1
|
Kim YI, Lee H, Kim MJ, Jung CH, Kim YS, Ahn J. Identification of Peucedanum japonicum Thunb. extract components and their protective effects against dexamethasone-induced muscle atrophy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155449. [PMID: 38518644 DOI: 10.1016/j.phymed.2024.155449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/21/2023] [Accepted: 02/11/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND Peucedanum japonicum Thunb. (PJ) is a vegetable widely consumed in East Asia and is known to have anticancer and anti-inflammatory effects. However, the effect of PJ on muscle atrophy remains elusive. PURPOSE This study aimed to investigate the effect of PJ and its active compound on dexamethasone (DEX)-induced muscle atrophy. METHODS We performed qualitative and quantitative analysis of PJ using ultra-performance liquid chromatography-mass spectrometry tandem mass spectrometry (UPLC-MS/MS) and high-performance liquid chromatography (HPLC), respectively. The efficacy of PJ and its main compound 4-caffeoylquinic acid (CQA) on muscle atrophy was evaluated in DEX-induced myotube atrophy and DEX-induced muscle atrophy in mouse myoblasts (C2C12) and C57BL/6 mice, in vitro and in vivo, respectively. RESULTS The UPLC-MS/MS and HPLC data showed that the concentration of 4-CQA in PJ was 18.845 mg/g. PJ and 4-CQA treatments significantly inhibited DEX-induced myotube atrophy by decreasing protein synthesis and glucocorticoid translocation to the nucleus in C2C12 myotubes. In addition, PJ enhanced myogenesis by upregulating myogenin and myogenic differentiation 1 in C2C12 cells. PJ supplementation effectively increased muscle function and mass, downregulated atrogenes, and decreased proteasome activity in C57BL/6 mice. Additionally, PJ effectively decreased the nuclear translocation of forkhead transcription factor 3 alpha by inhibiting glucocorticoid receptor. CONCLUSION Overall, PJ and its active compound 4-CQA alleviated skeletal muscle atrophy by inhibiting protein degradation. Hence, our findings present PJ as a potential novel pharmaceutical candidate for the treatment of muscle atrophy.
Collapse
Affiliation(s)
- Young In Kim
- Aging and Metabolism Research Group, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, South Korea; Department of Food Science and Technology, Jeonbuk National University, Jeonju-si, South Korea
| | - Hyunjung Lee
- Aging and Metabolism Research Group, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, South Korea
| | - Min Jung Kim
- Aging and Metabolism Research Group, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, South Korea
| | - Chang Hwa Jung
- Aging and Metabolism Research Group, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, South Korea; Department of Food Biotechnology, Korea National University of Science and Technology, Daejeon, South Korea
| | - Young-Soo Kim
- Department of Food Science and Technology, Jeonbuk National University, Jeonju-si, South Korea
| | - Jiyun Ahn
- Aging and Metabolism Research Group, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, South Korea; Department of Food Biotechnology, Korea National University of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
2
|
El-far AS, Kamiya M, Saneyasu T, Honda K. Effects of Amino Acid Supplementation to a Low-Protein Diet on the Growth Performance and Protein Metabolism-related Factors in Broiler Chicks. J Poult Sci 2024; 61:2024014. [PMID: 38726100 PMCID: PMC11074001 DOI: 10.2141/jpsa.2024014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/03/2024] [Indexed: 05/12/2024] Open
Abstract
A low-protein (LP) diet may alleviate the environmental impact of chicken meat production by reducing nitrogen excretion and ammonia emissions. Thus, this study investigated the effect of a 15% reduced protein diet with or without amino acid (AA) supplementation on the growth performance of broiler chicks from 10 to 35 days of age and the underlying mechanism for loss of skeletal muscle mass. Thirty-six male broiler chicks were allocated to three experimental groups based on body weight: control, LP, and essential AA-supplemented LP (LP+AA). The body weight gain, feed conversion ratio, and weight of breast muscles and legs significantly decreased only in the LP group at the end of the feeding period. Plasma uric acid levels were significantly lower in the LP+AA group than those of the other groups. In the LP group, mRNA levels of microtubule-associated protein 1 light chain 3 isoform B were significantly higher in the pectoralis major, whereas those of atrogin-1, muscle RING-finger protein-1, and myoblast determination protein 1 were significantly higher in the biceps femoris compared to those in the control group. There were no significant differences in insulin-like growth factor 1 mRNA levels in the liver or skeletal muscle between groups. These findings suggested that supplementation with essential AAs ameliorated the impaired effects of an LP diet on growth performance in broiler chicks, and that the transcriptional changes in proteolytic genes in skeletal muscles might be related to the impaired effects of the LP diet.
Collapse
Affiliation(s)
- Asmaa S. El-far
- Graduate School of
Agricultural Science, Kobe University, Kobe
657-8501, Japan
- Faculty of
Veterinary Medicine, Damanhour University,
Damanhour, Egypt
| | - Maho Kamiya
- Graduate School of
Agricultural Science, Kobe University, Kobe
657-8501, Japan
| | - Takaoki Saneyasu
- Graduate School of
Agricultural Science, Kobe University, Kobe
657-8501, Japan
| | - Kazuhisa Honda
- Graduate School of
Agricultural Science, Kobe University, Kobe
657-8501, Japan
| |
Collapse
|
3
|
Chae J, Hahn D, Nam JO. Macamide, a component of maca (Lepidium meyenii Walp) lipophilic extract, enhances myogenic differentiation via AKT/p38 signaling and attenuates dexamethasone-induced muscle atrophy. Biomed Pharmacother 2024; 172:116249. [PMID: 38340399 DOI: 10.1016/j.biopha.2024.116249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/15/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Maca (Lepidium meyenii) is a plant that grows in the central Andes region of Peru, and it has been reported to have various bioactive functions, such as improving or preventing osteoporosis, sexual dysfunction, and memory impairment. In this study, maca roots of various colors (yellow, red, or black) were extracted using different polar solvents (PE, HEX, or BuOH) to compare their effects on muscle differentiation. Among them, the red maca lipophilic extract, which showed the most effectiveness, was chosen for further investigation. Our results show that RMLE enhances muscle differentiation by inducing MyoD-E2A heterodimerization through the activation of the AKT/p38 pathway. Additionally, RMLE attenuated dexamethasone-induced muscle atrophy by inhibiting nuclear translocation of FoxO3a and expression of E3-ligase (MAFbx and MURF1) in vitro and in vivo. Therefore, based on these results suggest that lipophilic extract of maca, which can abundantly contain nonpolar compounds, macamides, can enhance the functional properties of maca in alleviating muscle homeostasis.
Collapse
Affiliation(s)
- Jongbeom Chae
- Department of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Dongyup Hahn
- School of Food Science and Biotechnology, Institute of Agricultural Science and Technology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea.
| | - Ju-Ock Nam
- Department of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea; Research Institute of Tailored Food Technology, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
4
|
Alev K, Aru M, Vain A, Pehme A, Kaasik P, Seene T. Short-time recovery skeletal muscle from dexamethasone-induced atrophy and weakness in old female rats. Clin Biomech (Bristol, Avon) 2022; 100:105808. [PMID: 36368193 DOI: 10.1016/j.clinbiomech.2022.105808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Several pathological conditions (atrophy, dystrophy, spasticity, inflammation) can change muscle biomechanical parameters. Our previous works have shown that dexamethasone treatment changes skeletal muscle tone, stiffness, elasticity. Exercise training may oppose the side effects observed during dexamethasone treatment. The purpose of this study was to examine the changes in biomechanical parameters (tone, stiffness, elasticity) of skeletal muscle occurring during dexamethasone treatment and subsequent short-time recovery from glucocorticoid-induced muscle atrophy and weakness, as well as the effect of mild therapeutic exercise. METHODS 17 old female rats, aged 22 months were used in this study. The hand-held and non-invasive device (MyotonPRO, Myoton Ltd., Tallinn, Estonia) was used to study changes in biomechanical properties of muscle. Additionally, body and muscle mass, hind limb grip strength were assessed. FINDINGS Results showed that dexamethasone treatment alters muscle tone, stiffness and elasticity. During 20-day recovery period all measured parameters gradually improved towards the average baseline, however, remaining significantly lower than these values. The body and muscle mass, hind limb grip strength of the rats decreased considerably in the groups that received glucocorticoids. After 20 days of recovery, hind limb grip strength of the animals was slightly lower than the baseline value and mild therapeutic exercise had a slight but not significant effect on hind limb grip strength. Biomechanical parameters improved during the recovery period, but only dynamic stiffness and decrement retuned to baseline value. INTERPRETATION The study results show that monitoring muscle biomechanical parameters allows to assess the recovery of atrophied muscle from steroid myopathy.
Collapse
Affiliation(s)
- Karin Alev
- Institute of Sport Sciences and Physiotherapy, Faculty of Medicine University of Tartu, Estonia.
| | - Maire Aru
- Clinical Research Centre University of Tartu, Estonia
| | - Arved Vain
- Institute of Physics, Faculty of Science and Technology University Tartu, Estonia
| | - Ando Pehme
- Institute of Sport Sciences and Physiotherapy, Faculty of Medicine University of Tartu, Estonia
| | - Priit Kaasik
- Institute of Sport Sciences and Physiotherapy, Faculty of Medicine University of Tartu, Estonia
| | - Teet Seene
- Institute of Sport Sciences and Physiotherapy, Faculty of Medicine University of Tartu, Estonia
| |
Collapse
|
5
|
Felice F, Cesare MM, Fredianelli L, De Leo M, Conti V, Braca A, Di Stefano R. Effect of Tomato Peel Extract Grown under Drought Stress Condition in a Sarcopenia Model. Molecules 2022; 27:molecules27082563. [PMID: 35458760 PMCID: PMC9031685 DOI: 10.3390/molecules27082563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/16/2022] Open
Abstract
Tomatoes and their derivates represent an important source of natural biologically active components. The present study aims to investigate the protective effect of tomato peel extracts, grown in normal (RED-Ctr) or in drought stress (RED-Ds) conditions, on an experimental model of sarcopenia. The phenolic profile and total polyphenols content (TPC) of RED-Ctr and RED-Ds were determined by Ultra High-Performance Liquid Chromatography (UHPLC) analyses coupled to electrospray ionization high-resolution mass spectrometry (ESI-HR-MS). Human skeletal muscle myoblasts (HSMM) were differentiated in myotubes, and sarcopenia was induced by dexamethasone (DEXA) treatment. Differentiation and sarcopenia were evaluated by both real-time PCR and immunofluorescent techniques. Data show that myosin heavy chain 2 (MYH2), troponin T (TNNT1), and miogenin (MYOG) were expressed in differentiated myotubes. 5 μg Gallic Acid Equivalent (GAE/mL) of TPC from RED-Ds extract significantly reduced muscle atrophy induced by DEXA. Moreover, Forkhead BoxO1 (FOXO1) expression, involved in cell atrophy, was significantly decreased by RED-Ds extract. The protective effect of tomato peel extracts depended on their qualitative polyphenolic composition, resulting effectively in the in vitro model of sarcopenia.
Collapse
Affiliation(s)
- Francesca Felice
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy;
- Correspondence:
| | - Maria Michela Cesare
- Department of Life Sciences, University of Siena, Via P.A. Mattioli 4, 53100 Siena, Italy; (M.M.C.); (V.C.)
| | - Luca Fredianelli
- Institute for Chemical-Physical Processes of the Italian Research Council (CNR-IPCF), Via Moruzzi 1, 56100 Pisa, Italy;
| | - Marinella De Leo
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.L.); (A.B.)
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56100 Pisa, Italy
- CISUP, Centre for Instrumentation Sharing, University of Pisa, 56126 Pisa, Italy
| | - Veronica Conti
- Department of Life Sciences, University of Siena, Via P.A. Mattioli 4, 53100 Siena, Italy; (M.M.C.); (V.C.)
| | - Alessandra Braca
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.L.); (A.B.)
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56100 Pisa, Italy
- CISUP, Centre for Instrumentation Sharing, University of Pisa, 56126 Pisa, Italy
| | - Rossella Di Stefano
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy;
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.L.); (A.B.)
| |
Collapse
|
6
|
Chang YB, Ahn Y, Suh HJ, Jo K. Yeast hydrolysate ameliorates dexamethasone-induced muscle atrophy by suppressing MuRF-1 expression in C2C12 cells and C57BL/6 mice. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
7
|
Kim HJ, Lee JH, Kim SW, Lee SH, Jung DW, Williams DR. Investigation of niclosamide as a repurposing agent for skeletal muscle atrophy. PLoS One 2021; 16:e0252135. [PMID: 34038481 PMCID: PMC8153455 DOI: 10.1371/journal.pone.0252135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle atrophy is a feature of aging (termed sarcopenia) and various diseases, such as cancer and kidney failure. Effective drug treatment options for muscle atrophy are lacking. The tapeworm medication, niclosamide is being assessed for repurposing to treat numerous diseases, including end-stage cancer metastasis and hepatic steatosis. In this study, we investigated the potential of niclosamide as a repurposing drug for muscle atrophy. In a myotube atrophy model using the glucocorticoid, dexamethasone, niclosamide did not prevent the reduction in myotube diameter or the decreased expression of phosphorylated FOXO3a, which upregulates the ubiquitin-proteasome pathway of muscle catabolism. Treatment of normal myotubes with niclosamide did not activate mTOR, a major regulator of muscle protein synthesis, and increased the expression of atrogin-1, which is induced in catabolic states. Niclosamide treatment also inhibited myogenesis in muscle precursor cells, enhanced the expression of myoblast markers Pax7 and Myf5, and downregulated the expression of differentiation markers MyoD, MyoG and Myh2. In an animal model of muscle atrophy, niclosamide did not improve muscle mass, grip strength or muscle fiber cross-sectional area. Muscle atrophy is also feature of cancer cachexia. IC50 analyses indicated that niclosamide was more cytotoxic for myoblasts than cancer cells. In addition, niclosamide did not suppress the induction of iNOS, a key mediator of atrophy, in an in vitro model of cancer cachexia and did not rescue myotube diameter. Overall, these results suggest that niclosamide may not be a suitable repurposing drug for glucocorticoid-induced skeletal muscle atrophy or cancer cachexia. Nevertheless, niclosamide may be employed as a compound to study mechanisms regulating myogenesis and catabolic pathways in skeletal muscle.
Collapse
Affiliation(s)
- Hyun-Jun Kim
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Jeollanam-do, Republic of Korea
| | - Ji-Hyung Lee
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Jeollanam-do, Republic of Korea
| | - Seon-Wook Kim
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Jeollanam-do, Republic of Korea
| | - Sang-Hoon Lee
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Jeollanam-do, Republic of Korea
| | - Da-Woon Jung
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Jeollanam-do, Republic of Korea
- * E-mail: (D-WJ); (DRW)
| | - Darren R. Williams
- New Drug Targets Laboratory, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Jeollanam-do, Republic of Korea
- * E-mail: (D-WJ); (DRW)
| |
Collapse
|
8
|
Antioxidants in Sport Sarcopenia. Nutrients 2020; 12:nu12092869. [PMID: 32961753 PMCID: PMC7551250 DOI: 10.3390/nu12092869] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/12/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022] Open
Abstract
The decline of skeletal muscle mass and strength that leads to sarcopenia is a pathology that might represent an emergency healthcare issue in future years. Decreased muscle mass is also a condition that mainly affects master athletes involved in endurance physical activities. Skeletal muscles respond to exercise by reshaping the biochemical, morphological, and physiological state of myofibrils. Adaptive responses involve the activation of intracellular signaling pathways and genetic reprogramming, causing alterations in contractile properties, metabolic status, and muscle mass. One of the mechanisms leading to sarcopenia is an increase in reactive oxygen and nitrogen species levels and a reduction in enzymatic antioxidant protection. The present review shows the recent experimental models of sarcopenia that explore molecular mechanisms. Furthermore, the clinical aspect of sport sarcopenia will be highlighted, and new strategies based on nutritional supplements, which may contribute to reducing indices of oxidative stress by reinforcing natural endogenous protection, will be suggested.
Collapse
|
9
|
Tokinoya K, Shirai T, Ota Y, Takemasa T, Takekoshi K. Denervation-induced muscle atrophy suppression in renalase-deficient mice via increased protein synthesis. Physiol Rep 2020; 8:e14475. [PMID: 32741114 PMCID: PMC7395909 DOI: 10.14814/phy2.14475] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/17/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022] Open
Abstract
Denervation-induced muscle atrophy increases signaling through both protein degradation and synthesis pathways. Renalase is a flavin adenine dinucleotide-dependent amine oxidase that inhibits apoptosis and inflammation and promotes cell survival. This study aimed to elucidate the effect of renalase on denervation-induced muscle atrophy. We used 7-week-old renalase knock-out (KO) mice (a model of denervation-induced muscle atrophy) and wild-type (WT) mice (KO: n = 6, weight = 20-26 g; WT: n = 5, weight = 19-23 g). After their left legs were denervated, these mice were killed 1 week later. KO mice had lighter muscle weight than the WT mice. We observed an increase in molecular signaling through protein degradation pathway as well as oxidative stress in denervated muscles compared with that in sham-operated muscles in both WT and KO mice. Additionally, we also observed the main effect of renalase in WT and KO mice. Mitochondrial oxidative phosphorylation protein content was lower in denervated muscles than in sham-operated muscles in both WT and KO mice. However, a significant difference was noted in the reaction with Akt and p70S6K (components of the protein synthesis pathway) between WT and KO mice. In conclusion, mice with renalase deficiency demonstrated an attenuation of denervation-induced muscle atrophy. This might be related to catecholamines because signaling through the protein synthesis pathway was increased following denervation in renalase KO mice compared with that in WT mice, despite showing no change in signaling through protein degradation pathways.
Collapse
Affiliation(s)
- Katsuyuki Tokinoya
- Doctoral Program in Sports MedicineGraduate School of Comprehensive Human SciencesUniversity of TsukubaTsukubaJapan
- Research Fellow of the Japan Society for the Promotion of ScienceTokyoJapan
| | - Takanaga Shirai
- Graduate School of Comprehensive Human SciencesUniversity of TsukubaTsukubaJapan
| | - Yuya Ota
- Graduate School of Comprehensive Human SciencesUniversity of TsukubaTsukubaJapan
| | - Tohru Takemasa
- Faculty of Health and Sport SciencesUniversity of TsukubaTsukubaJapan
| | - Kazuhiro Takekoshi
- Division of Clinical MedicineFaculty of MedicineUniversity of TsukubaTsukubaJapan
| |
Collapse
|
10
|
Acevedo A, DuBois D, Almon RR, Jusko WJ, Androulakis IP. Modeling Pathway Dynamics of the Skeletal Muscle Response to Intravenous Methylprednisolone (MPL) Administration in Rats: Dosing and Tissue Effects. Front Bioeng Biotechnol 2020; 8:759. [PMID: 32760706 PMCID: PMC7371857 DOI: 10.3389/fbioe.2020.00759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 06/15/2020] [Indexed: 12/27/2022] Open
Abstract
A model-based approach for the assessment of pathway dynamics is explored to characterize metabolic and signaling pathway activity changes characteristic of the dosing-dependent differences in response to methylprednisolone in muscle. To consistently compare dosing-induced changes we extend the principles of pharmacokinetics and pharmacodynamics and introduce a novel representation of pathway-level dynamic models of activity regulation. We hypothesize the emergence of dosing-dependent regulatory interactions is critical to understanding the mechanistic implications of MPL dosing in muscle. Our results indicate that key pathways, including amino acid and lipid metabolism, signal transduction, endocrine regulation, regulation of cellular functions including growth, death, motility, transport, protein degradation, and catabolism are dependent on dosing, exhibiting diverse dynamics depending on whether the drug is administered acutely of continuously. Therefore, the dynamics of drug presentation offer the possibility for the emergence of dosing-dependent models of regulation. Finally, we compared acute and chronic MPL response in muscle with liver. The comparison revealed systematic response differences between the two tissues, notably that muscle appears more prone to adapt to MPL.
Collapse
Affiliation(s)
- Alison Acevedo
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, United States
| | - Debra DuBois
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, United States.,Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - Richard R Almon
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, United States.,Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - William J Jusko
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, United States.,Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - Ioannis P Androulakis
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, United States.,Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, NJ, United States.,Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| |
Collapse
|
11
|
Yoshioka Y, Samukawa Y, Yamashita Y, Ashida H. 4-Hydroxyderricin and xanthoangelol isolated from Angelica keiskei prevent dexamethasone-induced muscle loss. Food Funct 2020; 11:5498-5512. [PMID: 32510085 DOI: 10.1039/d0fo00720j] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Since a decrease in muscle mass leads to an increased risk of mortality, the prevention of muscle wasting contributes to maintaining the quality of life. Recently, we reported that glabridin, a prenylated flavonoid in licorice, prevents dexamethasone-induced muscle loss. In this study, we focused on the other prenylated chalcones 4-hydroxyderricin and xanthoangelol in Ashitaba (Angelica keiskei) and investigated their prevention effect on dexamethasone-induced muscle loss. It was found that 4-hydroxyderricin and xanthoangelol significantly prevented dexamethasone-induced protein degradation in C2C12 myotubes by suppressing the expression of ubiquitin ligases, Cbl-b and MuRF-1. These prenylated chalcones acted as the antagonists of the glucocorticoid receptor and inhibited the binding of dexamethasone to this receptor and its subsequent nuclear translocation. In addition, the chalcones suppressed the phosphorylation of p38 and FoxO3a as the upstream factors for ubiquitin ligases. Dexamethasone-induced protein degradation and upregulation of Cbl-b were attenuated by the knockdown of the glucocorticoid receptor but not by the knockdown of p38. In male C57BL/6J mice, the Ashitaba extract, containing 4-hydroxyderricin and xanthoangelol, suppressed dexamethasone-induced muscle mass wasting accompanied by a decrease in the expression of ubiquitin ligases by inhibiting the nuclear translocation of the glucocorticoid receptor and phosphorylation of FoxO3a. In conclusion, 4-hydroxyderricin and xanthoangelol are effective compounds to inhibit steroid-induced muscle loss.
Collapse
Affiliation(s)
- Yasukiyo Yoshioka
- Faculty of Clinical Nutrition and Dietetics, Konan Women's University, Kobe, Hyogo 658-0001, Japan
| | - Yumi Samukawa
- Graduate school of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan.
| | - Yoko Yamashita
- Graduate school of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan.
| | - Hitoshi Ashida
- Graduate school of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan.
| |
Collapse
|
12
|
BST Stimulation Induces Atrophy and Changes in Aerobic Energy Metabolism in Rat Skeletal Muscles-The Biphasic Action of Endogenous Glucocorticoids. Int J Mol Sci 2020; 21:ijms21082787. [PMID: 32316389 PMCID: PMC7216182 DOI: 10.3390/ijms21082787] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/12/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
(1) The primary involvement in stress-induced disturbances in skeletal muscles is assigned to the release of glucocorticoids (GCs). The current study aims to investigate the impact of the biphasic action of the chronic stress response (CSR) induced by the electrical stimulation of the bed nucleus of the stria terminalis (BST) effects on muscle atrophy and aerobic energy metabolism in soleus (SOL) and extensor digitorum longus (EDL) muscles. (2) Male Wistar rats (n = 17) were used. The rats were divided randomly into three groups: the BST two weeks (ST2), four weeks (ST4), and the sham (SHM) electrically stimulated group. The plasma corticosterone (CORT) and irisin concentration were measured. Glucocorticoid and mineralocorticoid receptors (GR and MR), 11β-hydroxysteroid dehydrogenase type 1 and 2 (HSD11B1 and HSD11B2), atrogin-1, and insulin-like growth factor-1 (IGF-1) level were determined in SOL and EDL muscles. Citrate synthase (CS) activity was measured in both muscles. (3) We found elevated plasma concentration of CORT and irisin, raised the level of GR in SOL muscle, and the higher level of MR in both muscles in the ST4 group. The level of HSD11B1 was also higher in the ST4 group compared to the SHM group. Moreover, we observed increased activity of CS in SOL. (4) We suggest that biphasic action of the glucocorticoid induced by the CSR occurs and causes dysregulation of proteins involved in muscle atrophy and aerobic energy metabolism. Our findings potentially contribute to a better understanding of the mechanisms by which GCs and the CSR may regulate muscle atrophy and energy preservation of the red muscle.
Collapse
|
13
|
Geng H, Song Q, Cheng Y, Li H, Yang R, Liu S, Hao L. MicroRNA 322 Aggravates Dexamethasone-Induced Muscle Atrophy by Targeting IGF1R and INSR. Int J Mol Sci 2020; 21:E1111. [PMID: 32046161 PMCID: PMC7043225 DOI: 10.3390/ijms21031111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/02/2020] [Accepted: 02/03/2020] [Indexed: 01/08/2023] Open
Abstract
Dexamethasone (Dex) has been widely used as a potent anti-inflammatory, antishock, and immunosuppressive agent. However, high dose or long-term use of Dex is accompanied by side effects including skeletal muscle atrophy, whose underlying mechanisms remain incompletely understood. A number of microRNAs (miRNAs) have been shown to play key roles in skeletal muscle atrophy. Previous studies showed significantly increased miR-322 expression in Dex-treated C2C12 myotubes. In our study, the glucocorticoid receptor (GR) was required for Dex to increase miR-322 expression in C2C12 myotubes. miR-322 mimic or miR-322 inhibitor was used for regulating the expression of miR-322. Insulin-like growth factor 1 receptor (IGF1R) and insulin receptor (INSR) were identified as target genes of miR-322 using luciferase reporter assays and played key roles in Dex-induced muscle atrophy. miR-322 overexpression promoted atrophy in Dex-treated C2C12 myotubes and the gastrocnemius muscles of mice. Conversely, miR-322 inhibition showed the opposite effects. These data suggested that miR-322 contributes to Dex-induced muscle atrophy via targeting of IGF1R and INSR. Furthermore, miR-322 might be a potential target to counter Dex-induced muscle atrophy. miR-322 inhibition might also represent a therapeutic approach for Dex-induced muscle atrophy.
Collapse
Affiliation(s)
- Hongwei Geng
- College of Animal Science, Jilin University, Changchun 130062, China; (H.G.); (Y.C.); (H.L.); (S.L.)
| | - Qinglong Song
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing 100193, China;
- Beijing Key Laboratory of Bio-Feed Additives, Beijing 100193, China
| | - Yunyun Cheng
- College of Animal Science, Jilin University, Changchun 130062, China; (H.G.); (Y.C.); (H.L.); (S.L.)
| | - Haoyang Li
- College of Animal Science, Jilin University, Changchun 130062, China; (H.G.); (Y.C.); (H.L.); (S.L.)
| | - Rui Yang
- College of Animal Science, Jilin University, Changchun 130062, China; (H.G.); (Y.C.); (H.L.); (S.L.)
| | - Songcai Liu
- College of Animal Science, Jilin University, Changchun 130062, China; (H.G.); (Y.C.); (H.L.); (S.L.)
- Five-Star Animal Health Pharmaceutical Factory of Jilin Province, Changchun 130062, China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun 130062, China; (H.G.); (Y.C.); (H.L.); (S.L.)
| |
Collapse
|
14
|
|
15
|
Pyropia yezoensis Protein Supplementation Prevents Dexamethasone-Induced Muscle Atrophy in C57BL/6 Mice. Mar Drugs 2018; 16:md16090328. [PMID: 30208614 PMCID: PMC6163250 DOI: 10.3390/md16090328] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/05/2018] [Accepted: 09/09/2018] [Indexed: 12/29/2022] Open
Abstract
We investigated the protective effects of Pyropia yezoensis crude protein (PYCP) against dexamethasone (DEX)-induced myotube atrophy and its underlying mechanisms. DEX (3 mg/kg body weight, intraperitoneal injection) and PYCP (150 and 300 mg/kg body weight, oral) were administrated to mice for 18 days, and the effects of PYCP on DEX-induced muscle atrophy were evaluated. Body weight, calf thickness, and gastrocnemius and tibialis anterior muscle weight were significantly decreased by DEX administration (p < 0.05), while PYCP supplementation effectively prevented the DEX-induced decrease in body weight, calf thickness, and muscle weight. PYCP supplementation also attenuated the DEX-induced increase in serum glucose, creatine kinase, and lactate dehydrogenase levels. Additionally, PYCP supplementation reversed DEX-induced muscle atrophy via the regulation of the insulin-like growth factor-I/protein kinase B/rapamycin-sensitive mTOR complex I/forkhead box O signaling pathway. The mechanistic investigation revealed that PYCP inhibited the ubiquitin-proteasome and autophagy-lysosome pathways in DEX-administrated C57BL/6 mice. These findings demonstrated that PYCP increased protein synthesis and decreased protein breakdown to prevent muscle atrophy. Therefore, PYCP supplementation appears to be useful for preventing muscle atrophy.
Collapse
|
16
|
|
17
|
Zhang Y, Pan X, Sun Y, Geng YJ, Yu XY, Li Y. The Molecular Mechanisms and Prevention Principles of Muscle Atrophy in Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:347-368. [PMID: 30390260 DOI: 10.1007/978-981-13-1435-3_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Muscle atrophy in aging is characterized by progressive loss of muscle mass and function. Muscle mass is determined by the balance of synthesis and degradation of protein, which are regulated by several signaling pathways such as ubiquitin-proteasome system, autophagy-lysosome systems, oxidative stress, proinflammatory cytokines, hormones, and so on. Sufficient nutrition can enhance protein synthesis, while exercise can improve the quality of life in the elderly. This chapter will discuss the epidemiology, pathogenesis, as well as the current treatment for aging-induced muscular atrophy.
Collapse
Affiliation(s)
- Yu Zhang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Xiangbin Pan
- Department of Cardiac Surgery, Fuwai Hospital, Beijing, People's Republic of China
| | - Yi Sun
- Fuwai Yunnan Cardiovascular Hospital, Kunming, Yunnan, People's Republic of China
| | | | - Xi-Yong Yu
- Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yangxin Li
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
18
|
Excessive glucocorticoid-induced muscle MuRF1 overexpression is independent of Akt/FoXO1 pathway. Biosci Rep 2017; 37:BSR20171056. [PMID: 29046370 PMCID: PMC5691142 DOI: 10.1042/bsr20171056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/02/2017] [Accepted: 10/10/2017] [Indexed: 12/04/2022] Open
Abstract
The ubiquitin-proteasome system (UPS)-dependent proteolysis plays a major role in the muscle catabolic action of glucocorticoids (GCs). Atrogin-1 and muscle-specific RING finger protein 1 (MuRF1), two E3 ubiquitin ligases, are uniquely expressed in muscle. It has been previously demonstrated that GC treatment induced MuRF1 and atrogin-1 overexpression. However, it is yet unclear whether the higher pharmacological dose of GCs induced muscle protein catabolism through MuRF1 and atrogin-1. In the present study, the role of atrogin-1 and MuRF1 in C2C12 cells protein metabolism during excessive dexamethasone (DEX) was studied. The involvement of Akt/forkhead box O1 (FoXO1) signaling pathway and the cross-talk between anabolic regulator mammalian target of rapamycin (mTOR) and catabolic regulator FoXO1 were investigated. High concentration of DEX increased MuRF1 protein level in a time-dependent fashion (P<0.05), while had no detectable effect on atrogin-1 protein (P>0.05). FoXO1/3a (Thr24/32) phosphorylation was enhanced (P<0.05), mTOR phosphorylation was suppressed (P<0.05), while Akt protein expression was not affected (P>0.05) by DEX. RU486 treatment inhibited the DEX-induced increase of FoXO1/3a phosphorylation (P<0.05) and MuRF1 protein; LY294002 (LY) did not restore the stimulative effect of DEX on the FoXO1/3a phosphorylation (P>0.05), but inhibited the activation of MuRF1 protein induced by DEX (P<0.05); rapamycin (RAPA) inhibited the stimulative effect of DEX on the FoXO1/3a phosphorylation and MuRF1 protein (P<0.05).
Collapse
|
19
|
Son YH, Jang EJ, Kim YW, Lee JH. Sulforaphane prevents dexamethasone-induced muscle atrophy via regulation of the Akt/Foxo1 axis in C2C12 myotubes. Biomed Pharmacother 2017; 95:1486-1492. [DOI: 10.1016/j.biopha.2017.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/18/2017] [Accepted: 09/03/2017] [Indexed: 01/06/2023] Open
|
20
|
Kang SH, Lee HA, Kim M, Lee E, Sohn UD, Kim I. Forkhead box O3 plays a role in skeletal muscle atrophy through expression of E3 ubiquitin ligases MuRF-1 and atrogin-1 in Cushing's syndrome. Am J Physiol Endocrinol Metab 2017; 312:E495-E507. [PMID: 28246104 DOI: 10.1152/ajpendo.00389.2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 11/22/2022]
Abstract
Cushing's syndrome is caused by overproduction of the adrenocorticotropic hormone (ACTH), which stimulates the adrenal grand to make cortisol. Skeletal muscle wasting occurs in pathophysiological response to Cushing's syndrome. The forkhead box (FOX) protein family has been implicated as a key regulator of muscle loss under conditions such as diabetes and sepsis. However, the mechanistic role of the FOXO family in ACTH-induced muscle atrophy is not understood. We hypothesized that FOXO3a plays a role in muscle atrophy through expression of the E3 ubiquitin ligases, muscle RING finger protein-1 (MuRF-1), and atrogin-1 in Cushing's syndrome. For establishment of a Cushing's syndrome animal model, Sprague-Dawley rats were implanted with osmotic minipumps containing ACTH (40 ng·kg-1·day-1). ACTH infusion significantly reduced muscle weight. In ACTH-infused rats, MuRF-1, atrogin-1, and FOXO3a were upregulated and the FOXO3a promoter was targeted by the glucocorticoid receptor (GR). Transcriptional activity and expression of FOXO3a were significantly decreased by the GR antagonist RU486. Treatment with RU486 reduced MuRF-1 and atrogin-1 expression in accordance with reduced enrichment of FOXO3a and Pol II on the promoters. Knockdown of FOXO3a prevented dexamethasone-induced MuRF-1 and atrogin-1 expression. These results indicate that FOXO3a plays a role in muscle atrophy through expression of MuRF-1 and atrogin-1 in Cushing's syndrome.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Animals
- Cell Line
- Chromatin Immunoprecipitation
- Cushing Syndrome/metabolism
- Cushing Syndrome/pathology
- Cushing Syndrome/physiopathology
- Disease Models, Animal
- Forkhead Box Protein O3/agonists
- Forkhead Box Protein O3/antagonists & inhibitors
- Forkhead Box Protein O3/genetics
- Forkhead Box Protein O3/metabolism
- Gene Expression Regulation/drug effects
- Genes, Reporter/drug effects
- Glucocorticoids/pharmacology
- Hormone Antagonists/pharmacology
- Male
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Proteins/agonists
- Muscle Proteins/antagonists & inhibitors
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy/etiology
- Promoter Regions, Genetic/drug effects
- RNA Interference
- Rats, Sprague-Dawley
- Receptors, Glucocorticoid/agonists
- Receptors, Glucocorticoid/antagonists & inhibitors
- Receptors, Glucocorticoid/metabolism
- Response Elements/drug effects
- SKP Cullin F-Box Protein Ligases/antagonists & inhibitors
- SKP Cullin F-Box Protein Ligases/genetics
- SKP Cullin F-Box Protein Ligases/metabolism
- Tripartite Motif Proteins/agonists
- Tripartite Motif Proteins/antagonists & inhibitors
- Tripartite Motif Proteins/genetics
- Tripartite Motif Proteins/metabolism
- Ubiquitin-Protein Ligases/antagonists & inhibitors
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
Collapse
Affiliation(s)
- Seol-Hee Kang
- Department of Pharmacology, Cardiovascular Research Institute, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
- BK21 Plus Kyungpook National University Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Hae-Ahm Lee
- Department of Pharmacology, Cardiovascular Research Institute, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Mina Kim
- Department of Pharmacology, Cardiovascular Research Institute, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
- BK21 Plus Kyungpook National University Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Eunjo Lee
- Department of Pharmacology, Cardiovascular Research Institute, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
- BK21 Plus Kyungpook National University Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Uy Dong Sohn
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea; and
| | - Inkyeom Kim
- Department of Pharmacology, Cardiovascular Research Institute, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea;
- BK21 Plus Kyungpook National University Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
21
|
Lee MK, Kim YM, Kim IH, Choi YH, Nam TJ. Pyropia yezoensis peptide PYP1‑5 protects against dexamethasone‑induced muscle atrophy through the downregulation of atrogin1/MAFbx and MuRF1 in mouse C2C12 myotubes. Mol Med Rep 2017; 15:3507-3514. [PMID: 28393223 PMCID: PMC5436292 DOI: 10.3892/mmr.2017.6443] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 02/02/2017] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle atrophy refers to the decline in muscle mass and strength that occurs under various conditions, including aging, starvation, cancer and other cachectic diseases. Muscle atrophy caused by aging, known as sarcopenia, primarily occurs after 50 years of age. Muscle atrophy‑related genes, including atrogin1/muscle atrophy F‑box (MAFbx) and muscle RING finger 1 (MuRF1), are expressed early in the muscle atrophy process, and their expression precedes the loss of muscle mass. The present study investigated the potential anti‑atrophic effects of the Pyropia yezoensis peptide PYP1‑5. The MTS assay did not detect cytotoxic effects of PYP1‑5 on C2C12 mouse myoblast cells. Subsequently, the anti‑atrophic effects of PYP1‑5 on skeletal muscle cells was examined by treating C2C12 myotubes with 100 µM dexamethasone (DEX) and/or 500 ng/ml PYP1‑5 for 24 h. Compared with the control, myotube diameter was reduced in DEX‑treated cells, whereas PYP1‑5 treatment protected against DEX‑induced muscle atrophy. MAFbx and MuRF1 protein and mRNA expression levels were detected by western blot analysis and reverse transcription‑quantitative polymerase chain reaction, respectively. The results demonstrated that PYP1‑5 significantly reduced the expression of atrogin1/MAFbx and MuRF1. Therefore, data from the present study suggest that PYP1‑5 inhibits the expression of atrogin1/MAFbx and MuRF1 in C2C12 cells, and these characteristics may be of value in the development of anti‑atrophy functional foods.
Collapse
Affiliation(s)
- Min-Kyeong Lee
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Min Kim
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - In-Hye Kim
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Youn-Hee Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Taek-Jeong Nam
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
22
|
Leucine alleviates dexamethasone-induced suppression of muscle protein synthesis via synergy involvement of mTOR and AMPK pathways. Biosci Rep 2016; 36:BSR20160096. [PMID: 27129299 PMCID: PMC5293580 DOI: 10.1042/bsr20160096] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 04/28/2016] [Indexed: 12/19/2022] Open
Abstract
Both mTOR and AMPK pathways are involved in the DEX-induced suppression of protein synthesis in muscle cells. Leucine supplementation relieves DEX-induced inhibition on protein synthesis by evoking mTOR and suppressing AMPK pathway. Glucocorticoids (GCs) are negative muscle protein regulators that contribute to the whole-body catabolic state during stress. Mammalian target of rapamycin (mTOR)-signalling pathway, which acts as a central regulator of protein metabolism, can be activated by branched-chain amino acids (BCAA). In the present study, the effect of leucine on the suppression of protein synthesis induced by GCs and the pathway involved were investigated. In vitro experiments were conducted using cultured C2C12 myoblasts to study the effect of GCs on protein synthesis, and the involvement of mTOR pathway was investigated as well. After exposure to dexamethasone (DEX, 100 μmol/l) for 24 h, protein synthesis in muscle cells was significantly suppressed (P<0.05), the phosphorylations of mTOR, ribosomal protein S6 protein kinase 1 (p70s6k1) and eukaryotic initiation factor 4E binding protein 1 (4EBP1) were significantly reduced (P<0.05). Leucine supplementation (5 mmol/l, 10 mmol/l and 15 mmol/l) for 1 h alleviated the suppression of protein synthesis induced by DEX (P<0.05) and was accompanied with the increased phosphorylation of mTOR and decreased phosphorylation of AMPK (P<0.05). Branched-chain amino transferase 2 (BCAT2) mRNA level was not influenced by DEX (P>0.05) but was increased by leucine supplementation at a dose of 5 mmol/l (P<0.05).
Collapse
|
23
|
Hinds TD, Peck B, Shek E, Stroup S, Hinson J, Arthur S, Marino JS. Overexpression of Glucocorticoid Receptor β Enhances Myogenesis and Reduces Catabolic Gene Expression. Int J Mol Sci 2016; 17:232. [PMID: 26875982 PMCID: PMC4783964 DOI: 10.3390/ijms17020232] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 11/16/2022] Open
Abstract
Unlike the glucocorticoid receptor α (GRα), GR β (GRβ) has a truncated ligand-binding domain that prevents glucocorticoid binding, implicating GRα as the mediator of glucocorticoid-induced skeletal muscle loss. Because GRβ causes glucocorticoid resistance, targeting GRβ may be beneficial in impairing muscle loss as a result of GRα activity. The purpose of this study was to determine how the overexpression of GRβ affects myotube formation and dexamethasone (Dex) responsiveness. We measured GR isoform expression in C₂C12 muscle cells in response to Dex and insulin, and through four days of myotube formation. Next, lentiviral-mediated overexpression of GRβ in C₂C12 was performed, and these cells were characterized for cell fusion and myotube formation, as well as sensitivity to Dex via the expression of ubiquitin ligases. GRβ overexpression increased mRNA levels of muscle regulatory factors and enhanced proliferation in myoblasts. GRβ overexpressing myotubes had an increased fusion index. Myotubes overexpressing GRβ had lower forkhead box O3 (Foxo3a) mRNA levels and a blunted muscle atrophy F-box/Atrogen-1 (MAFbx) and muscle ring finger 1 (MuRF1) response to Dex. We showed that GRβ may serve as a pharmacological target for skeletal muscle growth and protection from glucocorticoid-induced catabolic signaling. Increasing GRβ levels in skeletal muscle may cause a state of glucocorticoid resistance, stabilizing muscle mass during exposure to high doses of glucocorticoids.
Collapse
Affiliation(s)
- Terry D Hinds
- Center for Hypertension and Personalized Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA.
| | - Bailey Peck
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina Charlotte, Charlotte, NC 28223, USA.
| | - Evan Shek
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina Charlotte, Charlotte, NC 28223, USA.
| | - Steven Stroup
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina Charlotte, Charlotte, NC 28223, USA.
| | - Jennifer Hinson
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina Charlotte, Charlotte, NC 28223, USA.
| | - Susan Arthur
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina Charlotte, Charlotte, NC 28223, USA.
| | - Joseph S Marino
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina Charlotte, Charlotte, NC 28223, USA.
| |
Collapse
|
24
|
Evaluation of follistatin as a therapeutic in models of skeletal muscle atrophy associated with denervation and tenotomy. Sci Rep 2015; 5:17535. [PMID: 26657343 PMCID: PMC4675991 DOI: 10.1038/srep17535] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 11/02/2015] [Indexed: 01/25/2023] Open
Abstract
Follistatin is an inhibitor of TGF-β superfamily ligands that repress skeletal muscle growth and promote muscle wasting. Accordingly, follistatin has emerged as a potential therapeutic to ameliorate the deleterious effects of muscle atrophy. However, it remains unclear whether the anabolic effects of follistatin are conserved across different modes of non-degenerative muscle wasting. In this study, the delivery of a recombinant adeno-associated viral vector expressing follistatin (rAAV:Fst) to the hind-limb musculature of mice two weeks prior to denervation or tenotomy promoted muscle hypertrophy that was sufficient to preserve muscle mass comparable to that of untreated sham-operated muscles. However, administration of rAAV:Fst to muscles at the time of denervation or tenotomy did not prevent subsequent muscle wasting. Administration of rAAV:Fst to innervated or denervated muscles increased protein synthesis, but markedly reduced protein degradation only in innervated muscles. Phosphorylation of the signalling proteins mTOR and S6RP, which are associated with protein synthesis, was increased in innervated muscles administered rAAV:Fst, but not in treated denervated muscles. These results demonstrate that the anabolic effects of follistatin are influenced by the interaction between muscle fibres and motor nerves. These findings have important implications for understanding the potential efficacy of follistatin-based therapies for non-degenerative muscle wasting.
Collapse
|
25
|
Bodine SC, Furlow JD. Glucocorticoids and Skeletal Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [PMID: 26215994 DOI: 10.1007/978-1-4939-2895-8_7] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glucocorticoids are known to regulate protein metabolism in skeletal muscle, producing a catabolic effect that is opposite that of insulin. In many catabolic diseases, such as sepsis, starvation, and cancer cachexia, endogenous glucocorticoids are elevated contributing to the loss of muscle mass and function. Further, exogenous glucocorticoids are often given acutely and chronically to treat inflammatory conditions such as asthma, chronic obstructive pulmonary disease, and rheumatoid arthritis, resulting in muscle atrophy. This chapter will detail the nature of glucocorticoid-induced muscle atrophy and discuss the mechanisms thought to be responsible for the catabolic effects of glucocorticoids on muscle.
Collapse
Affiliation(s)
- Sue C Bodine
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA,
| | | |
Collapse
|
26
|
Pearson SJ, Hussain SR. A review on the mechanisms of blood-flow restriction resistance training-induced muscle hypertrophy. Sports Med 2015; 45:187-200. [PMID: 25249278 DOI: 10.1007/s40279-014-0264-9] [Citation(s) in RCA: 258] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
It has traditionally been believed that resistance training can only induce muscle growth when the exercise intensity is greater than 65% of the 1-repetition maximum (RM). However, more recently, the use of low-intensity resistance exercise with blood-flow restriction (BFR) has challenged this theory and consistently shown that hypertrophic adaptations can be induced with much lower exercise intensities (<50% 1-RM). Despite the potent hypertrophic effects of BFR resistance training being demonstrated by numerous studies, the underlying mechanisms responsible for such effects are not well defined. Metabolic stress has been suggested to be a primary factor responsible, and this is theorised to activate numerous other mechanisms, all of which are thought to induce muscle growth via autocrine and/or paracrine actions. However, it is noteworthy that some of these mechanisms do not appear to be mediated to any great extent by metabolic stress but rather by mechanical tension (another primary factor of muscle hypertrophy). Given that the level of mechanical tension is typically low with BFR resistance exercise (<50% 1-RM), one may question the magnitude of involvement of these mechanisms aligned to the adaptations reported with BFR resistance training. However, despite the low level of mechanical tension, it is plausible that the effects induced by the primary factors (mechanical tension and metabolic stress) are, in fact, additive, which ultimately contributes to the adaptations seen with BFR resistance training. Exercise-induced mechanical tension and metabolic stress are theorised to signal a number of mechanisms for the induction of muscle growth, including increased fast-twitch fibre recruitment, mechanotransduction, muscle damage, systemic and localised hormone production, cell swelling, and the production of reactive oxygen species and its variants, including nitric oxide and heat shock proteins. However, the relative extent to which these specific mechanisms are induced by the primary factors with BFR resistance exercise, as well as their magnitude of involvement in BFR resistance training-induced muscle hypertrophy, requires further exploration.
Collapse
Affiliation(s)
- Stephen John Pearson
- Centre for Health, Sport and Rehabilitation Sciences Research, University of Salford, Manchester, M6 6PU, UK,
| | | |
Collapse
|
27
|
Son YH, Lee SJ, Lee KB, Lee JH, Jeong EM, Chung SG, Park SC, Kim IG. Dexamethasone downregulates caveolin-1 causing muscle atrophy via inhibited insulin signaling. J Endocrinol 2015; 225:27-37. [PMID: 25688118 DOI: 10.1530/joe-14-0490] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Glucocorticoids play a major role in the development of muscle atrophy in various medical conditions, such as cancer, burn injury, and sepsis, by inhibiting insulin signaling. In this study, we report a new pathway in which glucocorticoids reduce the levels of upstream insulin signaling components by downregulating the transcription of the gene encoding caveolin-1 (CAV1), a scaffolding protein present in the caveolar membrane. Treatment with the glucocorticoid dexamethasone (DEX) decreased CAV1 protein and Cav1 mRNA expression, with a concomitant reduction in insulin receptor alpha (IRα) and IR substrate 1 (IRS1) levels in C2C12 myotubes. On the basis of the results of promoter analysis using deletion mutants and site-directed mutagenesis a negative glucocorticoid-response element in the regulatory region of the Cav1 gene was identified, confirming that Cav1 is a glucocorticoid-target gene. Cav1 knockdown using siRNA decreased the protein levels of IRα and IRS1, and overexpression of Cav1 prevented the DEX-induced decrease in IRα and IRS1 proteins, demonstrating a causal role of Cav1 in the inhibition of insulin signaling. Moreover, injection of adenovirus expressing Cav1 into the gastrocnemius muscle of mice prevented DEX-induced atrophy. These results indicate that CAV1 is a critical regulator of muscle homeostasis, linking glucocorticoid signaling to the insulin signaling pathway, thereby providing a novel target for the prevention of glucocorticoid-induced muscle atrophy.
Collapse
Affiliation(s)
- Young Hoon Son
- Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea
| | - Seok-Jin Lee
- Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea
| | - Ki-Baek Lee
- Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea
| | - Jin-Haeng Lee
- Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea
| | - Eui Man Jeong
- Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea
| | - Sun Gun Chung
- Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea
| | - Sang-Chul Park
- Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea
| | - In-Gyu Kim
- Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea Department of Biochemistry and Molecular BiologyInstitute of Human-Environment Interface BiologyDepartment of Rehabilitation MedicineSeoul National University College of Medicine, 103 Daehak-ro, Jongno-Gu, Seoul 110-799, Korea
| |
Collapse
|
28
|
Schmidt U, Buell DR, Ionescu IA, Gassen NC, Holsboer F, Cox MB, Novak B, Huber C, Hartmann J, Schmidt MV, Touma C, Rein T, Herrmann L. A role for synapsin in FKBP51 modulation of stress responsiveness: Convergent evidence from animal and human studies. Psychoneuroendocrinology 2015; 52:43-58. [PMID: 25459892 DOI: 10.1016/j.psyneuen.2014.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/04/2014] [Accepted: 11/04/2014] [Indexed: 01/09/2023]
Abstract
Both the molecular co-chaperone FKBP51 and the presynaptic vesicle protein synapsin (alternatively spliced from SYN1-3) are intensively discussed players in the still insufficiently explored pathobiology of psychiatric disorders such as major depression, schizophrenia and posttraumatic stress disorder (PTSD). To address their still unknown interaction, we compared the expression levels of synapsin and five other neurostructural and HPA axis related marker proteins in the prefrontal cortex (PFC) and the hippocampus of restrained-stressed and unstressed Fkbp5 knockout mice and corresponding wild-type littermates. In addition, we compared and correlated the gene expression levels of SYN1, SYN2 and FKBP5 in three different online datasets comprising expression data of human healthy subjects as well as of predominantly medicated patients with different psychiatric disorders. In summary, we found that Fkbp5 deletion, which we previously demonstrated to improve stress-coping behavior in mice, prevents the stress-induced decline in prefrontal cortical (pc), but not in hippocampal synapsin expression. Accordingly, pc, but not hippocampal, synapsin protein levels correlated positively with a more active mouse stress coping behavior. Searching for an underlying mechanism, we found evidence that deletion of Fkbp5 might prevent stress-induced pc synapsin loss, at least in part, through improvement of pc Akt kinase activity. These results, together with our finding that FKBP5 and SYN1 mRNA levels were regulated in opposite directions in the PFC of schizophrenic patients, who are known for exhibiting an altered stress-coping behavior, provide the first evidence of a role for pc synapsin in FKBP51 modulation of stress responsiveness. This role might extend to other tissues, as we found FKBP5 and SYN1 levels to correlate inversely not only in human PFC samples but also in other expression sites. The main limitation of this study is the small number of individuals included in the correlation analyses. Future studies will have to verify the here-postulated role of the FKBP51-Akt kinase-synapsin pathway in stress responsiveness.
Collapse
Affiliation(s)
- Ulrike Schmidt
- Max Planck Institute of Psychiatry, Department of Clinical Research, RG Molecular Psychotraumatology, Munich, Germany.
| | - Dominik R Buell
- Max Planck Institute of Psychiatry, Department of Clinical Research, RG Molecular Psychotraumatology, Munich, Germany
| | - Irina A Ionescu
- Max Planck Institute of Psychiatry, Department of Clinical Research, RG Molecular Psychotraumatology, Munich, Germany
| | - Nils C Gassen
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Germany
| | - Florian Holsboer
- Max Planck Institute of Psychiatry, Department of Clinical Research, Munich, Germany
| | - Marc B Cox
- University of Texas at El Paso, 500 West University Avenue, El Paso, TX 79968, United States
| | - Bozidar Novak
- Max Planck Institute of Psychiatry, Department of Clinical Research, RG Molecular Psychotraumatology, Munich, Germany
| | - Christine Huber
- Max Planck Institute of Psychiatry, Department of Clinical Research, RG Molecular Psychotraumatology, Munich, Germany
| | - Jakob Hartmann
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, Germany
| | - Mathias V Schmidt
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, Germany
| | - Chadi Touma
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, Germany
| | - Theo Rein
- Max Planck Institute of Psychiatry, Department of Translational Research in Psychiatry, Germany
| | - Leonie Herrmann
- Max Planck Institute of Psychiatry, Department of Clinical Research, RG Molecular Psychotraumatology, Munich, Germany
| |
Collapse
|
29
|
Ye F, McCoy SC, Ross HH, Bernardo JA, Beharry AW, Senf SM, Judge AR, Beck DT, Conover CF, Cannady DF, Smith BK, Yarrow JF, Borst SE. Transcriptional regulation of myotrophic actions by testosterone and trenbolone on androgen-responsive muscle. Steroids 2014; 87:59-66. [PMID: 24928725 PMCID: PMC8396102 DOI: 10.1016/j.steroids.2014.05.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 05/23/2014] [Accepted: 05/27/2014] [Indexed: 12/31/2022]
Abstract
Androgens regulate body composition and skeletal muscle mass in males, but the molecular mechanisms are not fully understood. Recently, we demonstrated that trenbolone (a potent synthetic testosterone analogue that is not a substrate for 5-alpha reductase or for aromatase) induces myotrophic effects in skeletal muscle without causing prostate enlargement, which is in contrast to the known prostate enlarging effects of testosterone. These previous results suggest that the 5α-reduction of testosterone is not required for myotrophic action. We now report differential gene expression in response to testosterone versus trenbolone in the highly androgen-sensitive levator ani/bulbocavernosus (LABC) muscle complex of the adult rat after 6weeks of orchiectomy (ORX), using real time PCR. The ORX-induced expression of atrogenes (Muscle RING-finger protein-1 [MuRF1] and atrogin-1) was suppressed by both androgens, with trenbolone producing a greater suppression of atrogin-1 mRNA compared to testosterone. Both androgens elevated expression of anabolic genes (insulin-like growth factor-1 and mechano-growth factor) after ORX. ORX-induced increases in expression of glucocorticoid receptor (GR) mRNA were suppressed by trenbolone treatment, but not testosterone. In ORX animals, testosterone promoted WNT1-inducible-signaling pathway protein 2 (WISP-2) gene expression while trenbolone did not. Testosterone and trenbolone equally enhanced muscle regeneration as shown by increases in LABC mass and in protein expression of embryonic myosin by western blotting. In addition, testosterone increased WISP-2 protein levels. Together, these findings identify specific mechanisms by which testosterone and trenbolone may regulate skeletal muscle maintenance and growth.
Collapse
Affiliation(s)
- Fan Ye
- Geriatric Research, Education and Clinical Center, VA Medical Center, Gainesville, FL, United States; Applied Physiology & Kinesiology, University of Florida, Gainesville, FL, United States.
| | - Sean C McCoy
- Rural Health, VA Medical Center, Gainesville, FL, United States; Animal Sciences, University of Florida, Gainesville, FL, United States
| | - Heather H Ross
- Physical Therapy, University of Florida, Gainesville, FL, United States
| | - Joseph A Bernardo
- Physical Therapy, University of Florida, Gainesville, FL, United States
| | - Adam W Beharry
- Physical Therapy, University of Florida, Gainesville, FL, United States
| | - Sarah M Senf
- Physical Therapy, University of Florida, Gainesville, FL, United States
| | - Andrew R Judge
- Physical Therapy, University of Florida, Gainesville, FL, United States
| | - Darren T Beck
- Geriatric Research, Education and Clinical Center, VA Medical Center, Gainesville, FL, United States
| | - Christine F Conover
- Geriatric Research, Education and Clinical Center, VA Medical Center, Gainesville, FL, United States
| | - Darryl F Cannady
- Geriatric Research, Education and Clinical Center, VA Medical Center, Gainesville, FL, United States
| | - Barbara K Smith
- Physical Therapy, University of Florida, Gainesville, FL, United States
| | - Joshua F Yarrow
- Applied Physiology & Kinesiology, University of Florida, Gainesville, FL, United States; Research, VA Medical Center, Gainesville, FL, United States
| | - Stephen E Borst
- Geriatric Research, Education and Clinical Center, VA Medical Center, Gainesville, FL, United States; Applied Physiology & Kinesiology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
30
|
Feng B, He Q, Xu H. FOXO1-dependent up-regulation of MAP kinase phosphatase 3 (MKP-3) mediates glucocorticoid-induced hepatic lipid accumulation in mice. Mol Cell Endocrinol 2014; 393:46-55. [PMID: 24946098 PMCID: PMC4130747 DOI: 10.1016/j.mce.2014.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 05/15/2014] [Accepted: 06/04/2014] [Indexed: 11/16/2022]
Abstract
Long-term treatment with glucocorticoids (GCs) or dysregulation of endogenous GC levels induces a series of metabolic diseases, such as insulin resistance, obesity and type 2 diabetes. We previously showed that MAP kinase phosphatase-3 (MKP-3) plays an important role in glucose metabolism. The aim of this study is to investigate the role of MKP-3 in GC-induced metabolic disorders. Dexamethasone (Dex), a synthetic GC, increases MKP-3 protein expression both in cultured hepatoma cells and in the liver of lean mice. This effect is likely mediated by forkhead box protein O1 (FOXO1) because disruption of endogenous FOXO1 function by either interfering RNA mediated FOXO1 knockdown or overexpression of a dominant negative FOXO1 mutant blocks Dex-induced upregulation of MKP-3 protein. In addition, overexpression of FOXO1 is sufficient to induce MKP-3 protein expression. MKP-3 deficient mice are protected from several side effects of chronic Dex exposure, such as body weight gain, adipose tissue enlargement, hepatic lipid accumulation, and insulin resistance. The beneficial phenotypes in mice lacking MKP-3 are largely attributed to the absence of MKP-3 in the liver since only hepatic insulin signaling has been preserved among the three insulin target tissues (liver, muscle and adipose tissue).
Collapse
Affiliation(s)
- Bin Feng
- Hallett Center for Diabetes and Endocrinology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA.
| | - Qin He
- Hallett Center for Diabetes and Endocrinology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA.
| | - Haiyan Xu
- Hallett Center for Diabetes and Endocrinology, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA.
| |
Collapse
|
31
|
Patel R, Williams-Dautovich J, Cummins CL. Minireview: new molecular mediators of glucocorticoid receptor activity in metabolic tissues. Mol Endocrinol 2014; 28:999-1011. [PMID: 24766141 DOI: 10.1210/me.2014-1062] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The glucocorticoid receptor (GR) was one of the first nuclear hormone receptors cloned and represents one of the most effective drug targets available today for the treatment of severe inflammation. The physiologic consequences of endogenous or exogenous glucocorticoid excess are well established and include hyperglycemia, insulin resistance, fatty liver, obesity, and muscle wasting. However, at the molecular and tissue-specific level, there are still many unknown protein mediators of glucocorticoid response and thus, much remains to be uncovered that will help determine whether activation of the GR can be tailored to improve therapeutic efficacy while minimizing unwanted side effects. This review summarizes recent discoveries of tissue-selective modulators of glucocorticoid signaling that are important in mediating the unwanted side effects of therapeutic glucocorticoid use, emphasizing the downstream molecular effects of GR activation in the liver, adipose tissue, muscle, and pancreas.
Collapse
Affiliation(s)
- Rucha Patel
- Department of Pharmaceutical Sciences (R.P., J.W-D., C.L.C.), University of Toronto, Toronto, Ontario, M5S 3M2, Canada; and Banting and Best Diabetes Centre (C.L.C.), Toronto, Ontario M5G 2C4 Canada
| | | | | |
Collapse
|
32
|
Molecular patterns of neurodevelopmental preconditioning: a study of the effects of antenatal steroid therapy in a protein-restriction mouse model. ISRN OBSTETRICS AND GYNECOLOGY 2014; 2014:193816. [PMID: 25006477 PMCID: PMC3976831 DOI: 10.1155/2014/193816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 12/25/2013] [Indexed: 01/21/2023]
Abstract
Introduction. Prenatal programming secondary to maternal protein restriction renders an inherent susceptibility to neural compromise in neonates and any addition of glucocorticosteroids results in further damage. This is an investigation of consequent global gene activity due to effects of antenatal steroid therapy on a protein restriction mouse model. Methods. C57BL/6N pregnant mice were administered control or protein restricted diets and subjected to either 100 μg/Kg of dexamethasone sodium phosphate with normosaline or normosaline alone during late gestation (E10–E17). Nontreatment groups were also included. Brain samples were collected on embryonic day 17 and analyzed by mRNA microarray analysis. Results. Microarray analyses presented 332 significantly regulated genes. Overall, neurodevelopmental genes were overrepresented and a subset of 8 genes allowed treatment segregation through the hierarchical clustering method. The addition of stress or steroids greatly affected gene regulation through glucocorticoid receptor and stress signaling pathways. Furthermore, differences between dexamethasone-administered treatments implied a harmful effect during conditions of high stress. Microarray analysis was validated using qPCR. Conclusion. The effects of antenatal steroid therapy vary in fetuses according to maternal-fetal factors and environmental stimuli. Defining the key regulatory networks that signal either beneficial or damaging corticosteroid action would result in valuable adjustments to current treatment protocols.
Collapse
|
33
|
Abstract
The molecular mechanisms underlying skeletal muscle maintenance involve interplay between multiple signaling pathways. Under normal physiological conditions, a network of interconnected signals serves to control and coordinate hypertrophic and atrophic messages, culminating in a delicate balance between muscle protein synthesis and proteolysis. Loss of skeletal muscle mass, termed "atrophy", is a diagnostic feature of cachexia seen in settings of cancer, heart disease, chronic obstructive pulmonary disease, kidney disease, and burns. Cachexia increases the likelihood of death from these already serious diseases. Recent studies have further defined the pathways leading to gain and loss of skeletal muscle as well as the signaling events that induce differentiation and post-injury regeneration, which are also essential for the maintenance of skeletal muscle mass. In this review, we summarize and discuss the relevant recent literature demonstrating these previously undiscovered mediators governing anabolism and catabolism of skeletal muscle.
Collapse
Affiliation(s)
- Marc A Egerman
- Novartis Institutes for Biomedical Research , Cambridge, MA , USA
| | | |
Collapse
|
34
|
Kuo T, Harris CA, Wang JC. Metabolic functions of glucocorticoid receptor in skeletal muscle. Mol Cell Endocrinol 2013; 380:79-88. [PMID: 23523565 PMCID: PMC4893778 DOI: 10.1016/j.mce.2013.03.003] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 03/01/2013] [Accepted: 03/03/2013] [Indexed: 12/12/2022]
Abstract
Glucocorticoids (GCs) exert key metabolic influences on skeletal muscle. GCs increase protein degradation and decrease protein synthesis. The released amino acids are mobilized from skeletal muscle to liver, where they serve as substrates for hepatic gluconeogenesis. This metabolic response is critical for mammals' survival under stressful conditions, such as fasting and starvation. GCs suppress insulin-stimulated glucose uptake and utilization and glycogen synthesis, and play a permissive role for catecholamine-induced glycogenolysis, thus preserving the level of circulating glucose, the major energy source for the brain. However, chronic or excess exposure of GCs can induce muscle atrophy and insulin resistance. GCs convey their signal mainly through the intracellular glucocorticoid receptor (GR). While GR can act through different mechanisms, one of its major actions is to regulate the transcription of its primary target genes through genomic glucocorticoid response elements (GREs) by directly binding to DNA or tethering onto other DNA-binding transcription factors. These GR primary targets trigger physiological and pathological responses of GCs. Much progress has been made to understand how GCs regulate protein and glucose metabolism. In this review, we will discuss how GR primary target genes confer metabolic functions of GCs, and the mechanisms governing the transcriptional regulation of these targets. Comprehending these processes not only contributes to the fundamental understanding of mammalian physiology, but also will provide invaluable insight for improved GC therapeutics.
Collapse
Affiliation(s)
- Taiyi Kuo
- Department of Nutritional Science & Toxicology, University of California at Berkeley, Berkeley, CA 94720, United States
- Graduate Program of Endocrinology, University of California at Berkeley, Berkeley, CA 94720, United States
| | - Charles A. Harris
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, United States
- Department of Medicine, University of California at San Francisco, San Francisco, CA 94143, United States
| | - Jen-Chywan Wang
- Department of Nutritional Science & Toxicology, University of California at Berkeley, Berkeley, CA 94720, United States
- Graduate Program of Endocrinology, University of California at Berkeley, Berkeley, CA 94720, United States
- Corresponding author. Address: Department of Nutritional Science and Toxicology, 315 Morgan Hall, University of California at Berkeley, Berkeley, CA 94720-3104, United States. Tel.: +1 510 643 1039. (J.-C. Wang)
| |
Collapse
|
35
|
Owens J, Moreira K, Bain G. Characterization of primary human skeletal muscle cells from multiple commercial sources. In Vitro Cell Dev Biol Anim 2013; 49:695-705. [PMID: 23860742 PMCID: PMC3824271 DOI: 10.1007/s11626-013-9655-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 06/24/2013] [Indexed: 11/29/2022]
Abstract
There is a significant unmet need for safe, anabolic muscle therapies to treat diseases and conditions associated with severe muscle weakness and frailty. The identification of such therapies requires appropriate cell-based screening assays to select compounds for further development using animal models. Primary human skeletal muscle cells have recently become available from a number of commercial vendors. Such cells may be valuable for studying the mechanisms that direct muscle differentiation, and for identifying and characterizing novel therapeutic approaches for the treatment of age- and injury-induced muscle disorders. However, only limited characterization of these cells has been reported to date. Therefore, we have examined four primary human muscle cell preparations from three different vendors for their capacity to differentiate into multinucleated myotubes. Two of the preparations demonstrated robust myotube formation and expressed characteristic markers of muscle differentiation. Furthermore, these myotubes could be induced to undergo morphological atrophy- and hypertrophy-like responses, and atrophy could be blocked with an inhibitor of myostatin signaling, a pathway that is known to negatively regulate muscle mass. Finally, the myotubes were efficiently infected with recombinant adenovirus, providing a tool for genetic modification. Taken together, our results indicate that primary human muscle cells can be a useful system for studying muscle differentiation, and may also provide tools for studying new therapeutic molecules for the treatment of muscle disease.
Collapse
Affiliation(s)
- Jane Owens
- Tissue Repair Research Unit, Pfizer, 200 Cambridge Park Drive, Cambridge, MA, 02140, USA,
| | | | | |
Collapse
|
36
|
Glucocorticoid-induced skeletal muscle atrophy. Int J Biochem Cell Biol 2013; 45:2163-72. [PMID: 23806868 DOI: 10.1016/j.biocel.2013.05.036] [Citation(s) in RCA: 415] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Revised: 05/27/2013] [Accepted: 05/29/2013] [Indexed: 12/11/2022]
Abstract
Many pathological states characterized by muscle atrophy (e.g., sepsis, cachexia, starvation, metabolic acidosis and severe insulinopenia) are associated with an increase in circulating glucocorticoids (GC) levels, suggesting that GC could trigger the muscle atrophy observed in these conditions. GC-induced muscle atrophy is characterized by fast-twitch, glycolytic muscles atrophy illustrated by decreased fiber cross-sectional area and reduced myofibrillar protein content. GC-induced muscle atrophy results from increased protein breakdown and decreased protein synthesis. Increased muscle proteolysis, in particular through the activation of the ubiquitin proteasome and the lysosomal systems, is considered to play a major role in the catabolic action of GC. The stimulation by GC of these two proteolytic systems is mediated through the increased expression of several Atrogenes ("genes involved in atrophy"), such as FOXO, Atrogin-1, and MuRF-1. The inhibitory effect of GC on muscle protein synthesis is thought to result mainly from the inhibition of the mTOR/S6 kinase 1 pathway. These changes in muscle protein turnover could be explained by changes in the muscle production of two growth factors, namely Insulin-like Growth Factor (IGF)-I, a muscle anabolic growth factor and Myostatin, a muscle catabolic growth factor. This review will discuss the recent progress made in the understanding of the mechanisms involved in GC-induced muscle atrophy and consider the implications of these advancements in the development of new therapeutic approaches for treating GC-induced myopathy. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
|
37
|
Kukreti H, Amuthavalli K, Harikumar A, Sathiyamoorthy S, Feng PZ, Anantharaj R, Tan SLK, Lokireddy S, Bonala S, Sriram S, McFarlane C, Kambadur R, Sharma M. Muscle-specific microRNA1 (miR1) targets heat shock protein 70 (HSP70) during dexamethasone-mediated atrophy. J Biol Chem 2013; 288:6663-78. [PMID: 23297411 DOI: 10.1074/jbc.m112.390369] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
High doses of dexamethasone (Dex) or myostatin (Mstn) induce severe atrophy of skeletal muscle. Here we show a novel microRNA1 (miR1)-mediated mechanism through which Dex promotes skeletal muscle atrophy. Using both C2C12 myotubes and mouse models of Dex-induced atrophy we show that Dex induces miR1 expression through glucocorticoid receptor (GR). We further show that Mstn treatment facilitates GR nuclear translocation and thereby induces miR1 expression. Inhibition of miR1 in C2C12 myotubes attenuated the Dex-induced increase in atrophy-related proteins confirming a role for miR1 in atrophy. Analysis of miR1 targets revealed that HSP70 is regulated by miR1 during atrophy. Our results demonstrate that increased miR1 during atrophy reduced HSP70 levels, which resulted in decreased phosphorylation of AKT, as HSP70 binds to and protects phosphorylation of AKT. We further show that loss of pAKT leads to decreased phosphorylation, and thus, enhanced activation of FOXO3, up-regulation of MuRF1 and Atrogin-1, and progression of skeletal muscle atrophy. Based on these results, we propose a model whereby Dex- and Mstn-mediated atrophic signals are integrated through miR1, which then either directly or indirectly, inhibits the proteins involved in providing protection against atrophy.
Collapse
Affiliation(s)
- Himani Kukreti
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive MD7, Singapore 117597
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fanzani A, Conraads VM, Penna F, Martinet W. Molecular and cellular mechanisms of skeletal muscle atrophy: an update. J Cachexia Sarcopenia Muscle 2012; 3:163-79. [PMID: 22673968 PMCID: PMC3424188 DOI: 10.1007/s13539-012-0074-6] [Citation(s) in RCA: 234] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 05/13/2012] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle atrophy is defined as a decrease in muscle mass and it occurs when protein degradation exceeds protein synthesis. Potential triggers of muscle wasting are long-term immobilization, malnutrition, severe burns, aging as well as various serious and often chronic diseases, such as chronic heart failure, obstructive lung disease, renal failure, AIDS, sepsis, immune disorders, cancer, and dystrophies. Interestingly, a cooperation between several pathophysiological factors, including inappropriately adapted anabolic (e.g., growth hormone, insulin-like growth factor 1) and catabolic proteins (e.g., tumor necrosis factor alpha, myostatin), may tip the balance towards muscle-specific protein degradation through activation of the proteasomal and autophagic systems or the apoptotic pathway. Based on the current literature, we present an overview of the molecular and cellular mechanisms that contribute to muscle wasting. We also focus on the multifacetted therapeutic approach that is currently employed to prevent the development of muscle wasting and to counteract its progression. This approach includes adequate nutritional support, implementation of exercise training, and possible pharmacological compounds.
Collapse
Affiliation(s)
- Alessandro Fanzani
- Department of Biomedical Sciences and Biotechnologies and Interuniversitary Institute of Myology (IIM), University of Brescia, viale Europa 11, 25123, Brescia, Italy,
| | | | | | | |
Collapse
|
39
|
Genome-wide analysis of glucocorticoid receptor-binding sites in myotubes identifies gene networks modulating insulin signaling. Proc Natl Acad Sci U S A 2012; 109:11160-5. [PMID: 22733784 DOI: 10.1073/pnas.1111334109] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Glucocorticoids elicit a variety of biological responses in skeletal muscle, including inhibiting protein synthesis and insulin-stimulated glucose uptake and promoting proteolysis. Thus, excess or chronic glucocorticoid exposure leads to muscle atrophy and insulin resistance. Glucocorticoids propagate their signal mainly through glucocorticoid receptors (GR), which, upon binding to ligands, translocate to the nucleus and bind to genomic glucocorticoid response elements to regulate the transcription of nearby genes. Using a combination of chromatin immunoprecipitation sequencing and microarray analysis, we identified 173 genes in mouse C2C12 myotubes. The mouse genome contains GR-binding regions in or near these genes, and gene expression is regulated by glucocorticoids. Eight of these genes encode proteins known to regulate distinct signaling events in insulin/insulin-like growth factor 1 pathways. We found that overexpression of p85α, one of these eight genes, caused a decrease in C2C12 myotube diameters, mimicking the effect of glucocorticoids. Moreover, reducing p85α expression by RNA interference in C2C12 myotubes significantly compromised the ability of glucocorticoids to inhibit Akt and p70 S6 kinase activity and reduced glucocorticoid induction of insulin receptor substrate 1 phosphorylation at serine 307. This phosphorylation is associated with insulin resistance. Furthermore, decreasing p85α expression abolished glucocorticoid inhibition of protein synthesis and compromised glucocorticoid-induced reduction of cell diameters in C2C12 myotubes. Finally, a glucocorticoid response element was identified in the p85α GR-binding regions. In summary, our studies identified GR-regulated transcriptional networks in myotubes and showed that p85α plays a critical role in glucocorticoid-induced insulin resistance and muscle atrophy in C2C12 myotubes.
Collapse
|
40
|
Watson ML, Baehr LM, Reichardt HM, Tuckermann JP, Bodine SC, Furlow JD. A cell-autonomous role for the glucocorticoid receptor in skeletal muscle atrophy induced by systemic glucocorticoid exposure. Am J Physiol Endocrinol Metab 2012; 302:E1210-20. [PMID: 22354783 PMCID: PMC3361985 DOI: 10.1152/ajpendo.00512.2011] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Glucocorticoids (GCs) are important regulators of skeletal muscle mass, and prolonged exposure will induce significant muscle atrophy. To better understand the mechanism of skeletal muscle atrophy induced by elevated GC levels, we examined three different models: exogenous synthetic GC treatment [dexamethasone (DEX)], nutritional deprivation, and denervation. Specifically, we tested the direct contribution of the glucocorticoid receptor (GR) in skeletal muscle atrophy by creating a muscle-specific GR-knockout mouse line (MGR(e3)KO) using Cre-lox technology. In MGR(e3)KO mice, we found that the GR is essential for muscle atrophy in response to high-dose DEX treatment. In addition, DEX regulation of multiple genes, including two important atrophy markers, MuRF1 and MAFbx, is eliminated completely in the MGR(e3)KO mice. In a condition where endogenous GCs are elevated, such as nutritional deprivation, induction of MuRF1 and MAFbx was inhibited, but not completely blocked, in MGR(e3)KO mice. In response to sciatic nerve lesion and hindlimb muscle denervation, muscle atrophy and upregulation of MuRF1 and MAFbx occurred to the same extent in both wild-type and MGR(e3)KO mice, indicating that a functional GR is not required to induce atrophy under these conditions. Therefore, we demonstrate conclusively that the GR is an important mediator of skeletal muscle atrophy and associated gene expression in response to exogenous synthetic GCs in vivo and that the MGR(e3)KO mouse is a useful model for studying the role of the GR and its target genes in multiple skeletal muscle atrophy models.
Collapse
Affiliation(s)
- Monica L Watson
- Dept. of Neurobiology, Physiology, and Behavior, Univ. of California, One Shields Ave., Davis, CA 95616-8519, USA
| | | | | | | | | | | |
Collapse
|
41
|
Sato S, Suzuki H, Tsujimoto H, Shirato K, Tachiyashiki K, Imaizumi K. Casted-immobilization downregulates glucocorticoid receptor expression in rat slow-twitch soleus muscle. Life Sci 2011; 89:962-7. [DOI: 10.1016/j.lfs.2011.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 09/20/2011] [Accepted: 10/03/2011] [Indexed: 02/04/2023]
|
42
|
Muscle plasticity and β₂-adrenergic receptors: adaptive responses of β₂-adrenergic receptor expression to muscle hypertrophy and atrophy. J Biomed Biotechnol 2011; 2011:729598. [PMID: 22190857 PMCID: PMC3228688 DOI: 10.1155/2011/729598] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 08/23/2011] [Indexed: 02/04/2023] Open
Abstract
We discuss the functional roles of β2-adrenergic receptors in skeletal
muscle hypertrophy and atrophy as well as
the adaptive responses of β2-adrenergic receptor expression to anabolic and catabolic conditions. β2-Adrenergic receptor stimulation using
anabolic drugs increases muscle mass by promoting
muscle protein synthesis and/or attenuating
protein degradation. These effects are prevented
by the downregulation of the receptor. Endurance
training improves oxidative performance partly
by increasing β2-adrenergic receptor density in
exercise-recruited slow-twitch muscles. However,
excessive stimulation of β2-adrenergic receptors negates their beneficial effects. Although the preventive effects of β2-adrenergic receptor stimulation on
atrophy induced by muscle disuse and catabolic
hormones or drugs are observed, these catabolic
conditions decrease β2-adrenergic receptor expression in
slow-twitch muscles. These findings present
evidence against the use of β2-adrenergic agonists in therapy for muscle wasting and weakness. Thus, β2-adrenergic receptors in the skeletal
muscles play an important physiological role in
the regulation of protein and energy balance.
Collapse
|
43
|
The role and regulation of MAFbx/atrogin-1 and MuRF1 in skeletal muscle atrophy. Pflugers Arch 2011; 461:325-35. [DOI: 10.1007/s00424-010-0919-9] [Citation(s) in RCA: 203] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 12/21/2010] [Indexed: 10/18/2022]
|
44
|
Protection against dexamethasone-induced muscle atrophy is related to modulation by testosterone of FOXO1 and PGC-1α. Biochem Biophys Res Commun 2010; 403:473-8. [DOI: 10.1016/j.bbrc.2010.11.061] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 11/15/2010] [Indexed: 12/11/2022]
|
45
|
Kfir-Erenfeld S, Sionov RV, Spokoini R, Cohen O, Yefenof E. Protein kinase networks regulating glucocorticoid-induced apoptosis of hematopoietic cancer cells: fundamental aspects and practical considerations. Leuk Lymphoma 2010; 51:1968-2005. [PMID: 20849387 DOI: 10.3109/10428194.2010.506570] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids (GCs) are integral components in the treatment protocols of acute lymphoblastic leukemia, multiple myeloma, and non-Hodgkin lymphoma owing to their ability to induce apoptosis of these malignant cells. Resistance to GC therapy is associated with poor prognosis. Although they have been used in clinics for decades, the signal transduction pathways involved in GC-induced apoptosis have only partly been resolved. Accumulating evidence shows that this cell death process is mediated by a communication between nuclear GR affecting gene transcription of pro-apoptotic genes such as Bim, mitochondrial GR affecting the physiology of the mitochondria, and the protein kinase glycogen synthase kinase-3 (GSK3), which interacts with Bim following exposure to GCs. Prevention of Bim up-regulation, mitochondrial GR translocation, and/or GSK3 activation are common causes leading to GC therapy failure. Various protein kinases positively regulating the pro-survival Src-PI3K-Akt-mTOR and Raf-Ras-MEK-ERK signal cascades have been shown to be activated in malignant leukemic cells and antagonize GC-induced apoptosis by inhibiting GSK3 activation and Bim expression. Targeting these protein kinases has proven effective in sensitizing GR-positive malignant lymphoid cells to GC-induced apoptosis. Thus, intervening with the pro-survival kinase network in GC-resistant cells should be a good means of improving GC therapy of hematopoietic malignancies.
Collapse
Affiliation(s)
- Shlomit Kfir-Erenfeld
- The Lautenberg Center of Immunology and Cancer Research, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
46
|
Alamdari N, Smith IJ, Aversa Z, Hasselgren PO. Sepsis and glucocorticoids upregulate p300 and downregulate HDAC6 expression and activity in skeletal muscle. Am J Physiol Regul Integr Comp Physiol 2010; 299:R509-20. [PMID: 20538901 DOI: 10.1152/ajpregu.00858.2009] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Muscle wasting during sepsis is in part regulated by glucocorticoids. In recent studies, treatment of cultured muscle cells in vitro with dexamethasone upregulated expression and activity of p300, a histone acetyl transferase (HAT), and reduced expression and activity of the histone deacetylases-3 (HDAC3) and -6, changes that favor hyperacetylation. Here, we tested the hypothesis that sepsis and glucocorticoids regulate p300 and HDAC3 and -6 in skeletal muscle in vivo. Because sepsis-induced metabolic changes are particularly pronounced in white, fast-twitch skeletal muscle, most experiments were performed in extensor digitorum longus muscles. Sepsis in rats upregulated p300 mRNA and protein levels, stimulated HAT activity, and reduced HDAC6 expression and HDAC activity. The sepsis-induced changes in p300 and HDAC expression were prevented by the glucocorticoid receptor antagonist RU38486. Treatment of rats with dexamethasone increased expression of p300 and HAT activity, reduced expression of HDAC3 and -6, and inhibited HDAC activity. Finally, treatment with the HDAC inhibitor trichostatin A resulted in increased muscle proteolysis and expression of the ubiquitin ligase atrogin-1. Taken together, our results suggest for the first time that sepsis-induced muscle wasting may be regulated by glucocorticoid-dependent hyperacetylation caused by increased p300 and reduced HDAC expression and activity. The recent development of pharmacological HDAC activators may provide a novel avenue to prevent and treat muscle wasting in sepsis and other catabolic conditions.
Collapse
Affiliation(s)
- Nima Alamdari
- Dept. of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | |
Collapse
|
47
|
Frijters R, Fleuren W, Toonen EJM, Tuckermann JP, Reichardt HM, van der Maaden H, van Elsas A, van Lierop MJ, Dokter W, de Vlieg J, Alkema W. Prednisolone-induced differential gene expression in mouse liver carrying wild type or a dimerization-defective glucocorticoid receptor. BMC Genomics 2010; 11:359. [PMID: 20525385 PMCID: PMC2895630 DOI: 10.1186/1471-2164-11-359] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 06/05/2010] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Glucocorticoids (GCs) control expression of a large number of genes via binding to the GC receptor (GR). Transcription may be regulated either by binding of the GR dimer to DNA regulatory elements or by protein-protein interactions of GR monomers with other transcription factors. Although the type of regulation for a number of individual target genes is known, the relative contribution of both mechanisms to the regulation of the entire transcriptional program remains elusive. To study the importance of GR dimerization in the regulation of gene expression, we performed gene expression profiling of livers of prednisolone-treated wild type (WT) and mice that have lost the ability to form GR dimers (GRdim). RESULTS The GR target genes identified in WT mice were predominantly related to glucose metabolism, the cell cycle, apoptosis and inflammation. In GRdim mice, the level of prednisolone-induced gene expression was significantly reduced compared to WT, but not completely absent. Interestingly, for a set of genes, involved in cell cycle and apoptosis processes and strongly related to Foxo3a and p53, induction by prednisolone was completely abolished in GRdim mice. In contrast, glucose metabolism-related genes were still modestly upregulated in GRdim mice upon prednisolone treatment. Finally, we identified several novel GC-inducible genes from which Fam107a, a putative histone acetyltransferase complex interacting protein, was most strongly dependent on GR dimerization. CONCLUSIONS This study on prednisolone-induced effects in livers of WT and GRdim mice identified a number of interesting candidate genes and pathways regulated by GR dimers and sheds new light onto the complex transcriptional regulation of liver function by GCs.
Collapse
Affiliation(s)
- Raoul Frijters
- Computational Drug Discovery (CDD), Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, the Netherlands
| | - Wilco Fleuren
- Computational Drug Discovery (CDD), Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, the Netherlands
| | - Erik JM Toonen
- Department of Immunotherapeutics, Schering-Plough, Molenstraat 110, 5342 CC Oss, the Netherlands
| | - Jan P Tuckermann
- Tuckermann Lab, Leibniz Institute for Age Research - Fritz Lipmann Institute, Beutenbergstraße 11, 07745 Jena, Germany
| | - Holger M Reichardt
- Department of Cellular and Molecular Immunology, University of Göttingen Medical School, Humboldtallee 34, 37073 Göttingen, Germany
| | - Hans van der Maaden
- Molecular Pharmacology Department, Schering-Plough, Molenstraat 110, 5342 CC Oss, the Netherlands
| | - Andrea van Elsas
- Department of Immunotherapeutics, Schering-Plough, Molenstraat 110, 5342 CC Oss, the Netherlands
| | - Marie-Jose van Lierop
- Department of Immunotherapeutics, Schering-Plough, Molenstraat 110, 5342 CC Oss, the Netherlands
| | - Wim Dokter
- Department of Immunotherapeutics, Schering-Plough, Molenstraat 110, 5342 CC Oss, the Netherlands
| | - Jacob de Vlieg
- Computational Drug Discovery (CDD), Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen Medical Centre, Geert Grooteplein Zuid 26-28, 6525 GA Nijmegen, the Netherlands
- Department of Molecular Design & Informatics, Schering-Plough, Molenstraat 110, 5342 CC Oss, the Netherlands
| | - Wynand Alkema
- Department of Molecular Design & Informatics, Schering-Plough, Molenstraat 110, 5342 CC Oss, the Netherlands
| |
Collapse
|
48
|
Smith IJ, Alamdari N, O'Neal P, Gonnella P, Aversa Z, Hasselgren PO. Sepsis increases the expression and activity of the transcription factor Forkhead Box O 1 (FOXO1) in skeletal muscle by a glucocorticoid-dependent mechanism. Int J Biochem Cell Biol 2010; 42:701-11. [PMID: 20079455 DOI: 10.1016/j.biocel.2010.01.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 12/18/2009] [Accepted: 01/06/2010] [Indexed: 12/18/2022]
Abstract
Sepsis-induced muscle wasting has severe clinical consequences, including muscle weakness, need for prolonged ventilatory support and stay in the intensive care unit, and delayed ambulation with risk for pulmonary and thromboembolic complications. Understanding molecular mechanisms regulating loss of muscle mass in septic patients therefore has significant clinical implications. Forkhead Box O (FOXO) transcription factors have been implicated in muscle wasting, partly reflecting upregulation of the ubiquitin ligases atrogin-1 and MuRF1. The influence of sepsis on FOXO transcription factors in skeletal muscle is poorly understood. We tested the hypothesis that sepsis upregulates expression and activity of FOXO transcription factors in skeletal muscle by a glucocorticoid-dependent mechanism. Sepsis in rats increased muscle FOXO1 and 3a mRNA and protein levels but did not influence FOXO4 expression. Nuclear FOXO1 levels and DNA binding activity were increased in septic muscle whereas FOXO3a nuclear levels were not increased during sepsis. Sepsis-induced expression of FOXO1 was reduced by the glucocorticoid receptor antagonist RU38486 and treatment of rats with dexamethasone increased FOXO1 mRNA levels suggesting that the expression of FOXO1 is regulated by glucocorticoids. Reducing FOXO1, but not FOXO3a, expression by siRNA in cultured L6 myotubes inhibited dexamethasone-induced atrogin-1 and MuRF1 expression, further supporting a role of FOXO1 in glucocorticoid-regulated muscle wasting. Results suggest that sepsis increases FOXO1 expression and activity in skeletal muscle by a glucocorticoid-dependent mechanism and that glucocorticoid-dependent upregulation of atrogin-1 and MuRF1 in skeletal muscle is regulated by FOXO1. The study is significant because it provides novel information about molecular mechanisms involved in sepsis-induced muscle wasting.
Collapse
Affiliation(s)
- Ira J Smith
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue ST919, Boston, MA, United States
| | | | | | | | | | | |
Collapse
|
49
|
Cho JE, Fournier M, Da X, Lewis MI. Time course expression of Foxo transcription factors in skeletal muscle following corticosteroid administration. J Appl Physiol (1985) 2009; 108:137-45. [PMID: 19850732 DOI: 10.1152/japplphysiol.00704.2009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Increased expression of forkhead box O (Foxo) transcription factors were reported in cultured myotubes and mouse limb muscle with corticosteroid (CS) treatment. We previously reported that administration of CS to rats resulted in muscle fiber atrophy only by day 7. The aim of this study, therefore, was to evaluate the time-course changes in the expression of Foxo transcription factors and muscle-specific ubiquitin E3 ligases in rat limb muscle following CS administration. Triamcinolone (TRI; 1 mg x kg(-1) x day(-1) im) was administered for 1, 3, or 7 days. Control (CTL) rats were given saline. Muscle mRNA was analyzed by real-time RT-PCR. Compared with CTL, body weights of TRI-treated animals decreased by 3, 12, and 21% at days 1, 3, and 7, respectively. Muscle IGF-1 mRNA levels decreased by 33, 65, and 58% at days 1, 3, and 7 in TRI-treated rats compared with CTL. Levels of phosphorylated Akt were 28, 50, and 36% lower in TRI animals at these time points. Foxo1 mRNA increased progressively by 1.2-, 1.4-, and 2.5-fold at days 1, 3, and 7 in TRI animals. Similar changes were noted in the expression of Foxo3a mRNA (1.3-, 1.4-, and 2.6-fold increments). By contrast, Foxo4 mRNA was not significantly changed in TRI animals. With TRI, muscle atrophy F box/Atrogin-1 increased by 1.8-, 4.1-, and 7.5-fold at days 1, 3, and 7 compared with CTL rats. By contrast, muscle RING finger 1 increased only from day 7 (2.7-fold). Gradual reduction in IGF-I expression with TRI over the time series paralleled that of Akt. These findings are consistent with a progressive stimulus to muscle protein degradation and the need to process/remove disassembled muscle proteins via the ubiquitin-proteasome system. Elucidating the dynamic catabolic responses to CS challenge is important in understanding the mechanisms underlying muscle atrophy and therapeutic measures to offset this.
Collapse
Affiliation(s)
- John E Cho
- Division of Pulmonary/Critical Care Medicine, The Burns and Allen Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | |
Collapse
|