension/physiology , Mechanotransduction, Cellular , MicroRNAs/genetics , Muscle Contraction , Muscle, Skeletal/physiology , Aging , Animals , Dystrophin/genetics , Male , Muscular Atrophy/physiopathology , Rats , Rats, Inbred BN , Rats, Inbred F344
13.
J Chromatogr Sci ; 56(7): 644-649, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-29701757

ABSTRACT

Ultra-performance liquid chromatography (UPLC) interfaced with atmospheric pressure chemical ionization mass-spectrometry was used to separate and quantify ursolic acid (UA) and oleanolic acid (OA) in human plasma. UA and OA were extracted from 0.5 mL human plasma using supported liquid extraction and separated utilizing an Acquity UPLC HSS column. The method has been validated for both UA and OA quantitation with a limit of detection of 0.5 ng/mL. The UPLC separations are carried out with isocratic elution with methanol and 5 mM ammonium acetate in water (85:15) as a mobile phase at a flow rate of 0.4 mL/min. The assay was linear from 1 ng/mL to 100 ng/mL for both analytes. The total analysis time was 7 min with the retention times of 3.25 (internal standard), 3.65 (UA) and 3.85 min (OA). Recovery of drug from plasma ranged from 70% to 115%. Analysis of quality control samples at 3, 30 and 80 ng/mL (n = 14) had an intra-day coefficient of variation of 9.9%, 4.3% and 5.5%, respectively. A proof-of-concept study in human patients who consumed apple peels indicates that this analytical method could be applied to clinical studies of UA and/or OA in human subjects.


Subject(s)
Chromatography, High Pressure Liquid/methods , Mass Spectrometry/methods , Oleanolic Acid/blood , Triterpenes/blood , Humans , Linear Models , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity , Triterpenes/chemistry , Triterpenes/pharmacokinetics , Ursolic Acid
14.
Curr Opin Clin Nutr Metab Care ; 20(3): 164-168, 2017 May.
Article in English | MEDLINE | ID: mdl-28376050

ABSTRACT

PURPOSE OF REVIEW: Here, we discuss recent work focused on the role of activating transcription factor 4 (ATF4) in skeletal muscle atrophy. RECENT FINDINGS: Muscle atrophy involves and requires widespread changes in skeletal muscle gene expression; however, the transcriptional regulatory proteins responsible for those changes are not yet well defined. Recent work indicates that some forms of muscle atrophy require ATF4, a stress-inducible bZIP transcription factor subunit that helps to mediate a broad range of stress responses in mammalian cells. ATF4 expression in skeletal muscle fibers is sufficient to induce muscle fiber atrophy and required for muscle atrophy during several stress conditions, including aging, fasting, and limb immobilization. By helping to activate specific genes in muscle fibers, ATF4 contributes to the expression of numerous mRNAs, including at least two mRNAs (Gadd45a and p21) that encode mediators of muscle fiber atrophy. Gadd45a promotes muscle fiber atrophy by activating the protein kinase MEKK4. p21 promotes atrophy by reducing expression of spermine oxidase, a metabolic enzyme that helps to maintain muscle fiber size under nonstressed conditions. SUMMARY: In skeletal muscle fibers, ATF4 is critical component of a complex and incompletely understood molecular signaling network that causes muscle atrophy during aging, fasting, and immobilization.


Subject(s)
Activating Transcription Factor 4/metabolism , Aging/physiology , Fasting/physiology , Immobilization/physiology , Muscular Atrophy/genetics , Animals , Humans , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/physiopathology , Signal Transduction/genetics
15.
Trends Endocrinol Metab ; 27(11): 796-806, 2016 11.
Article in English | MEDLINE | ID: mdl-27444066

ABSTRACT

Aging impairs skeletal muscle protein synthesis, leading to muscle weakness and atrophy. However, the underlying molecular mechanisms remain poorly understood. Here, we review evidence that mammalian/mechanistic target of rapamycin complex 1 (mTORC1)-mediated and activating transcription factor 4 (ATF4)-mediated amino acid (AA) sensing pathways, triggered by impaired AA delivery to aged skeletal muscle, may play important roles in skeletal muscle aging. Interventions that alleviate age-related impairments in muscle protein synthesis, strength, and/or muscle mass appear to do so by reversing age-related changes in skeletal muscle AA delivery, mTORC1 activity, and/or ATF4 activity. An improved understanding of the mechanisms and roles of AA sensing pathways in skeletal muscle may lead to evidence-based strategies to attenuate sarcopenia.


Subject(s)
Amino Acids/metabolism , Muscle, Skeletal/metabolism , Activating Transcription Factor 4/metabolism , Animals , Humans , TOR Serine-Threonine Kinases/metabolism , Triterpenes/metabolism , Ursolic Acid
16.
J Biol Chem ; 291(34): 17496-17509, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27358404

ABSTRACT

Skeletal muscle atrophy is a serious and highly prevalent condition that remains poorly understood at the molecular level. Previous work found that skeletal muscle atrophy involves an increase in skeletal muscle Gadd45a expression, which is necessary and sufficient for skeletal muscle fiber atrophy. However, the direct mechanism by which Gadd45a promotes skeletal muscle atrophy was unknown. To address this question, we biochemically isolated skeletal muscle proteins that associate with Gadd45a as it induces atrophy in mouse skeletal muscle fibers in vivo We found that Gadd45a interacts with multiple proteins in skeletal muscle fibers, including, most prominently, MEKK4, a mitogen-activated protein kinase kinase kinase that was not previously known to play a role in skeletal muscle atrophy. Furthermore, we found that, by forming a complex with MEKK4 in skeletal muscle fibers, Gadd45a increases MEKK4 protein kinase activity, which is both sufficient to induce skeletal muscle fiber atrophy and required for Gadd45a-mediated skeletal muscle fiber atrophy. Together, these results identify a direct biochemical mechanism by which Gadd45a induces skeletal muscle atrophy and provide new insight into the way that skeletal muscle atrophy occurs at the molecular level.


Subject(s)
Cell Cycle Proteins/metabolism , MAP Kinase Kinase Kinase 4/metabolism , Multiprotein Complexes/metabolism , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/metabolism , Nuclear Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , MAP Kinase Kinase Kinase 4/genetics , Mice , Multiprotein Complexes/genetics , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Nuclear Proteins/genetics
17.
J Biol Chem ; 290(42): 25497-511, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26338703

ABSTRACT

Aging reduces skeletal muscle mass and strength, but the underlying molecular mechanisms remain elusive. Here, we used mouse models to investigate molecular mechanisms of age-related skeletal muscle weakness and atrophy as well as new potential interventions for these conditions. We identified two small molecules that significantly reduce age-related deficits in skeletal muscle strength, quality, and mass: ursolic acid (a pentacyclic triterpenoid found in apples) and tomatidine (a steroidal alkaloid derived from green tomatoes). Because small molecule inhibitors can sometimes provide mechanistic insight into disease processes, we used ursolic acid and tomatidine to investigate the pathogenesis of age-related muscle weakness and atrophy. We found that ursolic acid and tomatidine generate hundreds of small positive and negative changes in mRNA levels in aged skeletal muscle, and the mRNA expression signatures of the two compounds are remarkably similar. Interestingly, a subset of the mRNAs repressed by ursolic acid and tomatidine in aged muscle are positively regulated by activating transcription factor 4 (ATF4). Based on this finding, we investigated ATF4 as a potential mediator of age-related muscle weakness and atrophy. We found that a targeted reduction in skeletal muscle ATF4 expression reduces age-related deficits in skeletal muscle strength, quality, and mass, similar to ursolic acid and tomatidine. These results elucidate ATF4 as a critical mediator of age-related muscle weakness and atrophy. In addition, these results identify ursolic acid and tomatidine as potential agents and/or lead compounds for reducing ATF4 activity, weakness, and atrophy in aged skeletal muscle.


Subject(s)
Activating Transcription Factor 4/antagonists & inhibitors , Aging/pathology , Muscle, Skeletal/metabolism , Sarcopenia/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/physiology , Animals , Gene Expression/drug effects , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , RNA, Messenger/genetics , Sarcopenia/pathology , Sarcopenia/prevention & control , Tomatine/analogs & derivatives , Tomatine/pharmacology , Triterpenes/pharmacology , Ursolic Acid
18.
Curr Opin Clin Nutr Metab Care ; 18(3): 263-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25807353

ABSTRACT

PURPOSE OF REVIEW: Here, we discuss a recently developed experimental strategy for discovering small molecules with potential to prevent and treat skeletal muscle atrophy. RECENT FINDINGS: Muscle atrophy involves and requires widespread changes in skeletal muscle gene expression, which generate complex but measurable patterns of positive and negative changes in skeletal muscle mRNA levels (a.k.a. mRNA expression signatures of muscle atrophy). Many bioactive small molecules generate their own characteristic mRNA expression signatures, and by identifying small molecules whose signatures approximate mirror images of muscle atrophy signatures, one may identify small molecules with potential to prevent and/or reverse muscle atrophy. Unlike a conventional drug discovery approach, this strategy does not rely on a predefined molecular target but rather exploits the complexity of muscle atrophy to identify small molecules that counter the entire spectrum of pathological changes in atrophic muscle. We discuss how this strategy has been used to identify two natural compounds, ursolic acid and tomatidine, that reduce muscle atrophy and improve skeletal muscle function. SUMMARY: Discovery strategies based on mRNA expression signatures can elucidate new approaches for preserving and restoring muscle mass and function.


Subject(s)
Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/drug therapy , RNA, Messenger/metabolism , Tomatine/analogs & derivatives , Triterpenes/therapeutic use , Drug Discovery/methods , Gene Expression , Humans , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Signal Transduction , Tomatine/therapeutic use , Ursolic Acid
19.
Am J Physiol Endocrinol Metab ; 308(2): E144-58, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25406264

ABSTRACT

Skeletal muscle atrophy is a common and debilitating condition that remains poorly understood at the molecular level. To better understand the mechanisms of muscle atrophy, we used mouse models to search for a skeletal muscle protein that helps to maintain muscle mass and is specifically lost during muscle atrophy. We discovered that diverse causes of muscle atrophy (limb immobilization, fasting, muscle denervation, and aging) strongly reduced expression of the enzyme spermine oxidase. Importantly, a reduction in spermine oxidase was sufficient to induce muscle fiber atrophy. Conversely, forced expression of spermine oxidase increased muscle fiber size in multiple models of muscle atrophy (immobilization, fasting, and denervation). Interestingly, the reduction of spermine oxidase during muscle atrophy was mediated by p21, a protein that is highly induced during muscle atrophy and actively promotes muscle atrophy. In addition, we found that spermine oxidase decreased skeletal muscle mRNAs that promote muscle atrophy (e.g., myogenin) and increased mRNAs that help to maintain muscle mass (e.g., mitofusin-2). Thus, in healthy skeletal muscle, a relatively low level of p21 permits expression of spermine oxidase, which helps to maintain basal muscle gene expression and fiber size; conversely, during conditions that cause muscle atrophy, p21 expression rises, leading to reduced spermine oxidase expression, disruption of basal muscle gene expression, and muscle fiber atrophy. Collectively, these results identify spermine oxidase as an important positive regulator of muscle gene expression and fiber size, and elucidate p21-mediated repression of spermine oxidase as a key step in the pathogenesis of skeletal muscle atrophy.


Subject(s)
GTP Phosphohydrolases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/metabolism , Myogenin/metabolism , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Animals , Fasting/physiology , GTP Phosphohydrolases/genetics , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Muscle Denervation , Muscle Fibers, Skeletal/enzymology , Muscular Atrophy/enzymology , Muscular Atrophy/genetics , Myogenin/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Restraint, Physical/physiology , Polyamine Oxidase
20.
Am J Physiol Endocrinol Metab ; 307(3): E245-61, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24895282

ABSTRACT

Immobilization causes skeletal muscle atrophy via complex signaling pathways that are not well understood. To better understand these pathways, we investigated the roles of p53 and ATF4, two transcription factors that mediate adaptations to a variety of cellular stresses. Using mouse models, we demonstrate that 3 days of muscle immobilization induces muscle atrophy and increases expression of p53 and ATF4. Furthermore, muscle fibers lacking p53 or ATF4 are partially resistant to immobilization-induced muscle atrophy, and forced expression of p53 or ATF4 induces muscle fiber atrophy in the absence of immobilization. Importantly, however, p53 and ATF4 do not require each other to promote atrophy, and coexpression of p53 and ATF4 induces more atrophy than either transcription factor alone. Moreover, muscle fibers lacking both p53 and ATF4 are more resistant to immobilization-induced atrophy than fibers lacking only p53 or ATF4. Interestingly, the independent and additive nature of the p53 and ATF4 pathways allows for combinatorial control of at least one downstream effector, p21. Using genome-wide mRNA expression arrays, we identified p21 mRNA as a skeletal muscle transcript that is highly induced in immobilized muscle via the combined actions of p53 and ATF4. Additionally, in mouse muscle, p21 induces atrophy in a manner that does not require immobilization, p53 or ATF4, and p21 is required for atrophy induced by immobilization, p53, and ATF4. Collectively, these results identify p53 and ATF4 as essential and complementary mediators of immobilization-induced muscle atrophy and discover p21 as a critical downstream effector of the p53 and ATF4 pathways.


Subject(s)
Activating Transcription Factor 4/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Immobilization/adverse effects , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Activating Transcription Factor 4/genetics , Amino Acid Substitution , Animals , Crosses, Genetic , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , Disease Resistance , Genome-Wide Association Study , Hindlimb Suspension , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Mutant Proteins/metabolism , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...