201
|
Kang C, Goodman CA, Hornberger TA, Ji LL. PGC-1α overexpression by in vivo transfection attenuates mitochondrial deterioration of skeletal muscle caused by immobilization. FASEB J 2015; 29:4092-106. [PMID: 26178167 DOI: 10.1096/fj.14-266619] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/20/2015] [Indexed: 12/29/2022]
Abstract
Prolonged immobilization (IM) causes skeletal muscle atrophy characterized by mitochondrial deterioration and proteolysis. Muscle remobilization (RM) increases reactive oxygen species generation, proinflammatory cytokine expression, and oxidative stress, preventing muscle from quick recovery. Thus, we hypothesized that overexpression of peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) via in vivo transfection would promote mitochondrial biogenesis and antioxidant defense, thus ameliorating the aforementioned deteriorations in a mouse model with 14-d IM followed by 5-d RM. PGC-1α transfection in tibialis anterior muscle resulted in a 7.2- and 4-fold increase in PGC-1α content in cytosol and nucleus, respectively. Mitochondrial biogenic (cytochrome c, mitochondrial transcription factor A), morphologic (mitochondrial density, mDNA/nDNA ratio), and functional (cytochrome c oxidase activity, ATP synthesis rate) markers, as well as fiber cross-sectional area, significantly increased in IM-RM muscle by PGC-1α overexpression. These effects were accompanied by an 18% decrease in H2O2, 30% decrease in nuclear factor-κB-DNA binding, and 25% reduction of IL-1β and-6 production in IM-RM muscle. There was a 34% increase in superoxide dismutase-2 activity, along with a 3.5-fold increase in NAD-dependent deacetylase sirtuin-3 expression caused by enhanced PGC-1α-estrogen-related receptor α binding. Our findings highlighted the importance of PGC-1α in protecting muscle from metabolic and redox disturbances caused by IM.
Collapse
Affiliation(s)
- Chounghun Kang
- *Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota at Twin Cities, Minneapolis, Minnesota, USA; and Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Craig A Goodman
- *Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota at Twin Cities, Minneapolis, Minnesota, USA; and Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Troy A Hornberger
- *Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota at Twin Cities, Minneapolis, Minnesota, USA; and Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Li Li Ji
- *Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota at Twin Cities, Minneapolis, Minnesota, USA; and Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
202
|
Tintignac LA, Brenner HR, Rüegg MA. Mechanisms Regulating Neuromuscular Junction Development and Function and Causes of Muscle Wasting. Physiol Rev 2015; 95:809-52. [DOI: 10.1152/physrev.00033.2014] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The neuromuscular junction is the chemical synapse between motor neurons and skeletal muscle fibers. It is designed to reliably convert the action potential from the presynaptic motor neuron into the contraction of the postsynaptic muscle fiber. Diseases that affect the neuromuscular junction may cause failure of this conversion and result in loss of ambulation and respiration. The loss of motor input also causes muscle wasting as muscle mass is constantly adapted to contractile needs by the balancing of protein synthesis and protein degradation. Finally, neuromuscular activity and muscle mass have a major impact on metabolic properties of the organisms. This review discusses the mechanisms involved in the development and maintenance of the neuromuscular junction, the consequences of and the mechanisms involved in its dysfunction, and its role in maintaining muscle mass during aging. As life expectancy is increasing, loss of muscle mass during aging, called sarcopenia, has emerged as a field of high medical need. Interestingly, aging is also accompanied by structural changes at the neuromuscular junction, suggesting that the mechanisms involved in neuromuscular junction maintenance might be disturbed during aging. In addition, there is now evidence that behavioral paradigms and signaling pathways that are involved in longevity also affect neuromuscular junction stability and sarcopenia.
Collapse
Affiliation(s)
- Lionel A. Tintignac
- Biozentrum, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland; and INRA, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France
| | - Hans-Rudolf Brenner
- Biozentrum, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland; and INRA, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France
| | - Markus A. Rüegg
- Biozentrum, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland; and INRA, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France
| |
Collapse
|
203
|
Tryon LD, Crilly MJ, Hood DA. Effect of denervation on the regulation of mitochondrial transcription factor A expression in skeletal muscle. Am J Physiol Cell Physiol 2015; 309:C228-38. [PMID: 26063705 DOI: 10.1152/ajpcell.00266.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 06/03/2015] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to determine how the expression of mitochondrial transcription factor A (Tfam), a protein that governs mitochondrial DNA (mtDNA) transcription and replication, is regulated during a state of reduced organelle content imposed by muscle disuse. We measured Tfam expression at 8 h, 16 h, 24 h, 3 days, or 7 days following denervation and hypothesized that decreases in Tfam expression would precede mitochondrial loss. Muscle mass was lowered by 13% and 38% at 3 and 7 days postdenervation, while cytochrome c oxidase activity fell by 33% and 39% at the same time points. Tfam promoter activation in vivo was reduced by 30-65% between 8 h and 3 days of denervation, while Tfam transcript half-life was increased following 8-24 h of denervation. Protein expression of RNA-binding proteins that promote mRNA degradation (CUG repeat-binding protein and K homology splicing regulator protein) was elevated at 3 and 7 days of denervation. Tfam localization within subsarcolemmal mitochondria was reduced after 3 and 7 days of denervation and was associated with suppression of the cytochrome c oxidase type I transcript at 3 days, indicating that denervation impairs both mitochondrial Tfam import and mtDNA transcription during an early period following denervation. These data suggest that putative signals downregulate Tfam transcription during the earliest stages following denervation but are counteracted by increases in Tfam mRNA stability. Import of Tfam into the mitochondrion seems to be the most critical point of regulation of this protein during the early onset of denervation, an impairment of which is coincident with the loss of mitochondria during muscle disuse.
Collapse
Affiliation(s)
- Liam D Tryon
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada; and School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Matthew J Crilly
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada; and School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - David A Hood
- Muscle Health Research Centre, York University, Toronto, Ontario, Canada; and School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
204
|
Barreiro E, Sznajder JI, Nader GA, Budinger GRS. Muscle dysfunction in patients with lung diseases: a growing epidemic. Am J Respir Crit Care Med 2015; 191:616-9. [PMID: 25767924 DOI: 10.1164/rccm.201412-2189oe] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Esther Barreiro
- 1 Respiratory Medicine Department-Lung Cancer Research Group, Institute of Medical Research of Hospital del Mar (IMIM)-Hospital del Mar, Parc de Salut Mar, Barcelona Biomedical Research Park, Barcelona, Spain
| | | | | | | |
Collapse
|
205
|
Ziaaldini MM, Koltai E, Csende Z, Goto S, Boldogh I, Taylor AW, Radak Z. Exercise training increases anabolic and attenuates catabolic and apoptotic processes in aged skeletal muscle of male rats. Exp Gerontol 2015; 67:9-14. [PMID: 25910622 DOI: 10.1016/j.exger.2015.04.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/18/2015] [Accepted: 04/18/2015] [Indexed: 12/20/2022]
Abstract
Aging results in significant loss of mass and function of the skeletal muscle, which negatively impacts the quality of life. In this study we investigated whether aerobic exercise training has the potential to alter anabolic and catabolic pathways in the skeletal muscle. Five and twenty eight month old rats were used in the study. Aging resulted in decreased levels of follistatin/mTOR/Akt/Erk activation and increased myostatin/Murf1/2, proteasome subunits, and protein ubiquitination levels. In addition, TNF-α, reactive oxygen species (ROS), p53, and Bax levels were increased while Bcl-2 levels were decreased in the skeletal muscle of aged rats. Six weeks of exercise training at 60% of VO2max reversed the age-associated activation of catabolic and apoptotic pathways and increased anabolic signaling. The results suggest that the age-associated loss of muscle mass and cachexia could be due to the orchestrated down-regulation of anabolic and up-regulation of catabolic and pro-apoptotic processes. These metabolic changes can be attenuated by exercise training.
Collapse
Affiliation(s)
| | - Erika Koltai
- Research Institute of Sport Science, Semmelweis University, Budapest, Hungary
| | - Zsolt Csende
- Department of Biomechanics, Semmelweis University, Budapest, Hungary
| | - Sataro Goto
- Department of Exercise Physiology, School of Health and Sport Science, Juntendo University, Chiba, Japan
| | - Istvan Boldogh
- Department of Microbiology and Immunology, Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Albert W Taylor
- Faculty of Health Sciences, The University of Western Ontario, London, Ontario, Canada
| | - Zsolt Radak
- Research Institute of Sport Science, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
206
|
Milan G, Romanello V, Pescatore F, Armani A, Paik JH, Frasson L, Seydel A, Zhao J, Abraham R, Goldberg AL, Blaauw B, DePinho RA, Sandri M. Regulation of autophagy and the ubiquitin-proteasome system by the FoxO transcriptional network during muscle atrophy. Nat Commun 2015; 6:6670. [PMID: 25858807 PMCID: PMC4403316 DOI: 10.1038/ncomms7670] [Citation(s) in RCA: 507] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 02/18/2015] [Indexed: 12/19/2022] Open
Abstract
Stresses like low nutrients, systemic inflammation, cancer or infections provoke a catabolic state characterized by enhanced muscle proteolysis and amino acid release to sustain liver gluconeogenesis and tissue protein synthesis. These conditions activate the family of Forkhead Box (Fox) O transcription factors. Here we report that muscle-specific deletion of FoxO members protects from muscle loss as a result of the role of FoxOs in the induction of autophagy–lysosome and ubiquitin–proteasome systems. Notably, in the setting of low nutrient signalling, we demonstrate that FoxOs are required for Akt activity but not for mTOR signalling. FoxOs control several stress–response pathways such as the unfolded protein response, ROS detoxification, DNA repair and translation. Finally, we identify FoxO-dependent ubiquitin ligases including MUSA1 and a previously uncharacterised ligase termed SMART (Specific of Muscle Atrophy and Regulated by Transcription). Our findings underscore the central function of FoxOs in coordinating a variety of stress-response genes during catabolic conditions. FoxO transcription factors promote muscle atrophy in response to stresses such as low nutrient availability. By generating muscle-specific FoxO triple-knockout mice, Milan et al. identify mechanisms by which the FoxO transcriptional network coordinates autophagic and proteasomal protein degradation.
Collapse
Affiliation(s)
- Giulia Milan
- Venetian Institute of Molecular Medicine, via Orus 2, 35129 Padova, Italy
| | - Vanina Romanello
- Venetian Institute of Molecular Medicine, via Orus 2, 35129 Padova, Italy
| | - Francesca Pescatore
- 1] Venetian Institute of Molecular Medicine, via Orus 2, 35129 Padova, Italy [2] Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Andrea Armani
- Venetian Institute of Molecular Medicine, via Orus 2, 35129 Padova, Italy
| | - Ji-Hye Paik
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York 10065, USA
| | - Laura Frasson
- Venetian Institute of Molecular Medicine, via Orus 2, 35129 Padova, Italy
| | - Anke Seydel
- Venetian Institute of Molecular Medicine, via Orus 2, 35129 Padova, Italy
| | - Jinghui Zhao
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Reimar Abraham
- Department of Cancer Biology, U3 Pharma GmbH, Martinsried 82152, Germany
| | - Alfred L Goldberg
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Bert Blaauw
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Marco Sandri
- 1] Venetian Institute of Molecular Medicine, via Orus 2, 35129 Padova, Italy [2] Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy [3] Institute of Neuroscience, Consiglio Nazionale delle Ricerche, 35129 Padova, Italy [4] Department of Medicine, McGill University, Montreal, Quebec H3AoG4, Canada [5] Dulbecco Telethon Institute at Telethon Institute of Genetics and Medicine (TIGEM), 80131 Napoli, Italy
| |
Collapse
|
207
|
Nakao R, Yamamoto S, Horikawa K, Yasumoto Y, Nikawa T, Mukai C, Oishi K. Atypical expression of circadian clock genes in denervated mouse skeletal muscle. Chronobiol Int 2015; 32:486-96. [PMID: 25798696 DOI: 10.3109/07420528.2014.1003350] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The central circadian clock in the suprachiasmatic nucleus of the hypothalamus synchronizes peripheral clocks through neural and humoral signals in most mammalian tissues. Here, we analyzed the effects of unilateral sciatic denervation on the expression of circadian clock- and clock-controlled genes in the gastrocnemius muscles of mice twice per day on days 0, 3, 7, 9, 11 and 14 after denervation and six times on each of days 7 and 28 after denervation to assess the regulation mechanism of the circadian clock in skeletal muscle. Sciatic denervation did not affect systemic circadian rhythms since core body temperature (Day 7), corticosterone secretion (Days 7 and 28), and hepatic clock gene expression remained intact (Days 7 and 28). Expression levels of most circadian clock-related genes such as Arntl, Per1, Rora, Nr1d1 and Dbp were reduced in accordance with the extent of muscle atrophy, although circadian Per2 expression was significantly augmented (Day 28). Cosinor analysis revealed that the circadian expression of Arntl (Days 7 and 28) and Dbp (Day 28) was phase advanced in denervated muscle. The mRNA expression of Clock was significantly increased in denervated muscle on Day 3 when the severe atrophy was absent, and it was not affected by atrophic progression for 28 days. Sciatic denervation did not affect the expression of these genes in the contralateral muscle (Days 7 and 28), suggesting that humoral changes were not involved in denervation-induced muscle clock disruption. We then analyzed genome-wide gene expression using microarrays to determine the effects of disrupting the molecular clock in muscle on circadian rhythms at Day 7. Among 478 circadian genes, 313 lost rhythmicity in the denervated muscles. These denervation-sensitive genes included the lipid metabolism-related genes, Nrip1, Bbs1, Ptgis, Acot1, Scd2, Hpgd, Insig1, Dhcr24, Ldlr and Mboat1. Our findings revealed that sciatic denervation disrupts the circadian expression of clock and clock-controlled genes either directly or indirectly via muscle atrophy in the gastrocnemius muscles of mice in a gene-specific manner.
Collapse
Affiliation(s)
- Reiko Nakao
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST) , Tsukuba, Ibaraki , Japan
| | | | | | | | | | | | | |
Collapse
|
208
|
Abstract
Atrophy occurs in specific muscles with inactivity (for example, during plaster cast immobilization) or denervation (for example, in patients with spinal cord injuries). Muscle wasting occurs systemically in older people (a condition known as sarcopenia); as a physiological response to fasting or malnutrition; and in many diseases, including chronic obstructive pulmonary disorder, cancer-associated cachexia, diabetes, renal failure, cardiac failure, Cushing syndrome, sepsis, burns and trauma. The rapid loss of muscle mass and strength primarily results from excessive protein breakdown, which is often accompanied by reduced protein synthesis. This loss of muscle function can lead to reduced quality of life, increased morbidity and mortality. Exercise is the only accepted approach to prevent or slow atrophy. However, several promising therapeutic agents are in development, and major advances in our understanding of the cellular mechanisms that regulate the protein balance in muscle include the identification of several cytokines, particularly myostatin, and a common transcriptional programme that promotes muscle wasting. Here, we discuss these new insights and the rationally designed therapies that are emerging to combat muscle wasting.
Collapse
|
209
|
Wiggs MP. Can endurance exercise preconditioning prevention disuse muscle atrophy? Front Physiol 2015; 6:63. [PMID: 25814955 PMCID: PMC4356230 DOI: 10.3389/fphys.2015.00063] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/17/2015] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence suggests that exercise training can provide a level of protection against disuse muscle atrophy. Endurance exercise training imposes oxidative, metabolic, and heat stress on skeletal muscle which activates a variety of cellular signaling pathways that ultimately leads to the increased expression of proteins that have been demonstrated to protect muscle from inactivity -induced atrophy. This review will highlight the effect of exercise-induced oxidative stress on endogenous enzymatic antioxidant capacity (i.e., superoxide dismutase, glutathione peroxidase, and catalase), the role of oxidative and metabolic stress on PGC1-α, and finally highlight the effect heat stress and HSP70 induction. Finally, this review will discuss the supporting scientific evidence that these proteins can attenuate muscle atrophy through exercise preconditioning.
Collapse
Affiliation(s)
- Michael P Wiggs
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida Gainesville, FL, USA
| |
Collapse
|
210
|
Halievski K, Mo K, Westwood JT, Monks DA. Transcriptional profile of muscle following acute induction of symptoms in a mouse model of Kennedy's disease/spinobulbar muscular atrophy. PLoS One 2015; 10:e0118120. [PMID: 25719894 PMCID: PMC4341878 DOI: 10.1371/journal.pone.0118120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 01/07/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Kennedy's disease/Spinobulbar muscular atrophy (KD/SBMA) is a degenerative neuromuscular disease affecting males. This disease is caused by polyglutamine expansion mutations of the androgen receptor (AR) gene. Although KD/SBMA has been traditionally considered a motor neuron disease, emerging evidence points to a central etiological role of muscle. We previously reported a microarray study of genes differentially expressed in muscle of three genetically unique mouse models of KD/SBMA but were unable to detect those which are androgen-dependent or are associated with onset of symptoms. METHODOLOGY/PRINCIPAL FINDINGS In the current study we examined the time course and androgen-dependence of transcriptional changes in the HSA-AR transgenic (Tg) mouse model, in which females have a severe phenotype after acute testosterone treatment. Using microarray analysis we identified differentially expressed genes at the onset and peak of muscle weakness in testosterone-treated Tg females. We found both transient and persistent groups of differentially expressed genes and analysis of gene function indicated functional groups such as mitochondrion, ion and nucleotide binding, muscle development, and sarcomere maintenance. CONCLUSIONS/SIGNIFICANCE By comparing the current results with those from the three previously reported models we were able to identify KD/SBMA candidate genes that are androgen dependent, and occur early in the disease process, properties which are promising for targeted therapeutics.
Collapse
Affiliation(s)
- Katherine Halievski
- Department of Psychology, University of Toronto Mississauga,
Mississauga, Ontario, Canada
| | - Kaiguo Mo
- Department of Psychology, University of Toronto Mississauga,
Mississauga, Ontario, Canada
| | - J. Timothy Westwood
- Department of Psychology, University of Toronto Mississauga,
Mississauga, Ontario, Canada
| | - Douglas A. Monks
- Department of Psychology, University of Toronto Mississauga,
Mississauga, Ontario, Canada
- * E-mail:
| |
Collapse
|
211
|
Braun TP, Marks DL. The regulation of muscle mass by endogenous glucocorticoids. Front Physiol 2015; 6:12. [PMID: 25691871 PMCID: PMC4315033 DOI: 10.3389/fphys.2015.00012] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/08/2015] [Indexed: 01/16/2023] Open
Abstract
Glucocorticoids are highly conserved fundamental regulators of energy homeostasis. In response to stress in the form of perceived danger or acute inflammation, glucocorticoids are released from the adrenal gland, rapidly mobilizing energy from carbohydrate, fat and protein stores. In the case of inflammation, mobilized protein is critical for the rapid synthesis of acute phase reactants and an efficient immune response to infection. While adaptive in response to infection, chronic mobilization can lead to a profound depletion of energy stores. Skeletal muscle represents the major body store of protein, and can become substantially atrophied under conditions of chronic inflammation. Glucocorticoids elicit the atrophy of muscle by increasing the rate of protein degradation by the ubiquitin-proteasome system and autophagy lysosome system. Protein synthesis is also suppressed at the level of translational initiation, preventing the production of new myofibrillar protein. Glucocorticoids also antagonize the action of anabolic regulators such as insulin further exacerbating the loss of protein and muscle mass. The loss of muscle mass in the context of chronic disease is a key feature of cachexia and contributes substantially to morbidity and mortality. A growing body of evidence demonstrates that glucocorticoid signaling is a common mediator of wasting, irrespective of the underlying initiator or disease state. This review will highlight fundamental mechanisms of glucocorticoid signaling and detail the mechanisms of glucocorticoid-induced muscle atrophy. Additionally, the evidence for glucocorticoids as a driver of muscle wasting in numerous disease states will be discussed. Given the burden of wasting diseases and the nodal nature of glucocorticoid signaling, effective anti-glucocorticoid therapy would be a valuable clinical tool. Therefore, the progress and potential pitfalls in the development of glucocorticoid antagonists for muscle wasting will be discussed.
Collapse
Affiliation(s)
- Theodore P Braun
- Department of Internal Medicine, University of Washington Medical Center Seattle, WA, USA ; Papé Family Pediatric Research Institute, Oregon Health and Science University Portland, OR, USA
| | - Daniel L Marks
- Department of Internal Medicine, University of Washington Medical Center Seattle, WA, USA
| |
Collapse
|
212
|
Bongers KS, Fox DK, Kunkel SD, Stebounova LV, Murry DJ, Pufall MA, Ebert SM, Dyle MC, Bullard SA, Dierdorff JM, Adams CM. Spermine oxidase maintains basal skeletal muscle gene expression and fiber size and is strongly repressed by conditions that cause skeletal muscle atrophy. Am J Physiol Endocrinol Metab 2015; 308:E144-58. [PMID: 25406264 PMCID: PMC4297781 DOI: 10.1152/ajpendo.00472.2014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 11/13/2014] [Indexed: 01/02/2023]
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.
Collapse
Affiliation(s)
- Kale S Bongers
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine
| | - Daniel K Fox
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine
| | - Steven D Kunkel
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine
| | | | - Daryl J Murry
- College of Pharmacy, Roy J. and Lucille A. Carver College of Medicine
| | - Miles A Pufall
- Department of Biochemistry, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa, and
| | - Scott M Ebert
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine
| | - Michael C Dyle
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine
| | - Steven A Bullard
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine, Iowa City Veterans Affairs Medical Center, Iowa City, Iowa
| | - Jason M Dierdorff
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine
| | - Christopher M Adams
- Departments of Internal Medicine and Molecular Physiology and Biophysics and Fraternal Order of Eagles Diabetes Research Center, Roy J. and Lucille A. Carver College of Medicine, Iowa City Veterans Affairs Medical Center, Iowa City, Iowa
| |
Collapse
|
213
|
Villena JA. New insights into PGC-1 coactivators: redefining their role in the regulation of mitochondrial function and beyond. FEBS J 2015; 282:647-72. [DOI: 10.1111/febs.13175] [Citation(s) in RCA: 252] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/31/2014] [Accepted: 12/10/2014] [Indexed: 12/25/2022]
Affiliation(s)
- Josep A. Villena
- Laboratory of Metabolism and Obesity; Vall d'Hebron-Institut de Recerca; Universitat Autònoma de Barcelona; Spain
- CIBERDEM (CIBER de Diabetes y Enfermedades Metabólicas Asociadas); Instituto de Salud Carlos III; Barcelona Spain
| |
Collapse
|
214
|
Qin W, Pan J, Wu Y, Bauman WA, Cardozo C. Anabolic steroids activate calcineurin-NFAT signaling and thereby increase myotube size and reduce denervation atrophy. Mol Cell Endocrinol 2015; 399:336-45. [PMID: 25450864 DOI: 10.1016/j.mce.2014.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/22/2014] [Accepted: 09/24/2014] [Indexed: 01/11/2023]
Abstract
Anabolic androgens have been shown to reduce muscle loss due to immobilization, paralysis and many other medical conditions, but the molecular basis for these actions is poorly understood. We have recently demonstrated that nandrolone, a synthetic androgen, slows muscle atrophy after nerve transection associated with down-regulation of regulator of calcineurin 2 (RCAN2), a calcineurin inhibitor, suggesting a possible role of calcineurin-NFAT signaling. To test this possibility, rat gastrocnemius muscle was analyzed at 56 days after denervation. In denervated muscle, calcineurin activity declined and NFATc4 was excluded from the nucleus and these effects were reversed by nandrolone. Similarly, nandrolone increased calcineurin activity and nuclear NFATc4 levels in cultured L6 myotubes. Nandrolone also induced cell hypertrophy that was blocked by cyclosporin A or overexpression of RCAN2. Finally protection against denervation atrophy by nandrolone in rats was blocked by cyclosporin A. These results demonstrate for the first time that nandrolone activates calcineurin-NFAT signaling, and that such signaling is important in nandrolone-induced cell hypertrophy and protection against paralysis-induced muscle atrophy.
Collapse
Affiliation(s)
- Weiping Qin
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, Bronx, New York 10468, USA; Departments of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA.
| | - Jiangping Pan
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, Bronx, New York 10468, USA
| | - Yong Wu
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, Bronx, New York 10468, USA
| | - William A Bauman
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, Bronx, New York 10468, USA; Departments of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA; Rehabilitation Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA
| | - Christopher Cardozo
- Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, Bronx, New York 10468, USA; Departments of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA; Rehabilitation Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA.
| |
Collapse
|
215
|
Parry SM, Puthucheary ZA. The impact of extended bed rest on the musculoskeletal system in the critical care environment. EXTREME PHYSIOLOGY & MEDICINE 2015; 4:16. [PMID: 26457181 PMCID: PMC4600281 DOI: 10.1186/s13728-015-0036-7] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/30/2015] [Indexed: 04/14/2023]
Abstract
Prolonged immobility is harmful with rapid reductions in muscle mass, bone mineral density and impairment in other body systems evident within the first week of bed rest which is further exacerbated in individuals with critical illness. Our understanding of the aetiology and secondary consequences of prolonged immobilization in the critically ill is improving with recent and ongoing research to establish the cause, effect, and best treatment options. This review aims to describe the current literature on bed rest models for examining immobilization-induced changes in the musculoskeletal system and pathophysiology of immobilisation in critical illness including examination of intracellular signalling processes involved. Finally, the review examines the current barriers to early activity and mobilization and potential rehabilitation strategies, which are being, investigated which may reverse the effects of prolonged bed rest. Addressing the deleterious effects of immobilization is a major step in treatment and prevention of the public health issue, that is, critical illness survivorship.
Collapse
Affiliation(s)
- Selina M. Parry
- />Department of Physiotherapy, School of Health Sciences, The University of Melbourne, Level 7 Alan Gilbert Building, Parkville, Melbourne, VIC 3010 Australia
| | - Zudin A. Puthucheary
- />Division of Respiratory and Critical Care Medicine, National University Health System, Singapore, Singapore
- />Institute of Health and Human Performance, University College London, London, UK
| |
Collapse
|
216
|
Sepulveda PV, Bush ED, Baar K. Pharmacology of manipulating lean body mass. Clin Exp Pharmacol Physiol 2015; 42:1-13. [PMID: 25311629 PMCID: PMC4383600 DOI: 10.1111/1440-1681.12320] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 01/04/2023]
Abstract
Dysfunction and wasting of skeletal muscle as a consequence of illness decreases the length and quality of life. Currently, there are few, if any, effective treatments available to address these conditions. Hence, the existence of this unmet medical need has fuelled large scientific efforts. Fortunately, these efforts have shown many of the underlying mechanisms adversely affecting skeletal muscle health. With increased understanding have come breakthrough disease-specific and broad spectrum interventions, some progressing through clinical development. The present review focuses its attention on the role of the antagonistic process regulating skeletal muscle mass before branching into prospective promising therapeutic targets and interventions. Special attention is given to therapies in development against cancer cachexia and Duchenne muscular dystrophy before closing remarks on design and conceptualization of future therapies are presented to the reader.
Collapse
Affiliation(s)
- Patricio V Sepulveda
- Department of Physiology, Monash University, Monash College Wellington Rd, Melbourne Victoria, Australia
| | - Ernest D Bush
- Akashi Therapeutics, Cambridge, MA, University of California Davis, Davis, CA, USA
| | - Keith Baar
- Departments of Neurobiology, Physiology and Behaviour and Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
217
|
Judge SM, Wu CL, Beharry AW, Roberts BM, Ferreira LF, Kandarian SC, Judge AR. Genome-wide identification of FoxO-dependent gene networks in skeletal muscle during C26 cancer cachexia. BMC Cancer 2014; 14:997. [PMID: 25539728 PMCID: PMC4391468 DOI: 10.1186/1471-2407-14-997] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/11/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Evidence from cachectic cancer patients and animal models of cancer cachexia supports the involvement of Forkhead box O (FoxO) transcription factors in driving cancer-induced skeletal muscle wasting. However, the genome-wide gene networks and associated biological processes regulated by FoxO during cancer cachexia are unknown. We hypothesize that FoxO is a central upstream regulator of diverse gene networks in skeletal muscle during cancer that may act coordinately to promote the wasting phenotype. METHODS To inhibit endogenous FoxO DNA-binding, we transduced limb and diaphragm muscles of mice with AAV9 containing the cDNA for a dominant negative (d.n.) FoxO protein (or GFP control). The d.n.FoxO construct consists of only the FoxO3a DNA-binding domain that is highly homologous to that of FoxO1 and FoxO4, and which outcompetes and blocks endogenous FoxO DNA binding. Mice were subsequently inoculated with Colon-26 (C26) cells and muscles harvested 26 days later. RESULTS Blocking FoxO prevented C26-induced muscle fiber atrophy of both locomotor muscles and the diaphragm and significantly spared force deficits. This sparing of muscle size and function was associated with the differential regulation of 543 transcripts (out of 2,093) which changed in response to C26. Bioinformatics analysis of upregulated gene transcripts that required FoxO revealed enrichment of the proteasome, AP-1 and IL-6 pathways, and included several atrophy-related transcription factors, including Stat3, Fos, and Cebpb. FoxO was also necessary for the cancer-induced downregulation of several gene transcripts that were enriched for extracellular matrix and sarcomere protein-encoding genes. We validated these findings in limb muscles and the diaphragm through qRT-PCR, and further demonstrate that FoxO1 and/or FoxO3a are sufficient to increase Stat3, Fos, Cebpb, and the C/EBPβ target gene, Ubr2. Analysis of the Cebpb proximal promoter revealed two bona fide FoxO binding elements, which we further establish are necessary for Cebpb promoter activation in response to IL-6, a predominant cytokine in the C26 cancer model. CONCLUSIONS These findings provide new evidence that FoxO-dependent transcription is a central node controlling diverse gene networks in skeletal muscle during cancer cachexia, and identifies novel candidate genes and networks for further investigation as causative factors in cancer-induced wasting.
Collapse
Affiliation(s)
- Sarah M Judge
- Department of Physical Therapy, University of Florida, 1225 Center Drive, HPNP Building 1142, Gainesville, Florida, USA.
| | - Chia-Ling Wu
- Department of Health Sciences, Boston University, Boston, Massachusetts, USA.
| | - Adam W Beharry
- Department of Physical Therapy, University of Florida, 1225 Center Drive, HPNP Building 1142, Gainesville, Florida, USA.
| | - Brandon M Roberts
- Department of Physical Therapy, University of Florida, 1225 Center Drive, HPNP Building 1142, Gainesville, Florida, USA.
| | - Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA.
| | - Susan C Kandarian
- Department of Health Sciences, Boston University, Boston, Massachusetts, USA.
| | - Andrew R Judge
- Department of Physical Therapy, University of Florida, 1225 Center Drive, HPNP Building 1142, Gainesville, Florida, USA.
| |
Collapse
|
218
|
Nakao R, Yamamoto S, Yasumoto Y, Kadota K, Oishi K. Impact of denervation-induced muscle atrophy on housekeeping gene expression in mice. Muscle Nerve 2014; 51:276-81. [DOI: 10.1002/mus.24310] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2014] [Indexed: 01/30/2023]
Affiliation(s)
- Reiko Nakao
- Biological Clock Research Group, Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Saori Yamamoto
- Biological Clock Research Group, Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
| | - Yuki Yasumoto
- Biological Clock Research Group, Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
- Department of Applied Biological Science, Graduate School of Science and Technology; Tokyo University of Science; Chiba Japan
| | - Koji Kadota
- Agricultural Bioinformatics Research Unit, Graduate School of Agricultural and Life Sciences; University of Tokyo; Tokyo Japan
| | - Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute; National Institute of Advanced Industrial Science and Technology; Central 6, 1-1-1 Higashi Tsukuba Ibaraki 305-8566 Japan
- Department of Applied Biological Science, Graduate School of Science and Technology; Tokyo University of Science; Chiba Japan
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences; University of Tokyo; Kashiwa Chiba Japan
| |
Collapse
|
219
|
Muscle-specific GSK-3β ablation accelerates regeneration of disuse-atrophied skeletal muscle. Biochim Biophys Acta Mol Basis Dis 2014; 1852:490-506. [PMID: 25496993 DOI: 10.1016/j.bbadis.2014.12.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 11/26/2014] [Accepted: 12/03/2014] [Indexed: 01/08/2023]
Abstract
Muscle wasting impairs physical performance, increases mortality and reduces medical intervention efficacy in chronic diseases and cancer. Developing proficient intervention strategies requires improved understanding of the molecular mechanisms governing muscle mass wasting and recovery. Involvement of muscle protein- and myonuclear turnover during recovery from muscle atrophy has received limited attention. The insulin-like growth factor (IGF)-I signaling pathway has been implicated in muscle mass regulation. As glycogen synthase kinase 3 (GSK-3) is inhibited by IGF-I signaling, we hypothesized that muscle-specific GSK-3β deletion facilitates the recovery of disuse-atrophied skeletal muscle. Wild-type mice and mice lacking muscle GSK-3β (MGSK-3β KO) were subjected to a hindlimb suspension model of reversible disuse-induced muscle atrophy and followed during recovery. Indices of muscle mass, protein synthesis and proteolysis, and post-natal myogenesis which contribute to myonuclear accretion, were monitored during the reloading of atrophied muscle. Early muscle mass recovery occurred more rapidly in MGSK-3β KO muscle. Reloading-associated changes in muscle protein turnover were not affected by GSK-3β ablation. However, coherent effects were observed in the extent and kinetics of satellite cell activation, proliferation and myogenic differentiation observed during reloading, suggestive of increased myonuclear accretion in regenerating skeletal muscle lacking GSK-3β. This study demonstrates that muscle mass recovery and post-natal myogenesis from disuse-atrophy are accelerated in the absence of GSK-3β.
Collapse
|
220
|
Jesinkey SR, Korrapati MC, Rasbach KA, Beeson CC, Schnellmann RG. Atomoxetine prevents dexamethasone-induced skeletal muscle atrophy in mice. J Pharmacol Exp Ther 2014; 351:663-73. [PMID: 25292181 PMCID: PMC4244586 DOI: 10.1124/jpet.114.217380] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 10/06/2014] [Indexed: 01/12/2023] Open
Abstract
Skeletal muscle atrophy remains a clinical problem in numerous pathologic conditions. β2-Adrenergic receptor agonists, such as formoterol, can induce mitochondrial biogenesis (MB) to prevent such atrophy. Additionally, atomoxetine, an FDA-approved norepinephrine reuptake inhibitor, was positive in a cellular assay for MB. We used a mouse model of dexamethasone-induced skeletal muscle atrophy to investigate the potential role of atomoxetine and formoterol to prevent muscle mass loss. Mice were administered dexamethasone once daily in the presence or absence of formoterol (0.3 mg/kg), atomoxetine (0.1 mg/kg), or sterile saline. Animals were euthanized at 8, 16, and 24 hours or 8 days later. Gastrocnemius muscle weights, changes in mRNA and protein expression of peroxisome proliferator-activated receptor-γ coactivator-1 α (PGC-1α) isoforms, ATP synthase β, cytochrome c oxidase subunit I, NADH dehydrogenase (ubiquinone) 1 β subcomplex, 8, ND1, insulin-like growth factor 1 (IGF-1), myostatin, muscle Ring-finger protein-1 (muscle atrophy), phosphorylated forkhead box protein O 3a (p-FoxO3a), Akt, mammalian target of rapamycin (mTOR), and ribosomal protein S6 (rp-S6; muscle hypertrophy) in naive and muscle-atrophied mice were measured. Atomoxetine increased p-mTOR 24 hours after treatment in naïve mice, but did not change any other biomarkers. Formoterol robustly activated the PGC-1α-4-IGF1-Akt-mTOR-rp-S6 pathway and increased p-FoxO3a as early as 8 hours and repressed myostatin at 16 hours. In contrast to what was observed with acute treatment, chronic treatment (7 days) with atomoxetine increased p-Akt and p-FoxO3a, and sustained PGC-1α expression and skeletal muscle mass in dexamethasone-treated mice, in a manner comparable to formoterol. In conclusion, chronic treatment with a low dose of atomoxetine prevented dexamethasone-induced skeletal muscle wasting and supports a potential role in preventing muscle atrophy.
Collapse
Affiliation(s)
- Sean R Jesinkey
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (S.R.J., M.C.K., K.A.R., C.C.B., R.G.S.); and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| | - Midhun C Korrapati
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (S.R.J., M.C.K., K.A.R., C.C.B., R.G.S.); and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| | - Kyle A Rasbach
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (S.R.J., M.C.K., K.A.R., C.C.B., R.G.S.); and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| | - Craig C Beeson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (S.R.J., M.C.K., K.A.R., C.C.B., R.G.S.); and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| | - Rick G Schnellmann
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina (S.R.J., M.C.K., K.A.R., C.C.B., R.G.S.); and Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina (R.G.S.)
| |
Collapse
|
221
|
Bowman CJ, Ayer DE, Dynlacht BD. Foxk proteins repress the initiation of starvation-induced atrophy and autophagy programs. Nat Cell Biol 2014; 16:1202-14. [PMID: 25402684 PMCID: PMC4250422 DOI: 10.1038/ncb3062] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 10/13/2014] [Indexed: 12/14/2022]
Abstract
Autophagy is the primary catabolic process triggered in response to starvation. Although autophagic regulation within the cytosolic compartment is well established, it is becoming clear that nuclear events also regulate the induction or repression of autophagy. Nevertheless, a thorough understanding of the mechanisms by which sequence-specific transcription factors modulate expression of genes required for autophagy is lacking. Here, we identify Foxk proteins (Foxk1 and Foxk2) as transcriptional repressors of autophagy in muscle cells and fibroblasts. Interestingly, Foxk1/2 serve to counter-balance another forkhead transcription factor, Foxo3, which induces an overlapping set of autophagic and atrophic targets in muscle. Foxk1/2 specifically recruits Sin3A-HDAC complexes to restrict acetylation of histone H4 and expression of critical autophagy genes. Remarkably, mTOR promotes the transcriptional activity of Foxk1 by facilitating nuclear entry to specifically limit basal levels of autophagy in nutrient-rich conditions. Our study highlights an ancient, conserved mechanism whereby nutritional status is interpreted by mTOR to restrict autophagy by repressing essential autophagy genes via Foxk-Sin3-mediated transcriptional control.
Collapse
Affiliation(s)
- Christopher John Bowman
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, New York 10016, USA
| | - Donald E Ayer
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Brian David Dynlacht
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, New York 10016, USA
| |
Collapse
|
222
|
Watts FZ, Baldock R, Jongjitwimol J, Morley SJ. Weighing up the possibilities: Controlling translation by ubiquitylation and sumoylation. ACTA ACUST UNITED AC 2014; 2:e959366. [PMID: 26779408 DOI: 10.4161/2169074x.2014.959366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/01/2014] [Accepted: 05/12/2014] [Indexed: 12/15/2022]
Abstract
Regulation of protein synthesis is of fundamental importance to cells. It has a critical role in the control of gene expression, and consequently cell growth and proliferation. The importance of this control is supported by the fact that protein synthesis is frequently upregulated in tumor cells. The major point at which regulation occurs is the initiation stage. Initiation of translation involves the interaction of several proteins to form the eIF4F complex, the recognition of the mRNA by this complex, and the subsequent recruitment of the 40S ribosomal subunit to the mRNA. This results in the formation of the 48S complex that then scans the mRNA for the start codon, engages the methionyl-tRNA and eventually forms the mature 80S ribosome which is elongation-competent. Formation of the 48S complex is regulated by the availability of individual initiation factors and through specific protein-protein interactions. Both of these events can be regulated by post-translational modification by ubiquitin or Ubls (ubiquitin-like modifiers) such as SUMO or ISG15. We provide here a summary of translation initiation factors that are modified by ubiquitin or Ubls and, where they have been studied in detail, describe the role of these modifications and their effects on regulating protein synthesis.
Collapse
Affiliation(s)
- Felicity Z Watts
- Genome Damage and Stability Center; School of Life Sciences; University of Sussex ; Falmer, Brighton, UK
| | - Robert Baldock
- Genome Damage and Stability Center; School of Life Sciences; University of Sussex ; Falmer, Brighton, UK
| | - Jirapas Jongjitwimol
- Genome Damage and Stability Center; School of Life Sciences; University of Sussex ; Falmer, Brighton, UK
| | - Simon J Morley
- Department of Biochemistry and Biomedical Science; School of Life Sciences; University of Sussex ; Brighton, UK
| |
Collapse
|
223
|
Fjällström AK, Evertsson K, Norrby M, Tågerud S. Forkhead box O1 and muscle RING finger 1 protein expression in atrophic and hypertrophic denervated mouse skeletal muscle. J Mol Signal 2014; 9:9. [PMID: 25276226 PMCID: PMC4177715 DOI: 10.1186/1750-2187-9-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 09/18/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Forkhead box O (FoxO) transcription factors and E3 ubiquitin ligases such as Muscle RING finger 1 (MuRF1) are believed to participate in the regulation of skeletal muscle mass. The function of FoxO transcription factors is regulated by post-translational modifications such as phosphorylation and acetylation. In the present study FoxO1 protein expression, phosphorylation and acetylation as well as MuRF1 protein expression, were examined in atrophic and hypertrophic denervated skeletal muscle. METHODS Protein expression, phosphorylation and acetylation were studied semi-quantitatively using Western blots. Muscles studied were 6-days denervated mouse hind-limb muscles (anterior tibial as well as pooled gastrocnemius and soleus muscles, all atrophic), 6-days denervated mouse hemidiaphragm muscles (hypertrophic) and innervated control muscles. Total muscle homogenates were used as well as separated nuclear and cytosolic fractions of innervated and 6-days denervated anterior tibial and hemidiaphragm muscles. RESULTS Expression of FoxO1 and MuRF1 proteins increased 0.3-3.7-fold in all 6-days denervated muscles studied, atrophic as well as hypertrophic. Phosphorylation of FoxO1 at S256 increased about 0.8-1-fold after denervation in pooled gastrocnemius and soleus muscles and in hemidiaphragm but not in unfractionated anterior tibial muscle. A small (0.2-fold) but statistically significant increase in FoxO1 phosphorylation was, however, observed in cytosolic fractions of denervated anterior tibial muscle. A statistically significant increase in FoxO1 acetylation (0.8-fold) was observed only in denervated anterior tibial muscle. Increases in total FoxO1 and in phosphorylated FoxO1 were only seen in cytosolic fractions of denervated atrophic anterior tibial muscle whereas in denervated hypertrophic hemidiaphragm both total FoxO1 and phosphorylated FoxO1 increased in cytosolic as well as in nuclear fractions. MuRF1 protein expression increased in cytosolic as well as in nuclear fractions of both denervated atrophic anterior tibial muscle and denervated hypertrophic hemidiaphragm muscle. CONCLUSIONS Increased expression of FoxO1 and MuRF1 in denervated muscles (atrophic as well as hypertrophic) suggests that these proteins participate in the tissue remodelling occurring after denervation. The effect of denervation on the level of phosphorylated and acetylated FoxO1 differed in the muscles studied and may be related to differences in fiber type composition of the muscles.
Collapse
Affiliation(s)
- Ann-Kristin Fjällström
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar SE-391 82, Sweden
| | - Kim Evertsson
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar SE-391 82, Sweden
| | - Marlene Norrby
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar SE-391 82, Sweden
| | - Sven Tågerud
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar SE-391 82, Sweden
| |
Collapse
|
224
|
Abstract
Muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx)/atrogin-1 were identified more than 10 years ago as two muscle-specific E3 ubiquitin ligases that are increased transcriptionally in skeletal muscle under atrophy-inducing conditions, making them excellent markers of muscle atrophy. In the past 10 years much has been published about MuRF1 and MAFbx with respect to their mRNA expression patterns under atrophy-inducing conditions, their transcriptional regulation, and their putative substrates. However, much remains to be learned about the physiological role of both genes in the regulation of mass and other cellular functions in striated muscle. Although both MuRF1 and MAFbx are enriched in skeletal, cardiac, and smooth muscle, this review will focus on the current understanding of MuRF1 and MAFbx in skeletal muscle, highlighting the critical questions that remain to be answered.
Collapse
Affiliation(s)
- Sue C Bodine
- Departments of Neurobiology, Physiology, and Behavior and Physiology and Membrane Biology, University of California Davis, Davis, California; and Northern California Veterans Affairs Health Systems, Mather, California
| | - Leslie M Baehr
- Membrane Biology, University of California Davis, Davis, California; and
| |
Collapse
|
225
|
Recent advances in mitochondrial turnover during chronic muscle disuse. Integr Med Res 2014; 3:161-171. [PMID: 28664093 PMCID: PMC5481769 DOI: 10.1016/j.imr.2014.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/06/2014] [Indexed: 12/21/2022] Open
Abstract
Chronic muscle disuse, such as that resulting from immobilization, denervation, or prolonged physical inactivity, produces atrophy and a loss of mitochondria, yet the molecular relationship between these events is not fully understood. In this review we attempt to identify the key regulatory steps mediating the loss of muscle mass and the decline in mitochondrial content and function. An understanding of common intracellular signaling pathways may provide much-needed insight into the possible therapeutic targets for treatments that will maintain aerobic energy metabolism and preserve muscle mass during disuse conditions.
Collapse
|
226
|
Blanqué R, Lepescheux L, Auberval M, Minet D, Merciris D, Cottereaux C, Clément-Lacroix P, Delerive P, Namour F. Characterization of GLPG0492, a selective androgen receptor modulator, in a mouse model of hindlimb immobilization. BMC Musculoskelet Disord 2014; 15:291. [PMID: 25185887 PMCID: PMC4167280 DOI: 10.1186/1471-2474-15-291] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 08/27/2014] [Indexed: 11/23/2022] Open
Abstract
Background Muscle wasting is a hallmark of many chronic conditions but also of aging and results in a progressive functional decline leading ultimately to disability. Androgens, such as testosterone were proposed as therapy to counteract muscle atrophy. However, this treatment is associated with potential cardiovascular and prostate cancer risks and therefore not acceptable for long-term treatment. Selective Androgen receptor modulators (SARM) are androgen receptor ligands that induce muscle anabolism while having reduced effects in reproductive tissues. Therefore, they represent an alternative to testosterone therapy. Our objective was to demonstrate the activity of SARM molecule (GLPG0492) on a immobilization muscle atrophy mouse model as compared to testosterone propionate (TP) and to identify putative biomarkers in the plasma compartment that might be related to muscle function and potentially translated into the clinical space. Methods GLPG0492, a non-steroidal SARM, was evaluated and compared to TP in a mouse model of hindlimb immobilization. Results GLPG0492 treatment partially prevents immobilization-induced muscle atrophy with a trend to promote muscle fiber hypertrophy in a dose-dependent manner. Interestingly, GLPG0492 was found as efficacious as TP at reducing muscle loss while sparing reproductive tissues. Furthermore, gene expression studies performed on tibialis samples revealed that both GLPG0492 and TP were slowing down muscle loss by negatively interfering with major signaling pathways controlling muscle mass homeostasis. Finally, metabolomic profiling experiments using 1H-NMR led to the identification of a plasma GLPG0492 signature linked to the modulation of cellular bioenergetic processes. Conclusions Taken together, these results unveil the potential of GLPG0492, a non-steroidal SARM, as treatment for, at least, musculo-skeletal atrophy consecutive to coma, paralysis, or limb immobilization. Electronic supplementary material The online version of this article (doi:10.1186/1471-2474-15-291) contains supplementary material, which is available to authorized users.
Collapse
|
227
|
Cannavino J, Brocca L, Sandri M, Bottinelli R, Pellegrino MA. PGC1-α over-expression prevents metabolic alterations and soleus muscle atrophy in hindlimb unloaded mice. J Physiol 2014; 592:4575-89. [PMID: 25128574 DOI: 10.1113/jphysiol.2014.275545] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Prolonged skeletal muscle inactivity causes muscle fibre atrophy. Redox imbalance has been considered one of the major triggers of skeletal muscle disuse atrophy, but whether redox imbalance is actually the major cause or simply a consequence of muscle disuse remains of debate. Here we hypothesized that a metabolic stress mediated by PGC-1α down-regulation plays a major role in disuse atrophy. First we studied the adaptations of soleus to mice hindlimb unloading (HU) in the early phase of disuse (3 and 7 days of HU) with and without antioxidant treatment (trolox). HU caused a reduction in cross-sectional area, redox status alteration (NRF2, SOD1 and catalase up-regulation), and induction of the ubiquitin proteasome system (MuRF-1 and atrogin-1 mRNA up-regulation) and autophagy (Beclin1 and p62 mRNA up-regulation). Trolox completely prevented the induction of NRF2, SOD1 and catalase mRNAs, but not atrophy or induction of catabolic systems in unloaded muscles, suggesting that oxidative stress is not a major cause of disuse atrophy. HU mice showed a marked alteration of oxidative metabolism. PGC-1α and mitochondrial complexes were down-regulated and DRP1 was up-regulated. To define the link between mitochondrial dysfunction and disuse muscle atrophy we unloaded mice overexpressing PGC-1α. Transgenic PGC-1α animals did not show metabolic alteration during unloading, preserving muscle size through the reduction of autophagy and proteasome degradation. Our results indicate that mitochondrial dysfunction plays a major role in disuse atrophy and that compounds inducing PGC-1α expression could be useful to treat/prevent muscle atrophy.
Collapse
Affiliation(s)
- Jessica Cannavino
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy
| | - Lorenza Brocca
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy
| | - Marco Sandri
- Venetian Institute of Molecular Medicine and Dulbecco Telethon Institute, 35129, Padova, Italy Interuniversity Institute of Myology, University of Pavia, Pavia, Italy
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy Fondazione Salvatore Maugeri (IRCCS), Scientific Institute of Pavia, Pavia, Italy Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia, Italy
| | - Maria Antonietta Pellegrino
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy Interuniversity Institute of Myology, University of Pavia, Pavia, Italy Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
228
|
Stefanetti RJ, Lamon S, Wallace M, Vendelbo MH, Russell AP, Vissing K. Regulation of ubiquitin proteasome pathway molecular markers in response to endurance and resistance exercise and training. Pflugers Arch 2014; 467:1523-1537. [DOI: 10.1007/s00424-014-1587-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 07/02/2014] [Accepted: 07/24/2014] [Indexed: 12/30/2022]
|
229
|
Sei H, Taguchi A, Nishida N, Hato N, Gyo K. Expression of atrophy-related transcription factors in the process of intrinsic laryngeal muscle atrophy after denervation. Eur Arch Otorhinolaryngol 2014; 272:137-41. [DOI: 10.1007/s00405-014-3226-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 07/29/2014] [Indexed: 10/24/2022]
|
230
|
Fox DK, Ebert SM, Bongers KS, Dyle MC, Bullard SA, Dierdorff JM, Kunkel SD, Adams CM. p53 and ATF4 mediate distinct and additive pathways to skeletal muscle atrophy during limb immobilization. Am J Physiol Endocrinol Metab 2014; 307:E245-61. [PMID: 24895282 PMCID: PMC4121573 DOI: 10.1152/ajpendo.00010.2014] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
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.
Collapse
Affiliation(s)
- Daniel K Fox
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Scott M Ebert
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Kale S Bongers
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Michael C Dyle
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Steven A Bullard
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and Iowa City Veterans Affairs Medical Center, Iowa City, Iowa
| | - Jason M Dierdorff
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Steven D Kunkel
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and
| | - Christopher M Adams
- Departments of Internal Medicine and Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa; and Iowa City Veterans Affairs Medical Center, Iowa City, Iowa
| |
Collapse
|
231
|
Vitamin E in sarcopenia: current evidences on its role in prevention and treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:914853. [PMID: 25097722 PMCID: PMC4109111 DOI: 10.1155/2014/914853] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/06/2014] [Indexed: 01/01/2023]
Abstract
Sarcopenia is a geriatric syndrome that is characterized by gradual loss of muscle mass and strength with increasing age. Although the underlying mechanism is still unknown, the contribution of increased oxidative stress in advanced age has been recognized as one of the risk factors of sarcopenia. Thus, eliminating reactive oxygen species (ROS) can be a strategy to combat sarcopenia. In this review, we discuss the potential role of vitamin E in the prevention and treatment of sarcopenia. Vitamin E is a lipid soluble vitamin, with potent antioxidant properties and current evidence suggesting a role in the modulation of signaling pathways. Previous studies have shown its possible beneficial effects on aging and age-related diseases. Although there are evidences suggesting an association between vitamin E and muscle health, they are still inconclusive compared to other more extensively studied chronic diseases such as neurodegenerative diseases and cardiovascular diseases. Therefore, we reviewed the role of vitamin E and its potential protective mechanisms on muscle health based on previous and current in vitro and in vivo studies.
Collapse
|
232
|
Qin W, Pan J, Qin Y, Lee DN, Bauman WA, Cardozo C. Identification of functional glucocorticoid response elements in the mouse FoxO1 promoter. Biochem Biophys Res Commun 2014; 450:979-83. [DOI: 10.1016/j.bbrc.2014.06.080] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 06/17/2014] [Indexed: 12/12/2022]
|
233
|
Sharma M, Juvvuna PK, Kukreti H, McFarlane C. Mega roles of microRNAs in regulation of skeletal muscle health and disease. Front Physiol 2014; 5:239. [PMID: 25018733 PMCID: PMC4072100 DOI: 10.3389/fphys.2014.00239] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 06/10/2014] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle is a dynamic tissue with remarkable plasticity. Skeletal muscle growth and regeneration are highly organized processes thus it is not surprising that a high degree of complexity exists in the regulation of these processes. Recent discovery of non-coding microRNAs (miRNAs) has prompted extensive research in understanding the roles of these molecules in skeletal muscle. Research so far shows that miRNAs play a very significant role at every aspect of muscle biology. Besides muscle growth, development, and regeneration miRNAs are also involved in the pathology of muscle diseases and metabolism. In this review, recent advancements in miRNA function during myogenesis, exercise, atrophy, aging, and dystrophy are discussed.
Collapse
Affiliation(s)
- Mridula Sharma
- Department of Biochemistry, YLL School of Medicine, National University of Singapore Singapore, Singapore
| | - Prasanna Kumar Juvvuna
- Department of Biochemistry, YLL School of Medicine, National University of Singapore Singapore, Singapore
| | - Himani Kukreti
- Department of Biochemistry, YLL School of Medicine, National University of Singapore Singapore, Singapore
| | - Craig McFarlane
- Brenner Centre for Molecular Medicine, Singapore Institute for Clinical Sciences (ASTAR) Singapore, Singapore
| |
Collapse
|
234
|
Bollinger LM, Witczak CA, Houmard JA, Brault JJ. SMAD3 augments FoxO3-induced MuRF-1 promoter activity in a DNA-binding-dependent manner. Am J Physiol Cell Physiol 2014; 307:C278-87. [PMID: 24920680 DOI: 10.1152/ajpcell.00391.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Muscle-specific RING finger-1 (MuRF-1), a ubiquitin ligase and key regulator of proteasome-dependent protein degradation, is highly expressed during skeletal muscle atrophy. The transcription factor forkhead box O3 (FoxO3) induces MuRF-1 expression, but the direct role of other major atrophy-related transcription factors, such as SMAD3, is largely unknown. The goal of this study was to determine whether SMAD3 individually regulates, or with FoxO3 coordinately regulates, MuRF-1 expression. In cultured myotubes or human embryonic kidney cells, MuRF-1 mRNA content and promoter activity were increased by FoxO3 but not by SMAD3 overexpression. However, FoxO3 and SMAD3 coexpression synergistically increased MuRF-1 mRNA and promoter activity. Mutation of the SMAD-binding element (SBE) in the proximal MuRF-1 promoter or overexpression of a SMAD3 DNA-binding mutant attenuated FoxO3-dependent MuRF-1 promoter activation, showing that SMAD binding to DNA is required for optimal activation of FoxO3-induced transcription of MuRF-1. Using chromatin immunoprecipitation, SMAD3 DNA binding increased FoxO3 abundance and SBE mutation reduced FoxO3 abundance on the MuRF-1 promoter. Furthermore, SMAD3 overexpression dose-dependently increased FoxO3 protein content, and coexpression of FoxO3 and SMAD3 synergistically increased FoxO-dependent gene transcription [assessed with a FoxO response element (FRE)-driven reporter]. Collectively, these results show that SMAD3 regulates transcription of MuRF-1 by increasing FoxO3 binding at a conserved FRE-SBE motif within the proximal promoter region, and by increasing FoxO3 protein content and transcriptional activity. These data are the first to indicate that two major transcription factors regulating protein degradation, FoxO3 and SMAD3, converge to coordinately and directly regulate transcription of MuRF-1.
Collapse
Affiliation(s)
- Lance M Bollinger
- Department of Kinesiology, Human Performance Laboratory, College of Health and Human Performance, East Carolina University, Greenville, North Carolina; Department of Biochemistry and Molecular Biology, Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Carol A Witczak
- Department of Kinesiology, Human Performance Laboratory, College of Health and Human Performance, East Carolina University, Greenville, North Carolina; Department of Biochemistry and Molecular Biology, Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Joseph A Houmard
- Department of Kinesiology, Human Performance Laboratory, College of Health and Human Performance, East Carolina University, Greenville, North Carolina; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - Jeffrey J Brault
- Department of Kinesiology, Human Performance Laboratory, College of Health and Human Performance, East Carolina University, Greenville, North Carolina; Department of Biochemistry and Molecular Biology, Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, North Carolina; and East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| |
Collapse
|
235
|
Soares RJ, Cagnin S, Chemello F, Silvestrin M, Musaro A, De Pitta C, Lanfranchi G, Sandri M. Involvement of microRNAs in the regulation of muscle wasting during catabolic conditions. J Biol Chem 2014; 289:21909-25. [PMID: 24891504 PMCID: PMC4139209 DOI: 10.1074/jbc.m114.561845] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Loss of muscle proteins and the consequent weakness has important clinical consequences in diseases such as cancer, diabetes, chronic heart failure, and in aging. In fact, excessive proteolysis causes cachexia, accelerates disease progression, and worsens life expectancy. Muscle atrophy involves a common pattern of transcriptional changes in a small subset of genes named atrophy-related genes or atrogenes. Whether microRNAs play a role in the atrophy program and muscle loss is debated. To understand the involvement of miRNAs in atrophy we performed miRNA expression profiling of mouse muscles under wasting conditions such as fasting, denervation, diabetes, and cancer cachexia. We found that the miRNA signature is peculiar of each catabolic condition. We then focused on denervation and we revealed that changes in transcripts and microRNAs expression did not occur simultaneously but were shifted. Indeed, whereas transcriptional control of the atrophy-related genes peaks at 3 days, changes of miRNA expression maximized at 7 days after denervation. Among the different miRNAs, microRNA-206 and -21 were the most induced in denervated muscles. We characterized their pattern of expression and defined their role in muscle homeostasis. Indeed, in vivo gain and loss of function experiments revealed that miRNA-206 and miRNA-21 were sufficient and required for atrophy program. In silico and in vivo approaches identified transcription factor YY1 and the translational initiator factor eIF4E3 as downstream targets of these miRNAs. Thus miRNAs are important for fine-tuning the atrophy program and their modulation can be a novel potential therapeutic approach to counteract muscle loss and weakness in catabolic conditions.
Collapse
Affiliation(s)
- Ricardo José Soares
- From the Dulbecco Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy, the Ph.D. Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Stefano Cagnin
- the Department of Biology and CRIBI Biotechnology Centre, University of Padova, 35121 Padova, Italy
| | - Francesco Chemello
- the Department of Biology and CRIBI Biotechnology Centre, University of Padova, 35121 Padova, Italy
| | - Matteo Silvestrin
- the Department of Biology and CRIBI Biotechnology Centre, University of Padova, 35121 Padova, Italy
| | - Antonio Musaro
- the DAHFMO-Unit of Histology and Medical Embryology, Sapienza University, 00161 Roma, Italy, and
| | - Cristiano De Pitta
- the Department of Biology and CRIBI Biotechnology Centre, University of Padova, 35121 Padova, Italy,
| | - Gerolamo Lanfranchi
- the Department of Biology and CRIBI Biotechnology Centre, University of Padova, 35121 Padova, Italy,
| | - Marco Sandri
- From the Dulbecco Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy, the Department of Biomedical Sciences and the Institute of Neuroscience, Consiglio Nazionale delle Ricerche, 35121 Padova, Italy, the Telethon Institute of Genetics and Medicine (TIGEM), 80131 Napoli, Italy
| |
Collapse
|
236
|
Hudson MB, Rahnert JA, Zheng B, Woodworth-Hobbs ME, Franch HA, Price SR. miR-182 attenuates atrophy-related gene expression by targeting FoxO3 in skeletal muscle. Am J Physiol Cell Physiol 2014; 307:C314-9. [PMID: 24871856 DOI: 10.1152/ajpcell.00395.2013] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Skeletal muscle atrophy occurs in response to a variety of conditions including chronic kidney disease, diabetes, cancer, and elevated glucocorticoids. MicroRNAs (miR) may play a role in the wasting process. Activation of the forkhead box O3 (FoxO3) transcription factor causes skeletal muscle atrophy in patients, animals, and cultured cells by increasing the expression of components of the ubiquitin-proteasome and autophagy-lysosome proteolytic systems. To identify microRNAs that potentially modulate the atrophy process, an in silico target analysis was performed and miR-182 was predicted to target FoxO3 mRNA. Using a combination of immunoblot analysis, quantitative real-time RT-PCR, and FoxO3 3'-UTR luciferase reporter genes, miR-182 was confirmed to regulate FoxO3 expression in C2C12 myotubes. Transfection of miR-182 into muscle cells decreased FoxO3 mRNA 30% and FoxO3 protein 67% (P < 0.05) and also prevented a glucocorticoid-induced upregulation of multiple FoxO3 gene targets including MAFbx/atrogin-1, autophagy-related protein 12 (ATG12), cathepsin L, and microtubule-associated protein light chain 3 (LC3). Treatment of C2C12 myotubes with dexamethasone (Dex) (1 μM, 6 h) to induce muscle atrophy decreased miR-182 expression by 63% (P < 0.05). Similarly, miR-182 was decreased 44% (P < 0.05) in the gastrocnemius muscle of rats injected with streptozotocin to induce diabetes compared with controls. Finally, miR-182 was present in exosomes isolated from the media of C2C12 myotubes and Dex increased its abundance. These data identify miR-182 as an important regulator of FoxO3 expression that participates in the control of atrophy-inducing genes during catabolic diseases.
Collapse
Affiliation(s)
- Matthew B Hudson
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia;
| | - Jill A Rahnert
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Bin Zheng
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - Myra E Woodworth-Hobbs
- Nutrition and Health Sciences Ph.D. Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, Georgia
| | - Harold A Franch
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia
| | - S Russ Price
- Department of Medicine, Renal Division, Emory University, Atlanta, Georgia; Biomedical Laboratory Research and Development Service, Atlanta VA Medical Center, Decatur, Georgia
| |
Collapse
|
237
|
Costa A, Toschi A, Murfuni I, Pelosi L, Sica G, Adamo S, Scicchitano BM. Local overexpression of V1a-vasopressin receptor enhances regeneration in tumor necrosis factor-induced muscle atrophy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:235426. [PMID: 24971321 PMCID: PMC4055243 DOI: 10.1155/2014/235426] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 01/25/2023]
Abstract
Skeletal muscle atrophy occurs during disuse and aging, or as a consequence of chronic diseases such as cancer and diabetes. It is characterized by progressive loss of muscle tissue due to hypotrophic changes, degeneration, and an inability of the regeneration machinery to replace damaged myofibers. Tumor necrosis factor (TNF) is a proinflammatory cytokine known to mediate muscle atrophy in many chronic diseases and to inhibit skeletal muscle regeneration. In this study, we investigated the role of Arg-vasopressin-(AVP-)dependent pathways in muscles in which atrophy was induced by local overexpression of TNF. AVP is a potent myogenesis-promoting factor and is able to enhance skeletal muscle regeneration by stimulating Ca(2+)/calmodulin-dependent kinase and calcineurin signaling. We performed morphological and molecular analyses and demonstrated that local over-expression of the AVP receptor V1a enhances regeneration of atrophic muscle. By upregulating the regeneration/differentiation markers, modulating the inflammatory response, and attenuating fibrogenesis, the stimulation of AVP-dependent pathways creates a favourable environment for efficient and sustained muscle regeneration and repair even in the presence of elevated levels of TNF. This study highlights a novel in vivo role for AVP-dependent pathways, which may represent an interesting strategy to counteract muscle decline in aging or in muscular pathologies.
Collapse
Affiliation(s)
- Alessandra Costa
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Via A. Scarpa 16, 00161 Rome, Italy
| | - Angelica Toschi
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Via A. Scarpa 16, 00161 Rome, Italy
| | - Ivana Murfuni
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Via A. Scarpa 16, 00161 Rome, Italy
| | - Laura Pelosi
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Via A. Scarpa 16, 00161 Rome, Italy
| | - Gigliola Sica
- Institute of Histology and Embryology, Catholic University School of Medicine, L.go F. Vito, 1, 00168 Rome, Italy
| | - Sergio Adamo
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Via A. Scarpa 16, 00161 Rome, Italy
| | - Bianca Maria Scicchitano
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Via A. Scarpa 16, 00161 Rome, Italy
- Institute of Histology and Embryology, Catholic University School of Medicine, L.go F. Vito, 1, 00168 Rome, Italy
| |
Collapse
|
238
|
Li W, Claypool MD, Friera AM, McLaughlin J, Baltgalvis KA, Smith IJ, Kinoshita T, White K, Lang W, Godinez G, Payan DG, Kinsella TM. Noninvasive imaging of in vivo MuRF1 expression during muscle atrophy. PLoS One 2014; 9:e94032. [PMID: 24710205 PMCID: PMC3977994 DOI: 10.1371/journal.pone.0094032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 03/11/2014] [Indexed: 11/20/2022] Open
Abstract
Numerous human diseases can lead to atrophy of skeletal muscle, and loss of this tissue has been correlated with increased mortality and morbidity rates. Clinically addressing muscle atrophy remains an unmet medical need, and the development of preclinical tools to assist drug discovery and basic research in this effort is important for advancing this goal. In this report, we describe the development of a bioluminescent gene reporter rat, based on the zinc finger nuclease-targeted insertion of a bicistronic luciferase reporter into the 3′ untranslated region of a muscle specific E3 ubiquitin ligase gene, MuRF1 (Trim63). In longitudinal studies, we noninvasively assess atrophy-related expression of this reporter in three distinct models of muscle loss (sciatic denervation, hindlimb unloading and dexamethasone-treatment) and show that these animals are capable of generating refined detail on in vivo MuRF1 expression with high temporal and anatomical resolution.
Collapse
Affiliation(s)
- Wei Li
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Mark D. Claypool
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Annabelle M. Friera
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - John McLaughlin
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Kristen A. Baltgalvis
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Ira J. Smith
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Taisei Kinoshita
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Kathy White
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Wayne Lang
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Guillermo Godinez
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Donald G. Payan
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
| | - Todd M. Kinsella
- Discovery Research, Rigel Pharmaceuticals Inc., South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
239
|
Forman DE, Daniels KM, Cahalin LP, Zavin A, Allsup K, Cao P, Santhanam M, Joseph J, Arena R, Lazzari A, Schulze PC, Lecker SH. Analysis of skeletal muscle gene expression patterns and the impact of functional capacity in patients with systolic heart failure. J Card Fail 2014; 20:422-30. [PMID: 24704539 DOI: 10.1016/j.cardfail.2014.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 02/24/2014] [Accepted: 03/25/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Declining physical function is common among systolic heart failure (HF) patients and heralds poor clinical outcomes. We hypothesized that coordinated shifts in expression of ubiquitin-mediated atrophy-promoting genes are associated with muscle atrophy and contribute to decreased physical function. METHODS Systolic HF patients (left ventricular ejection fraction [LVEF] ≤40%) underwent skeletal muscle biopsies (nondominant vastus lateralis) and comprehensive physical assessments. Skeletal muscle gene expression was assessed with the use of real-time polymerase chain reaction. Aerobic function was assessed with the use of cardiopulmonary exercise and 6-minute walk tests. Strength capacity was assessed with the use of pneumatic leg press (maximum strength and power). Serologic inflammatory markers also were assessed. RESULTS 54 male patients (66.6 ± 10.0 years) were studied: 24 systolic HF patients (mean LVEF 28.9 ± 7.8%) and 30 age-matched control subjects. Aerobic and strength parameters were diminished in HF versus control. FoxO1 and FoxO3 were increased in HF versus control (7.9 ± 6.2 vs 5.0 ± 3.5, 6.5 ± 4.3 vs 4.3 ± 2.8 relative units, respectively; P ≤ .05 in both). However, atrogin-1 and MuRF-1 were similar in both groups. PGC-1α was also increased in HF (7.9 ± 5.4 vs. 5.3 ± 3.6 relative units; P < .05). Muscle levels of insulin-like growth factor (IGF) 1 as well as serum levels of tumor necrosis factor α, C-reactive protein, interleukin (IL) 1β, and IL-6 were similar in HF and control. CONCLUSION Expression of the atrophy-promoting genes FoxO1 and FoxO3 were increased in skeletal muscle in systolic HF compared with control, but other atrophy gene expression patterns (atrogin-1 and MuRF-1), as well as growth promoting patterns (IGF-1), were similar. PGC-1α, a gene critical in enhancing mitochondrial function and moderating FoxO activity, may play an important counterregulatory role to offset ubiquitin pathway-mediated functional decrements.
Collapse
Affiliation(s)
- Daniel E Forman
- New England Geriatric Research, Education, and Clinical Center, Veterans Affairs Boston Healthcare System, Boston, Massachusetts; Division of Cardiovascular Medicine, Veterans Affairs Boston Healthcare System, Boston, Massachusetts; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical, School, Boston, Massachusetts.
| | - Karla M Daniels
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical, School, Boston, Massachusetts
| | - Lawrence P Cahalin
- Department of Physical Therapy, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida
| | - Alexandra Zavin
- New England Geriatric Research, Education, and Clinical Center, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Kelly Allsup
- New England Geriatric Research, Education, and Clinical Center, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Peirang Cao
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Mahalakshmi Santhanam
- Division of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jacob Joseph
- Division of Cardiovascular Medicine, Veterans Affairs Boston Healthcare System, Boston, Massachusetts; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical, School, Boston, Massachusetts
| | - Ross Arena
- Department of Physical Therapy and Integrative Physiology Laboratory - College of Applied Health Sciences, University of Illinois Chicago, Chicago, Illinois
| | - Antonio Lazzari
- Division of Rheumatology, Veterans Affairs Boston Healthcare System, Boston, Massachusetts; Boston University School of Medicine, Boston, Massachusetts
| | - P Christian Schulze
- Division of Cardiovascular Medicine, Columbia University Medical Center, New York, New York
| | | |
Collapse
|
240
|
Beharry AW, Sandesara PB, Roberts BM, Ferreira LF, Senf SM, Judge AR. HDAC1 activates FoxO and is both sufficient and required for skeletal muscle atrophy. J Cell Sci 2014; 127:1441-53. [PMID: 24463822 PMCID: PMC3970557 DOI: 10.1242/jcs.136390] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The Forkhead box O (FoxO) transcription factors are activated, and necessary for the muscle atrophy, in several pathophysiological conditions, including muscle disuse and cancer cachexia. However, the mechanisms that lead to FoxO activation are not well defined. Recent data from our laboratory and others indicate that the activity of FoxO is repressed under basal conditions via reversible lysine acetylation, which becomes compromised during catabolic conditions. Therefore, we aimed to determine how histone deacetylase (HDAC) proteins contribute to activation of FoxO and induction of the muscle atrophy program. Through the use of various pharmacological inhibitors to block HDAC activity, we demonstrate that class I HDACs are key regulators of FoxO and the muscle-atrophy program during both nutrient deprivation and skeletal muscle disuse. Furthermore, we demonstrate, through the use of wild-type and dominant-negative HDAC1 expression plasmids, that HDAC1 is sufficient to activate FoxO and induce muscle fiber atrophy in vivo and is necessary for the atrophy of muscle fibers that is associated with muscle disuse. The ability of HDAC1 to cause muscle atrophy required its deacetylase activity and was linked to the induction of several atrophy genes by HDAC1, including atrogin-1, which required deacetylation of FoxO3a. Moreover, pharmacological inhibition of class I HDACs during muscle disuse, using MS-275, significantly attenuated both disuse muscle fiber atrophy and contractile dysfunction. Together, these data solidify the importance of class I HDACs in the muscle atrophy program and indicate that class I HDAC inhibitors are feasible countermeasures to impede muscle atrophy and weakness.
Collapse
Affiliation(s)
- Adam W. Beharry
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610-0154, USA
| | - Pooja B. Sandesara
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610-0154, USA
| | - Brandon M. Roberts
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610-0154, USA
| | - Leonardo F. Ferreira
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32610-0154, USA
| | - Sarah M. Senf
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610-0154, USA
| | - Andrew R. Judge
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610-0154, USA
- Author for correspondence ()
| |
Collapse
|
241
|
Remels AHV, Pansters NA, Gosker HR, Schols AMWJ, Langen RCJ. Activation of alternative NF-κB signaling during recovery of disuse-induced loss of muscle oxidative phenotype. Am J Physiol Endocrinol Metab 2014; 306:E615-26. [PMID: 24425759 DOI: 10.1152/ajpendo.00452.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Physical inactivity-induced loss of skeletal muscle oxidative phenotype (OXPHEN), often observed in chronic disease, adversely affects physical functioning and quality of life. Potential therapeutic targets remain to be identified, since the molecular mechanisms involved in reloading-induced recovery of muscle OXPHEN remain incompletely understood. We hypothesized a role for alternative NF-κB, as a recently identified positive regulator of muscle OXPHEN, in reloading-induced alterations in muscle OXPHEN. Markers and regulators (including alternative NF-κB signaling) of muscle OXPHEN were investigated in gastrocnemius muscle of mice subjected to a hindlimb suspension/reloading (HLS/RL) protocol. Expression levels of oxidative phosphorylation subunits and slow myosin heavy chain isoforms I and IIA increased rapidly upon RL. After an initial decrease upon HLS, mRNA levels of peroxisome proliferator-activated receptor (PPAR)-γ coactivator (PGC) molecules PGC-1α and PGC-1β and mRNA levels of mitochondrial transcription factor A (Tfam) and estrogen-related receptor α increased upon RL. PPAR-δ, nuclear respiratory factor 1 (NRF-1), NRF-2α, and sirtuin 1 mRNA levels increased during RL although expression levels were unaltered upon HLS. In addition, both Tfam and NRF-1 protein levels increased significantly during the RL period. Moreover, upon RL, IKK-α mRNA and protein levels increased, and phosphorylation of P100 and subsequent processing to P52 were elevated, reflecting alternative NF-κB activation. We conclude that RL-induced recovery of muscle OXPHEN is associated with activation of alternative NF-κB signaling.
Collapse
Affiliation(s)
- A H V Remels
- NUTRIM School for Nutrition, Toxicology, and Metabolism, Department of Respiratory Medicine, Maastricht University Medical Center +, Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
242
|
Wollersheim T, Woehlecke J, Krebs M, Hamati J, Lodka D, Luther-Schroeder A, Langhans C, Haas K, Radtke T, Kleber C, Spies C, Labeit S, Schuelke M, Spuler S, Spranger J, Weber-Carstens S, Fielitz J. Dynamics of myosin degradation in intensive care unit-acquired weakness during severe critical illness. Intensive Care Med 2014; 40:528-38. [PMID: 24531339 DOI: 10.1007/s00134-014-3224-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 01/18/2014] [Indexed: 01/31/2023]
Abstract
IMPORTANCE Intensive care unit (ICU)-acquired muscle wasting is a devastating complication leading to persistent weakness and functional disability. The mechanisms of this myopathy are unclear, but a disturbed balance of myosin heavy chain (MyHC) is implicated. OBJECTIVE To investigate pathways of myosin turnover in severe critically ill patients at high risk of ICU-acquired weakness. DESIGN Prospective, mechanistic, observational study. SETTING Interdisciplinary ICUs of a university hospital. PARTICIPANTS Twenty-nine patients with Sequential Organ Failure Assessment (SOFA) scores of at least 8 on three consecutive days within the first 5 days in ICU underwent two consecutive open skeletal muscle biopsies from the vastus lateralis at median days 5 and 15. Control biopsy specimens were from healthy subjects undergoing hip-replacement surgery. INTERVENTIONS None. MAIN OUTCOME(S) AND MEASURE(S) Time-dependent changes in myofiber architecture, MyHC synthesis, and degradation were determined and correlated with clinical data. RESULTS ICU-acquired muscle wasting was characterized by early, disrupted myofiber ultrastructure followed by atrophy of slow- and fast-twitch myofibers at later time points. A rapid decrease in MyHC mRNA and protein expression occurred by day 5 and persisted at day 15 (P < 0.05). Expression of the atrophy genes MuRF-1 and Atrogin1 was increased at day 5 (P < 0.05). Early MuRF-1 protein content was closely associated with late myofiber atrophy and the severity of weakness. CONCLUSIONS AND RELEVANCE Decreased synthesis and increased degradation of MyHCs contribute to ICU-acquired muscle wasting. The rates and time frames suggest that pathogenesis of muscle failure is initiated very early during critical illness. The persisting reduction of MyHC suggests that sustained treatment is required.
Collapse
Affiliation(s)
- Tobias Wollersheim
- Anesthesiology and Operative Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Campus Virchow and Campus Mitte, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
MacDonald EM, Andres-Mateos E, Mejias R, Simmers JL, Mi R, Park JS, Ying S, Hoke A, Lee SJ, Cohn RD. Denervation atrophy is independent from Akt and mTOR activation and is not rescued by myostatin inhibition. Dis Model Mech 2014; 7:471-81. [PMID: 24504412 PMCID: PMC3974457 DOI: 10.1242/dmm.014126] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The purpose of our study was to compare two acquired muscle atrophies and the use of myostatin inhibition for their treatment. Myostatin naturally inhibits skeletal muscle growth by binding to ActRIIB, a receptor on the cell surface of myofibers. Because blocking myostatin in an adult wild-type mouse induces profound muscle hypertrophy, we applied a soluble ActRIIB receptor to models of disuse (limb immobilization) and denervation (sciatic nerve resection) atrophy. We found that treatment of immobilized mice with ActRIIB prevented the loss of muscle mass observed in placebo-treated mice. Our results suggest that this protection from disuse atrophy is regulated by serum and glucocorticoid-induced kinase (SGK) rather than by Akt. Denervation atrophy, however, was not protected by ActRIIB treatment, yet resulted in an upregulation of the pro-growth factors Akt, SGK and components of the mTOR pathway. We then treated the denervated mice with the mTOR inhibitor rapamycin and found that, despite a reduction in mTOR activation, there is no alteration of the atrophy phenotype. Additionally, rapamycin prevented the denervation-induced upregulation of the mTORC2 substrates Akt and SGK. Thus, our studies show that denervation atrophy is not only independent from Akt, SGK and mTOR activation but also has a different underlying pathophysiological mechanism than disuse atrophy.
Collapse
Affiliation(s)
- Elizabeth M MacDonald
- McKusick-Nathans Institute of Genetic Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Piccirillo R, Demontis F, Perrimon N, Goldberg AL. Mechanisms of muscle growth and atrophy in mammals and Drosophila. Dev Dyn 2014; 243:201-15. [PMID: 24038488 PMCID: PMC3980484 DOI: 10.1002/dvdy.24036] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 08/01/2013] [Accepted: 08/01/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The loss of skeletal muscle mass (atrophy) that accompanies disuse and systemic diseases is highly debilitating. Although the pathogenesis of this condition has been primarily studied in mammals, Drosophila is emerging as an attractive system to investigate some of the mechanisms involved in muscle growth and atrophy. RESULTS In this review, we highlight the outstanding unsolved questions that may benefit from a combination of studies in both flies and mammals. In particular, we discuss how different environmental stimuli and signaling pathways influence muscle mass and strength and how a variety of disease states can cause muscle wasting. CONCLUSIONS Studies in Drosophila and mammals should help identify molecular targets for the treatment of muscle wasting in humans.
Collapse
Affiliation(s)
- Rosanna Piccirillo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
- Department of Oncology, IRCCS - Mario Negri Institute for Pharmacological Research, Milano, Italy
| | - Fabio Demontis
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Department of Developmental Neurobiology, Division of Developmental Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115
| | | |
Collapse
|
245
|
Jespersen JG, Mikkelsen UR, Nedergaard A, Thorlund JB, Schjerling P, Suetta C, Christensen PA, Aagaard P. Alterations in molecular muscle mass regulators after 8 days immobilizing Special Forces mission. Scand J Med Sci Sports 2014; 25:175-83. [PMID: 24422600 DOI: 10.1111/sms.12170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2013] [Indexed: 01/01/2023]
Abstract
In military operations, declined physical capacity can endanger the life of soldiers. During special support and reconnaissance (SSR) missions, Special Forces soldiers sustain 1-2 weeks full-body horizontal immobilization, which impairs muscle strength and performance. Adequate muscle mass and strength are necessary in combat or evacuation situations, which prompt for improved understanding of muscle mass modulation during SSR missions. To explore the molecular regulation of myofiber size during a simulated SSR operation, nine male Special Forces soldiers were biopsied in m. vastus lateralis pre and post 8 days immobilizing restricted prone position. After immobilization, total mammalian target of rapamycin protein was reduced by 42% (P < 0.05), whereas total and phosphorylated protein levels of Akt, ribosomal protein S6k, 4E-BP1, and glycogen synthase kinase3β were unchanged. Messenger RNA (mRNA) levels of the atrogenes forkhead box O3 (FoxO3), atrogin1, and muscle ring finger protein1 (MuRF1) increased by 36%, 53%, and 71% (P < 0.01), MuRF1 protein by 51% (P = 0.05), whereas FoxO1 and peroxisome proliferator-activated receptor γ coactivator-1 β mRNAs decreased by 29% and 40% (P < 0.01). In conclusion, occupational immobilization in Special Forces soldiers led to modulations in molecular muscle mass regulators during 8 days prone SSR mission, which likely contribute to muscle loss observed in such operations. The present data expand our knowledge of human muscle mass regulation during short-term immobilization.
Collapse
Affiliation(s)
- J G Jespersen
- Institute of Sports Medicine, Department of Orthopedic Surgery M, Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
246
|
Joshi SK, Kim HT, Feeley BT, Liu X. Differential ubiquitin-proteasome and autophagy signaling following rotator cuff tears and suprascapular nerve injury. J Orthop Res 2014; 32:138-44. [PMID: 24018537 PMCID: PMC3856942 DOI: 10.1002/jor.22482] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 08/14/2013] [Indexed: 02/06/2023]
Abstract
Previous studies have evaluated role of Akt/mTOR signaling in rotator cuff muscle atrophy and determined that there was differential in signaling following tendon transection (TT) and suprascapular nerve (SSN) denervation (DN), suggesting that atrophy following TT and DN was modulated by different protein degradation pathways. In this study, two muscle proteolytic systems that have been shown to be potent regulators of muscle atrophy in other injury models, the ubiquitin-proteasome pathway and autophagy, were evaluated following TT and DN. In addition to examining protein degradation, this study assessed protein synthesis rate following these two surgical models to understand how the balance between protein degradation and synthesis results in atrophy following rotator cuff injury. In contrast to the traditional theory that protein synthesis is decreased during muscle atrophy, this study suggests that protein synthesis is up-regulated in rotator cuff muscle atrophy following both surgical models. While the ubiquitin-proteasome pathway was a major contributor to the atrophy seen following DN, autophagy was a major contributor following TT. The findings of this study suggest that protein degradation is the primary factor contributing to atrophy following rotator cuff injury. However, different proteolytic pathways are activated if SSN injury is involved.
Collapse
Affiliation(s)
- Sunil K. Joshi
- San Francisco Veterans Affairs Medical Center, Department of Veterans Affairs,Department of Orthopaedic Surgery, University of California, San Francisco
| | - Hubert T. Kim
- San Francisco Veterans Affairs Medical Center, Department of Veterans Affairs,Department of Orthopaedic Surgery, University of California, San Francisco
| | - Brian T. Feeley
- San Francisco Veterans Affairs Medical Center, Department of Veterans Affairs,Department of Orthopaedic Surgery, University of California, San Francisco
| | - Xuhui Liu
- San Francisco Veterans Affairs Medical Center, Department of Veterans Affairs,Department of Orthopaedic Surgery, University of California, San Francisco,Corresponding author: Xuhui Liu, M.D., San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA 94153, Tel: 415-221-4810 x 2742, Fax: 415-750-2181,
| |
Collapse
|
247
|
Hudson MB, Woodworth-Hobbs ME, Zheng B, Rahnert JA, Blount MA, Gooch JL, Searles CD, Price SR. miR-23a is decreased during muscle atrophy by a mechanism that includes calcineurin signaling and exosome-mediated export. Am J Physiol Cell Physiol 2013; 306:C551-8. [PMID: 24336651 DOI: 10.1152/ajpcell.00266.2013] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Skeletal muscle atrophy is prevalent in chronic diseases, and microRNAs (miRs) may play a key role in the wasting process. miR-23a was previously shown to inhibit the expression of atrogin-1 and muscle RING-finger protein-1 (MuRF1) in muscle. It also was reported to be regulated by cytoplasmic nuclear factor of activated T cells 3 (NFATc3) in cardiomyocytes. The objective of this study was to determine if miR-23a is regulated during muscle atrophy and to evaluate the relationship between calcineurin (Cn)/NFAT signaling and miR-23a expression in skeletal muscle cells during atrophy. miR-23a was decreased in the gastrocnemius of rats with acute streptozotocin-induced diabetes, a condition known to increase atrogin-1 and MuRF1 expression and cause atrophy. Treatment of C2C12 myotubes with dexamethasone (Dex) for 48 h also reduced miR-23a as well as RCAN1.4 mRNA, which is transcriptionally regulated by NFAT. NFATc3 nuclear localization and the amount of miR-23a decreased rapidly within 1 h of Dex administration, suggesting a link between Cn signaling and miR-23a. The level of miR-23a was lower in primary myotubes from mice lacking the α- or β-isoform of the CnA catalytic subunit than wild-type mice. Dex did not further suppress miR-23a in myotubes from Cn-deficient mice. Overexpression of CnAβ in C2C12 myotubes prevented Dex-induced suppression of miR-23a. Finally, miR-23a was present in exosomes isolated from the media of C2C12 myotubes, and Dex increased its exosomal abundance. Dex did not alter the number of exosomes released into the media. We conclude that atrophy-inducing conditions downregulate miR-23a in muscle by mechanisms involving attenuated Cn/NFAT signaling and selective packaging into exosomes.
Collapse
Affiliation(s)
- Matthew B Hudson
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | | | | | | | | | | | | | | |
Collapse
|
248
|
Durigan JL, Delfino GB, Peviani SM, Russo TL, Ramírez C, Da Silva Gomes AD, Salvini TF. Neuromuscular electrical stimulation alters gene expression and delays quadriceps muscle atrophy of rats after anterior cruciate ligament transection. Muscle Nerve 2013; 49:120-8. [DOI: 10.1002/mus.23883] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2013] [Indexed: 01/12/2023]
Affiliation(s)
- João L.Q. Durigan
- Department of Physical Therapy; University of Brasília; Distrito Federal Brazil
| | - Gabriel B. Delfino
- Muscle Plasticity Laboratory; Department of Physical Therapy; Federal University of São Carlos; São Paulo Brazil
| | - Sabrina M. Peviani
- Muscle Plasticity Laboratory; Department of Physical Therapy; Federal University of São Carlos; São Paulo Brazil
| | - Thiago L. Russo
- Muscle Plasticity Laboratory; Department of Physical Therapy; Federal University of São Carlos; São Paulo Brazil
| | - Carolina Ramírez
- School of Physical Therapy; Industrial University of Santander; Bucaramanga Santander Colombia
| | - André D.B. Da Silva Gomes
- Muscle Plasticity Laboratory; Department of Physical Therapy; Federal University of São Carlos; São Paulo Brazil
| | - Tania F. Salvini
- Muscle Plasticity Laboratory; Department of Physical Therapy; Federal University of São Carlos; São Paulo Brazil
| |
Collapse
|
249
|
Huang J, Wang J, Gu L, Bao J, Yin J, Tang Z, Wang L, Yuan W. Effect of a low-protein diet supplemented with ketoacids on skeletal muscle atrophy and autophagy in rats with type 2 diabetic nephropathy. PLoS One 2013; 8:e81464. [PMID: 24303049 PMCID: PMC3841136 DOI: 10.1371/journal.pone.0081464] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/14/2013] [Indexed: 02/07/2023] Open
Abstract
A low-protein diet supplemented with ketoacids maintains nutritional status in patients with diabetic nephropathy. The activation of autophagy has been shown in the skeletal muscle of diabetic and uremic rats. This study aimed to determine whether a low-protein diet supplemented with ketoacids improves muscle atrophy and decreases the increased autophagy observed in rats with type 2 diabetic nephropathy. In this study, 24-week-old Goto-Kakizaki male rats were randomly divided into groups that received either a normal protein diet (NPD group), a low-protein diet (LPD group) or a low-protein diet supplemented with ketoacids (LPD+KA group) for 24 weeks. Age- and weight-matched Wistar rats served as control animals and received a normal protein diet (control group). We found that protein restriction attenuated proteinuria and decreased blood urea nitrogen and serum creatinine levels. Compared with the NPD and LPD groups, the LPD+KA group showed a delay in body weight loss, an attenuation in soleus muscle mass loss and a decrease of the mean cross-sectional area of soleus muscle fibers. The mRNA and protein expression of autophagy-related genes, such as Beclin-1, LC3B, Bnip3, p62 and Cathepsin L, were increased in the soleus muscle of GK rats fed with NPD compared to Wistar rats. Importantly, LPD resulted in a slight reduction in the expression of autophagy-related genes; however, these differences were not statistically significant. In addition, LPD+KA abolished the upregulation of autophagy-related gene expression. Furthermore, the activation of autophagy in the NPD and LPD groups was confirmed by the appearance of autophagosomes or autolysosomes using electron microscopy, when compared with the Control and LPD+KA groups. Our results showed that LPD+KA abolished the activation of autophagy in skeletal muscle and decreased muscle loss in rats with type 2 diabetic nephropathy.
Collapse
Affiliation(s)
- Juan Huang
- Department of Nephrology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Jialin Wang
- Department of Nephrology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Lijie Gu
- Department of Nephrology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Jinfang Bao
- Department of Nephrology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Jun Yin
- Department of Nephrology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Zhihuan Tang
- Department of Nephrology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Ling Wang
- Department of Nephrology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Weijie Yuan
- Department of Nephrology, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
- * E-mail:
| |
Collapse
|
250
|
Functional crosstalk of PGC-1 coactivators and inflammation in skeletal muscle pathophysiology. Semin Immunopathol 2013; 36:27-53. [DOI: 10.1007/s00281-013-0406-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 10/29/2013] [Indexed: 02/06/2023]
|