1
|
Kim A, Kim J, Kim BY, Seo CS, Kim YR, Song KH, Kim NS. Aquo-ethanolic extract of Lilii Bulbus attenuates dexamethasone-induced muscle loss and enhances muscle strength in experimental mice. Biomed Pharmacother 2024; 181:117658. [PMID: 39486367 DOI: 10.1016/j.biopha.2024.117658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/12/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Traditionally, Lilium lancifolium bulb is known for its ability to nourish yin, nourish the lungs, clear the heart, soothe coughs, reduce irritability, and calm the mind. In Oriental Medicine, it is categorized as a tonic remedy for alleviating symptoms of fatigue and enhancing the strength of bones and muscles. In this study, we aimed to validate the effectiveness of the aquo-ethanolic extract of Lilli Bulbus (LBE) in a dexamethasone (DEX)-induced muscle atrophy model, both in vitro and in vivo, and elucidate its mechanism of action through muscle transcriptome analysis. The effects of LBE on the viability and myotube density of C2C12 myoblasts and differentiated C2C12 myotubes with and without DEX treatment were investigated. LBE pretreatment protected C2C12 myoblast cells and increased the muscle density of C2C12 myotubes in response to DEX. LBE showed potent free radical scavenging activities in cell-free biochemical assays as well as antioxidant activity in C2C12 myoblasts. LBE also exhibited protective effects in an experimental animal model of DEX-induced muscle atrophy, showing muscular function and motor coordination recovery. Transcriptomic analysis of three different muscle tissues from mice with DEX-induced muscle atrophy showed that the regulation of the extracellular matrix was perturbed by glucocorticoid treatment, and this perturbation was reversed by LBE treatment. Collectively, LBE alleviated skeletal muscle loss and maintained muscle function from the chronic toxicity of DEX by protecting muscle cells from various stressful conditions, as well as DEX itself, inhibiting muscle protein degradation, and preserving the muscle tissue microenvironment.
Collapse
Affiliation(s)
- Aeyung Kim
- KM Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea.
| | - Jinhee Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Bu-Yeo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Chang-Seob Seo
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Yu Ri Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Kwang Hoon Song
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - No Soo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| |
Collapse
|
2
|
Li D, Rongchun W, Lu W, Ma Y. Exploring the potential of MFG-E8 in neurodegenerative diseases. Crit Rev Food Sci Nutr 2024:1-15. [PMID: 39468823 DOI: 10.1080/10408398.2024.2417800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Milk fat globule-epidermal growth factor 8 (MFG-E8) is a multifunctional glycoprotein regulating intercellular interactions in various biological and pathological processes. This review summarizes the effects and mechanisms of MFG-E8 in neurodegenerative diseases (NDDs), emphasizing its roles in inflammation, apoptosis, and oxidative stress. In this review, will also explore the potential of MFG-E8 as a diagnostic biomarker and its therapeutic applications in neurodegenerative disorders. Recent studies have revealed intriguing characteristics of using MFG-E8 as a potential drug for treating various brain disorders. While the discovery, origin, expression, and physiological functions of MFG-E8 in various organs and tissues are well defined, its role in the brain remains less understood. This is particularly true for NDDs, indicating unmet medical needs. Elucidating its role in the brain could position MFG-E8 as a potential treatment for NDDs.
Collapse
Affiliation(s)
- Dan Li
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Wang Rongchun
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Weihong Lu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
3
|
Ye YL, Kuai Z, Qian DD, He YT, Shen JP, Wu KF, Ren WY, Hu Y. GLP-2 ameliorates D-galactose induced muscle aging by IGF-1/Pi3k/Akt/FoxO3a signaling pathway in C2C12 cells and mice. Arch Gerontol Geriatr 2024; 124:105462. [PMID: 38692155 DOI: 10.1016/j.archger.2024.105462] [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: 01/16/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND The study aimed to investigate the effect of Glucagon-like peptide-2 (GLP-2) on muscle aging in vivo and in vitro. METHODS Six-week-old C57BL/6J mice were administered with D-galactose (200 mg/kg/day, intraperitoneally) for 8weeks, followed by daily subcutaneous injections of GLP-2 (300 or 600 μg/kg/day) for 4weeks. Skeletal muscle function and mass were evaluated using relative grip strength and muscle weight. The sizes and types of muscle fibers and apoptosis were assessed through histological analysis, immunofluorescence staining, and TUNEL staining, respectively. C2C12 myotubes were treated with D-galactose (40 mg/mL) and GLP-2. Protein expression of differentiation-related myogenic differentiation factor D (MyoD), myogenin (MyoG), and myosin heavy chain (Myhc), degradation-related Muscle RING finger 1 (MuRF-1), and muscle atrophy F-box (MAFbx)/Atrogin-1, and apoptosis-related B-cell leukemia/lymphoma 2 (Bcl-2) and Bax, were assessed using western blots. The Pi3k inhibitor LY294002 was applied to investigate whether GLP-2 regulated myogenesis and myotube aging via IGF-1/Pi3k/Akt/FoxO3a signaling pathway. RESULTS The results demonstrated that GLP-2 significantly reversed the decline in muscles weight, relative grip strength, diameter, and cross-sectional area of muscle fibers induced by D-galactose in mice. Apart from suppressing the expressions of MuRF-1 and Atrogin-1 in the muscles and C2C12 myotubes, GLP-2 significantly increased the expressions of MyoD, MyoG, and Myhc compared to the D-galactose. GLP-2 significantly suppressed cell apoptosis. Western blot analysis indicated that the regulation of GLP-2 may be attributed to the activation of theIGF-1/Pi3k/Akt/FoxO3a phosphorylation pathway. CONCLUSIONS This study suggested that GLP-2 ameliorated D-galactose induced muscle aging by IGF-1/Pi3k/Akt/FoxO3a pathway.
Collapse
Affiliation(s)
- Yang-Li Ye
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, PR China
| | - Zheng Kuai
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, PR China
| | - Dian-Dian Qian
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, PR China
| | - Yu-Ting He
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, PR China
| | - Ji-Ping Shen
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, PR China
| | - Ke-Fen Wu
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, PR China
| | - Wei-Ying Ren
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, PR China.
| | - Yu Hu
- Department of Geriatrics, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, PR China; Center for Evidence Based Medicine and Clinical Epidemiology, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| |
Collapse
|
4
|
Hwangbo H, Kim MY, Ji SY, Kim DH, Park BS, Jeong SU, Yoon JH, Kim TH, Kim GY, Choi YH. A Mixture of Morus alba and Angelica keiskei Leaf Extracts Improves Muscle Atrophy by Activating the PI3K/Akt/mTOR Signaling Pathway and Inhibiting FoxO3a In Vitro and In Vivo. J Microbiol Biotechnol 2023; 33:1635-1647. [PMID: 37674382 PMCID: PMC10772550 DOI: 10.4014/jmb.2306.06012] [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: 06/07/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 09/08/2023]
Abstract
Muscle atrophy, which is defined as a decrease in muscle mass and strength, is caused by an imbalance between the anabolism and catabolism of muscle proteins. Thus, modulating the homeostasis between muscle protein synthesis and degradation represents an efficient treatment approach for this condition. In the present study, the protective effects against muscle atrophy of ethanol extracts of Morus alba L. (MA) and Angelica keiskei Koidz. (AK) leaves and their mixtures (MIX) were evaluated in vitro and in vivo. Our results showed that MIX increased 5-aminoimidazole-4-carboxamide ribonucleotide-induced C2C12 myotube thinning, and enhanced soleus and gastrocnemius muscle thickness compared to each extract alone in dexamethasone-induced muscle atrophy Sprague Dawley rats. In addition, although MA and AK substantially improved grip strength and histological changes for dexamethasone-induced muscle atrophy in vivo, the efficacy was superior in the MIX-treated group. Moreover, MIX further increased the expression levels of myogenic factors (MyoD and myogenin) and decreased the expression levels of E3 ubiquitin ligases (atrogin-1 and muscle-specific RING finger protein-1) in vitro and in vivo compared to the MA- and AK-alone treatment groups. Furthermore, MIX increased the levels of phosphorylated phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), and mammalian target of rapamycin (mTOR) that were reduced by dexamethasone, and downregulated the expression of forkhead box O3 (FoxO3a) induced by dexamethasone. These results suggest that MIX has a protective effect against muscle atrophy by enhancing muscle protein anabolism through the activation of the PI3K/Akt/mTOR signaling pathway and attenuating catabolism through the inhibition of FoxO3a.
Collapse
Affiliation(s)
- Hyun Hwangbo
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
| | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
| | - Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
| | - Da Hye Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
| | - Beom Su Park
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
| | - Seong Un Jeong
- Hamsoa Pharmaceutical Co., Ltd., Iksan 54524, Republic of Korea
| | - Jae Hyun Yoon
- Hamsoa Pharmaceutical Co., Ltd., Iksan 54524, Republic of Korea
| | - Tae Hee Kim
- Hamsoa Pharmaceutical Co., Ltd., Iksan 54524, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| |
Collapse
|
5
|
Wang J, Ma S, Wu Q, Xu Q, Wang J, Zhang R, Bai L, Li L, Liu H. Effects of testis testosterone deficiency on gene expression in the adrenal gland and skeletal muscle of ducks. Br Poult Sci 2023. [PMID: 36735924 DOI: 10.1080/00071668.2023.2176741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
1. Testosterone has an anabolic effect on skeletal muscle. The testes produce most of the testosterone in vivo, while the adrenal glands contribute smaller amounts. When testis testosterone is deficient the adrenal gland increases steroid hormone synthesis, which is referred to as compensatory testicular adaptation (CTA).2. To reveal the effects of testis testosterone deficiency on adrenal steroid hormones synthesis and skeletal muscle development, gene expression related to adrenal steroid hormones synthesis and skeletal muscle development were determined by RNA-seq.3. The results showed that castrating male ducks had significant effects on their body weight but no significant impact on cross-sectional area (CSA) or density of pectoral muscle fibres. In skeletal muscle protein metabolism, expression levels of the catabolic gene atrogin1/MAFbx and the anabolic gene eEF2 were significantly higher, with concomitant increases after castration. The adrenal glands' alteration of the steroid hormone 11β-hydroxylase (CYP11B1) was significantly lower following castration.4. Expression pattern analysis showed that the adrenal glands' glucocorticoid receptor (NR3C1/GR) had a potential regulatory relationship with the skeletal muscle-related genes (Pax7, mTOR, FBXO32, FOXO3, and FOXO4).5. The data showed that castration affected muscle protein metabolism, adrenal steroid and testosterone synthesis. In addition, it was speculated that, after castration, steroid hormones produced by the adrenal gland could have a compensatory effect, which might mediate the changes in skeletal muscle protein metabolism and development.
Collapse
Affiliation(s)
- J Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, P.R. China
| | - S Ma
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, P.R. China
| | - Q Wu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, P.R. China
| | - Q Xu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, P.R. China
| | - J Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, P.R. China
| | - R Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, P.R. China
| | - L Bai
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, P.R. China
| | - L Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, P.R. China
| | - H Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, P.R. China
| |
Collapse
|
6
|
Zhu DY, Lu J, Xu R, Yang JZ, Meng XR, Ou-Yang XN, Yan QY, Nie RF, Zhao T, Chen YD, Lu Y, Zhang YN, Li WJ, Shen X. FX5, a non-steroidal glucocorticoid receptor antagonist, ameliorates diabetic cognitive impairment in mice. Acta Pharmacol Sin 2022; 43:2495-2510. [PMID: 35260821 PMCID: PMC9525278 DOI: 10.1038/s41401-022-00884-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/06/2022] [Indexed: 12/15/2022] Open
Abstract
Diabetic cognitive impairment (DCI) is a common diabetic complication characterized by learning and memory deficits. In diabetic patients, hyperactivated hypothalamic-pituitary-adrenal (HPA) axis leads to abnormal increase of glucocorticoids (GCs), which causes the damage of hippocampal neurons and cognitive impairment. In this study we investigated the cognition-improving effects of a non-steroidal glucocorticoid receptor (GR) antagonist 5-chloro-N-[4-chloro-3-(trifluoromethyl) phenyl]thiophene-2-sulfonamide (FX5) in diabetic mice. Four weeks after T1DM or T2DM was induced, the mice were administered FX5 (20, 40 mg·kg-1·d-1, i.g.) for 8 weeks. Cognitive impairment was assessed in open field test, novel object recognition test, Y-maze test, and Morris water maze test. We showed that FX5 administration significantly ameliorated the cognitive impairments in both type 1 and 2 diabetic mice. Similar cognitive improvement was observed in diabetic mice following brain GR-specific knockdown by injecting AAV-si-GR. Moreover, AAV-si-GR injection occluded the cognition-improving effects of FX5, suggesting that FX5 functioning as a non-steroidal GR antagonist. In PA-treated primary neurons (as DCI model in vitro), we demonstrated that FX5 (2, 5, 10 μM) dose-dependently ameliorated synaptic impairment via upregulating GR/BDNF/TrkB/CREB pathway, protected against neuronal apoptosis through repressing GR/PI3K/AKT/GSK3β-mediated tauopathy and subsequent endoplasmic reticulum stress. In LPS-treated primary microglia, FX5 dose-dependently inhibited inflammation through GR/NF-κB/NLRP3/ASC/Caspase-1 pathway. These beneficial effects were also observed in the hippocampus of diabetic mice following FX5 administration. Collectively, we have elucidated the mechanisms underlying the beneficial effects of non-steroidal GR antagonist FX5 on DCI and highlighted the potential of FX5 in the treatment of the disease.
Collapse
Affiliation(s)
- Dan-Yang Zhu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jian Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Juan-Zhen Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiang-Rui Meng
- Faculty of Art and Science, Queens University, Kingston, ON, K7L 3N6, Canada
| | - Xing-Nan Ou-Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiu-Ying Yan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui-Fang Nie
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tong Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi-di Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi-Nan Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Wen-Jun Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
7
|
Ma K, Huang F, Qiao R, Miao L. Pathogenesis of sarcopenia in chronic obstructive pulmonary disease. Front Physiol 2022; 13:850964. [PMID: 35928562 PMCID: PMC9343800 DOI: 10.3389/fphys.2022.850964] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common pulmonary disease characterized by persistent respiratory symptoms and airflow obstruction. In addition to lung diseases, chronic obstructive pulmonary disease (COPD) is often associated with other organ diseases, and sarcopenia is one of the common diseases. In recent years, multiple factors have been proposed to influence muscle dysfunction in COPD patients, including systemic and local inflammation, oxidative stress, hypoxia, hypercapnia, protein synthesis, catabolic imbalance, nutritional changes, disuse, ageing, and the use of medications such as steroids. These factors alone or in combination can lead to a reduction in muscle mass and cross-sectional area, deterioration of muscle bioenergy metabolism, defects in muscle repair and regeneration mechanisms, apoptosis and other anatomical and/or functional pathological changes, resulting in a decrease in the muscle’s ability to work. This article reviews the research progress of possible pathogenesis of sarcopenia in COPD.
Collapse
|
8
|
Oh S, Choi CH, Lee BJ, Park JH, Son KH, Byun K. Fermented Oyster Extract Attenuated Dexamethasone-Induced Muscle Atrophy by Decreasing Oxidative Stress. Molecules 2021; 26:molecules26237128. [PMID: 34885708 PMCID: PMC8658907 DOI: 10.3390/molecules26237128] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/12/2021] [Accepted: 11/22/2021] [Indexed: 12/25/2022] Open
Abstract
It is well known that oxidative stress induces muscle atrophy, which decreases with the activation of Nrf2/HO-1. Fermented oyster extracts (FO), rich in γ-aminobutyric acid (GABA) and lactate, have shown antioxidative effects. We evaluated whether FO decreased oxidative stress by upregulating Nrf2/HO-1 and whether it decreased NF-κB, leading to decreased IL-6 and TNF-α. Decreased oxidative stress led to the downregulation of Cbl-b ubiquitin ligase, which increased IGF-1 and decreased FoxO3, atrogin1, and Murf1, and eventually decreased muscle atrophy in dexamethasone (Dexa)-induced muscle atrophy animal model. For four weeks, mice were orally administered with FO, GABA, lactate, or GABA+Lactate, and then Dexa was subcutaneously injected for ten days. During Dexa injection period, FO, GABA, lactate, or GABA+Lactate were also administered, and grip strength test and muscle harvesting were performed on the day of the last Dexa injection. We compared the attenuation effect of FO with GABA, lactate, and GABA+lactate treatment. Nrf2 and HO-1 expressions were increased by Dexa but decreased by FO; SOD activity and glutathione levels were decreased by Dexa but increased by FO; NADPH oxidase activity was increased by Dexa but decreased by FO; NF-κB, IL-6, and TNF-α activities were increased by Dexa were decreased by FO; Cbl-b expression was increased by Dexa but restored by FO; IGF-1 expression was decreased by Dexa but increased by FO; FoxO3, Atrogin-1, and MuRF1 expressions were increased by Dexa but decreased by FO. The gastrocnemius thickness and weight were decreased by Dexa but increased by FO. The cross-sectional area of muscle fiber and grip strength were decreased by Dexa but increased by FO. In conclusion, FO decreased Dexa-induced oxidative stress through the upregulation of Nrf2/HO-1. Decreased oxidative stress led to decreased Cbl-b, FoxO3, atrogin1, and MuRF1, which attenuated muscle atrophy.
Collapse
Affiliation(s)
- Seyeon Oh
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School and Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea;
| | - Chang Hu Choi
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Korea;
| | - Bae-Jin Lee
- Marine Bioprocess Co., Ltd., Smart Marine BioCenter, Busan 46048, Korea; (B.-J.L.); (J.-H.P.)
| | - Joung-Hyun Park
- Marine Bioprocess Co., Ltd., Smart Marine BioCenter, Busan 46048, Korea; (B.-J.L.); (J.-H.P.)
| | - Kuk-Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, Gachon University, Incheon 21565, Korea;
- Correspondence: (K.-H.S.); (K.B.); Tel.: +82-32-460-3666 (K.-H.S.); +82-32-899-6511 (K.B.)
| | - Kyunghee Byun
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School and Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon 21999, Korea;
- Department of Anatomy and Cell Biology, Gachon University College of Medicine, Incheon 21936, Korea
- Correspondence: (K.-H.S.); (K.B.); Tel.: +82-32-460-3666 (K.-H.S.); +82-32-899-6511 (K.B.)
| |
Collapse
|
9
|
Seok YM, Yoo JM, Nam Y, Kim J, Kim JS, Son JH, Kim HJ. Mountain ginseng inhibits skeletal muscle atrophy by decreasing muscle RING finger protein-1 and atrogin1 through forkhead box O3 in L6 myotubes. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113557. [PMID: 33161026 DOI: 10.1016/j.jep.2020.113557] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 09/29/2020] [Accepted: 11/01/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Mountain ginseng (Panax ginseng C.A. Meyer) is a medicinal herb with immune effects, muscle damage protection and energy metabolism effects. However, the pharmacological role of mountain ginseng in dexamethasone (DEXA)-induced muscle atrophy through the forkhead box O (FOXO) family is not understood. Therefore, we hypothesized that mountain ginseng inhibits skeletal muscle atrophy by decreasing muscle RING finger protein-1 (MuRF1) and atrogin1 through FOXO3 in L6 myotubes. METHODS Rat myoblast (L6) cells or Sprague-Dawley (SD) rats were exposed to DEXA and mountain ginseng. The expressions of muscle atrophy targets such as MuRF1, atrogin1, MyHC (myosin heavy chain), HSP90, p-Akt, Akt, p-ERK1/2, ERK, FOXO3a, FOXO1, myostatin, and follistatin were analyzed by using Western blot analysis or real-time PCR. The diameter of myotubes was measured. Recruitment of glucocorticoid receptor (GR) or FOXO3a was analyzed by performing a chromatin immunoprecipitation (ChIP) assay. RESULTS Mountain ginseng treatment reduced muscle weight loss and collagen deposition in DEXA-induced rats. Mountain ginseng treatment led to decreases in MuRF1, atrogin1, p-ERK1/2, FOXO3a, FOXO1, and myostatin. Also, mountain ginseng treatment led to increases in the diameter of myotubes, MyHC, HSP90, p-Akt, and follistatin. Treatment with mountain ginseng reduced enrichment of GR, FOXO3a, and RNA polymerase II on the promoters. CONCLUSIONS These results suggest that mountain ginseng inhibits skeletal muscle atrophy by decreasing MuRF1 and atrogin1 through FOXO3a in L6 myotubes.
Collapse
Affiliation(s)
- Young Mi Seok
- Korean Medicine R&D Team 1, National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea.
| | - Jae-Myung Yoo
- Korean Medicine R&D Team 1, National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea.
| | - Yoonju Nam
- Korean Medicine R&D Team 1, National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea.
| | - Jungeun Kim
- Korean Medicine R&D Team 1, National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea.
| | - Jin Soo Kim
- Korean Medicine R&D Team 1, National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea.
| | - Jun-Ho Son
- Korean Medicine R&D Team 1, National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea.
| | - Hyo Jung Kim
- Korean Medicine R&D Team 1, National Institute for Korean Medicine Development, Gyeongsan, 38540, Republic of Korea.
| |
Collapse
|
10
|
Bedada FB, Ntekim OE, Nwulia EO, Fungwe TV, Nadarajah SR, Obisesan TO. Exercise Training-Increased FBXO32 and FOXO1 in a Gender-Dependent Manner in Mild Cognitively Impaired African Americans: GEMS-1 Study. Front Aging Neurosci 2021; 13:641758. [PMID: 33935685 PMCID: PMC8079639 DOI: 10.3389/fnagi.2021.641758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/18/2021] [Indexed: 01/29/2023] Open
Abstract
The ubiquitin proteasome system (UPS) and FOXOs transcription factors play a pivotal role in cellular clearance and minimizing the accumulation of Aβ in neurodegeneration (ND). In African Americans (AAs) with mild cognitive impairment (MCI), the role of components of UPS and FOXOs; and whether they are amenable to exercise effects is unknown. We hypothesized that exercise can enhance cellular clearance systems during aging and ND by increasing expressions of FBXO32 and FOXO1. To test this hypothesis, we used TaqMan gene expression analysis in peripheral blood (PB) to investigate the component of UPS and FOXOs; and provide mechanistic insight at baseline, during exercise, and in both genders. At baseline, levels of FBXO32 were higher in women than in men. In our attempt to discern gender-specific exercise-related changes, we observed that levels of FBXO32 increased in men but not in women. Similarly, levels of FOXO1 increased in men only. These data suggest that a graded dose of FBXO32 and FOXO1 may be beneficial when PB cells carrying FBXO32 and FOXO1 summon into the brain in response to Alzheimer's disease (AD) perturbation (docking station PB cells). Our observation is consistent with emerging studies that exercise allows the trafficking of blood factors. Given the significance of FBXO32 and FOXO1 to ND and associated muscle integrity, our findings may explain, at least in part, the benefits of exercise on memory, associated gait, and balance perturbation acknowledged to herald the emergence of MCI.
Collapse
Affiliation(s)
- Fikru B. Bedada
- Department of Clinical Laboratory Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Oyonumo E. Ntekim
- Department of Nutritional Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Evaristus O. Nwulia
- Department of Psychiatry, Howard University Hospital, Washington, DC, United States
| | - Thomas V. Fungwe
- Department of Nutritional Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Sheeba Raaj Nadarajah
- Department of Nursing, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, United States
| | - Thomas O. Obisesan
- Division of Geriatrics, Department of Medicine, Howard University Hospital, Washington, DC, United States
| |
Collapse
|
11
|
Ulla A, Uchida T, Miki Y, Sugiura K, Higashitani A, Kobayashi T, Ohno A, Nakao R, Hirasaka K, Sakakibara I, Nikawa T. Morin attenuates dexamethasone-mediated oxidative stress and atrophy in mouse C2C12 skeletal myotubes. Arch Biochem Biophys 2021; 704:108873. [PMID: 33848514 DOI: 10.1016/j.abb.2021.108873] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/19/2021] [Accepted: 04/06/2021] [Indexed: 12/28/2022]
Abstract
Glucocorticoids are the drugs most commonly used to manage inflammatory diseases. However, they are prone to inducing muscle atrophy by increasing muscle proteolysis and decreasing protein synthesis. Various studies have demonstrated that antioxidants can mitigate glucocorticoid-induced skeletal muscle atrophy. Here, we investigated the effect of a potent antioxidative natural flavonoid, morin, on the muscle atrophy and oxidative stress induced by dexamethasone (Dex) using mouse C2C12 skeletal myotubes. Dex (10 μM) enhanced the production of reactive oxygen species (ROS) in C2C12 myotubes via glucocorticoid receptor. Moreover, Dex administration reduced the diameter and expression levels of the myosin heavy chain protein in C2C12 myotubes, together with the upregulation of muscle atrophy-associated ubiquitin ligases, such as muscle atrophy F-box protein 1/atrogin-1, muscle ring finger protein-1, and casitas B-lineage lymphoma proto-oncogene-b. Dex also significantly decreased phosphorylated Foxo3a and increased total Foxo3a expression. Interestingly, Dex-induced ROS accumulation and Foxo3a expression were inhibited by morin (10 μM) pretreatment. Morin also prevented the Dex-induced reduction of myotube thickness, together with muscle protein degradation and suppression of the upregulation of atrophy-associated ubiquitin ligases. In conclusion, our results suggest that morin effectively prevents glucocorticoid-induced muscle atrophy by reducing oxidative stress.
Collapse
Affiliation(s)
- Anayt Ulla
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Takayuki Uchida
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Yukari Miki
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Kosuke Sugiura
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan; Department of Orthopedics, Institute of Medical Bioscience, Tokushima University Graduate School, Tokushima, Japan
| | | | - Takeshi Kobayashi
- Department of Physiology, Nagoya University, School of Medicine, Nagoya, Japan
| | - Ayako Ohno
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Reiko Nakao
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Katsuya Hirasaka
- Organization for Marine Science and Technology, Nagasaki University, Nagasaki, Japan
| | - Iori Sakakibara
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Takeshi Nikawa
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan.
| |
Collapse
|
12
|
Chen TC, Kuo T, Dandan M, Lee RA, Chang M, Villivalam SD, Liao SC, Costello D, Shankaran M, Mohammed H, Kang S, Hellerstein MK, Wang JC. The role of striated muscle Pik3r1 in glucose and protein metabolism following chronic glucocorticoid exposure. J Biol Chem 2021; 296:100395. [PMID: 33567340 PMCID: PMC8010618 DOI: 10.1016/j.jbc.2021.100395] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 11/03/2022] Open
Abstract
Chronic glucocorticoid exposure causes insulin resistance and muscle atrophy in skeletal muscle. We previously identified phosphoinositide-3-kinase regulatory subunit 1 (Pik3r1) as a primary target gene of skeletal muscle glucocorticoid receptors involved in the glucocorticoid-mediated suppression of insulin action. However, the in vivo functions of Pik3r1 remain unclear. Here, we generated striated muscle-specific Pik3r1 knockout (MKO) mice and treated them with a dexamethasone (DEX), a synthetic glucocorticoid. Treating wildtype (WT) mice with DEX attenuated insulin activated Akt activity in liver, epididymal white adipose tissue, and gastrocnemius (GA) muscle. This DEX effect was diminished in GA muscle of MKO mice, therefore, resulting in improved glucose and insulin tolerance in DEX-treated MKO mice. Stable isotope labeling techniques revealed that in WT mice, DEX treatment decreased protein fractional synthesis rates in GA muscle. Furthermore, histology showed that in WT mice, DEX treatment reduced GA myotube diameters. In MKO mice, myotube diameters were smaller than in WT mice, and there were more fast oxidative fibers. Importantly, DEX failed to further reduce myotube diameters. Pik3r1 knockout also decreased basal protein synthesis rate (likely caused by lower 4E-BP1 phosphorylation at Thr37/Thr46) and curbed the ability of DEX to attenuate protein synthesis rate. Finally, the ability of DEX to inhibit eIF2α phosphorylation and insulin-induced 4E-BP1 phosphorylation was reduced in MKO mice. Taken together, these results demonstrate the role of Pik3r1 in glucocorticoid-mediated effects on glucose and protein metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Tzu-Chieh Chen
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Taiyi Kuo
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Mohamad Dandan
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Rebecca A Lee
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Maggie Chang
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Sneha D Villivalam
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Szu-Chi Liao
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Damian Costello
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Mahalakshmi Shankaran
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Hussein Mohammed
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA
| | - Sona Kang
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Marc K Hellerstein
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA
| | - Jen-Chywan Wang
- Metabolic Biology Graduate Program, University of California Berkeley, Berkeley, California, USA; Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, California, USA; Endocrinology Graduate Program, University of California Berkeley, Berkeley, California, USA.
| |
Collapse
|
13
|
Inhibitory effects of curcuminoids on dexamethasone-induced muscle atrophy in differentiation of C2C12 cells. PHYTOMEDICINE PLUS 2021. [DOI: 10.1016/j.phyplu.2020.100012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
14
|
Li Y, Shi H, Chen R, Zhou S, Lei S, She Y. Role of miRNAs and lncRNAs in dexamethasone-induced myotube atrophy in vitro. Exp Ther Med 2020; 21:146. [PMID: 33456513 PMCID: PMC7791919 DOI: 10.3892/etm.2020.9577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 11/17/2020] [Indexed: 12/02/2022] Open
Abstract
Skeletal muscle atrophy is a well-known adverse effect of long-term glucocorticoid (GC) therapy. MicroRNAs (miRNAs or miRs) and long non-coding RNAs (lncRNAs) are important regulators in a number of physiological and pathological processes. However, the role of miRNAs and lncRNAs in the regulation of GC-treated muscle atrophy remains poorly understood. In the current study, muscular atrophy was induced and the results indicated that C2C12 myotubes were thinner than normal, while the expression of muscle ring finger protein 1 and Atrogin-1 was increased. The expression of nine miRNAs and seven lncRNAs associated with proliferation and differentiation were analyzed in a dexamethasone (DEX)-induced muscle atrophy cell model. In addition, the mRNA expression of the downstream targets of lncRNAs that were differentially expressed between DEX-treated and control cells were determined. The results indicated that the expression of miR-133a, miR-133b, miR-206 and five lncRNAs (increased Atrolnc-1, Dum, MAR1, linc-MD1 and decreased Myolinc) were significantly different between the DEX and the control group. Furthermore, the relative mRNA expression of Wnt5a and MyoD was significantly different between the two groups. The results of the current study indicated that some important miRNAs and lncRNAs are associated with DEX-induced muscle atrophy and have the potential to be further developed as a diagnostic tool for this condition.
Collapse
Affiliation(s)
- Yang Li
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Huacai Shi
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Rui Chen
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Shanyao Zhou
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Si Lei
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Yanling She
- Guangdong Traditional Medical and Sports Injury Rehabilitation Research Institute, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| |
Collapse
|
15
|
Armstrong VS, Fitzgerald LW, Bathe OF. Cancer-Associated Muscle Wasting-Candidate Mechanisms and Molecular Pathways. Int J Mol Sci 2020; 21:ijms21239268. [PMID: 33291708 PMCID: PMC7729509 DOI: 10.3390/ijms21239268] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Excessive muscle loss is commonly observed in cancer patients and its association with poor prognosis has been well-established. Cancer-associated sarcopenia differs from age-related wasting in that it is not responsive to nutritional intervention and exercise. This is related to its unique pathogenesis, a result of diverse and interconnected mechanisms including inflammation, disordered metabolism, proteolysis and autophagy. There is a growing body of evidence that suggests that the tumor is the driver of muscle wasting by its elaboration of mediators that influence each of these pro-sarcopenic pathways. In this review, evidence for these tumor-derived factors and putative mechanisms for inducing muscle wasting will be reviewed. Potential targets for future research and therapeutic interventions will also be reviewed.
Collapse
Affiliation(s)
- Victoria S. Armstrong
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (V.S.A.); (L.W.F.)
- Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Liam W. Fitzgerald
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (V.S.A.); (L.W.F.)
- Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Oliver F. Bathe
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; (V.S.A.); (L.W.F.)
- Department of Medical Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Departments of Surgery and Oncology, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Correspondence: ; Tel.: +1-403-521-3275
| |
Collapse
|
16
|
Shang Y, Kuang M, Wang Z, Huang Y, Liu L, Zhao X, Zhang R, Zhao Y, Peng R, Sun S, Yang Q, Yang Z. An Ultrashort Peptide-Based Supramolecular Hydrogel Mimicking IGF-1 to Alleviate Glucocorticoid-Induced Sarcopenia. ACS APPLIED MATERIALS & INTERFACES 2020; 12:34678-34688. [PMID: 32668906 DOI: 10.1021/acsami.0c09973] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Sarcopenia is a common disease in older people due to aging, and it can also occur in midlife because of diseases including cancer. Sarcopenia, characterized by rapid loss of muscle mass and accelerated loss of function, can lead to adverse outcomes such as frailty, falls, and even mortality. The development of pharmacological and therapeutic approaches to treat sarcopenia remains challenging. The growth status and quantity of myoblasts are the key factors directly affecting muscle formation. Therefore, enhancing the function of myoblasts is crucial for the treatment of sarcopenia. In our study, we introduced an insulin-like growth factor-I (IGF-1) mimicking supramolecular nanofibers/hydrogel formed by Nap-FFGSSSR that effectively promoted proliferation and significantly reduced dexamethasone-induced apoptosis of myoblasts, assisted myoblasts to differentiate into myotubes, and prevented the fibrosis of muscle tissue and the deposition of collagen, ultimately achieving outstanding effects in the treatment of sarcopenia. The RNA-sequencing results revealed that our nanofibers possessed similar bioactivity to the growth factor IGF-1, which increased the phosphorylation of Akt by activating the insulin signaling pathway. We prepared novel supramolecular nanomaterials to reverse glucocorticoid-induced myoblast dysfunction, which was promising for the treatment of muscular atrophy. In addition, we envisioned the generation of biofunctional nanomaterials by molecular self-assembly for the treatment of chronic diseases in middle-aged and older people.
Collapse
Affiliation(s)
- Yuna Shang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, P. R. China
| | - Mingjie Kuang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong University, Shandong 250014, P. R. China
| | - Zhongyan Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, P. R. China
| | - Ying Huang
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, P. R. China
| | - Lulu Liu
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, P. R. China
| | - Xige Zhao
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, P. R. China
| | - Rui Zhang
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, P. R. China
| | - Yanhong Zhao
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, P. R. China
| | - Rong Peng
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, P. R. China
| | - Shenghuan Sun
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, P. R. China
| | - Qiang Yang
- Department of Orthopedics, Tianjin Hospital, Tianjin 300211, P. R. China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, P. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P. R. China
| |
Collapse
|
17
|
Gouvêa AL, Martinez CG, Kurtenbach E. Determining Maximal Muscle Strength in Mice: Validity and Reliability of an Adapted Swimming Incremental Overload Test. J Strength Cond Res 2020; 34:2360-2368. [DOI: 10.1519/jsc.0000000000002777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
18
|
Jaworska J, Rodziewicz-Flis E, Kortas J, Kozłowska M, Micielska K, Babińska A, Laskowski R, Lombardi G, Ziemann E. Short-Term Resistance Training Supported by Whole-Body Cryostimulation Induced a Decrease in Myostatin Concentration and an Increase in Isokinetic Muscle Strength. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17155496. [PMID: 32751455 PMCID: PMC7432449 DOI: 10.3390/ijerph17155496] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 01/26/2023]
Abstract
The study aimed to determine whether combining cryostimulation with resistance training would effectively increase muscle strength, and if so, whether this adaptation would be related to changes in circulating levels of exerkines (i.e., mediators of systemic adaptation to exercise). Twenty-five students completed 12 sessions of resistance training, each followed by either cryostimulation (n = 15, 3 min exposure at -110 °C) or passive recovery (n = 10). Prior to and post this intervention, participants performed two eccentric cycling bouts (before and after training). At these points, serum concentrations of muscle damage marker (myoglobin), exerkines (interleukin 6 (IL-6), interleukin 15 (IL-15), irisin, brain-derived neurotrophic factor), hypertrophy-related factors (myostatin, insulin-like growth factor 1), and muscle strength were measured. The applied procedure reduced the physiological burden of the second eccentric cycling bout and myoglobin concentrations only in the group subject to cryostimulation. The same group also exhibited decreased levels of myostatin (from 4.7 ± 1.7 to 3.8 ± 1.8 ng·mL-1, p < 0.05). A significant and large interaction between the group × time was noted in IL-15 concentration (p = 0.01, ηp2=0.27). Training and cryostimulation induced a positive and likely significant improvement of isokinetic muscle strength. Altogether, obtained results support the claim that resistance training combined with cold exposure modified muscle strength through modulation of myostatin and IL-15 concentrations.
Collapse
Affiliation(s)
- Joanna Jaworska
- Department of Physiology, Gdansk University of Physical Education and Sport, Kazimierza Gorskiego 1, 80-336 Gdansk, Poland; (J.J.); (M.K.); (R.L.)
| | - Ewa Rodziewicz-Flis
- Department of Physical Therapy and Biological Regeneration, Gdansk University of Physical Education and Sport, Kazimierza Gorskiego 1, 80-336 Gdansk, Poland;
| | - Jakub Kortas
- Department of Sport, Gdansk University of Physical Education and Sport, Kazimierza Gorskiego 1, 80-336 Gdansk, Poland;
| | - Marta Kozłowska
- Department of Physiology, Gdansk University of Physical Education and Sport, Kazimierza Gorskiego 1, 80-336 Gdansk, Poland; (J.J.); (M.K.); (R.L.)
| | - Katarzyna Micielska
- Department of Physical Education and Lifelong sports, Poznan University of Physical Education, Krolowej Jadwigi 27/39, 61-871 Poznan, Poland;
| | - Anna Babińska
- Department of Endocrinology and Internal Medicine, Medical University, Marii Sklodowskiej-Curie 3, 80-001 Gdansk, Poland;
| | - Radosław Laskowski
- Department of Physiology, Gdansk University of Physical Education and Sport, Kazimierza Gorskiego 1, 80-336 Gdansk, Poland; (J.J.); (M.K.); (R.L.)
| | - Giovanni Lombardi
- Department of Athletics, Strength and Conditioning, Poznan University of Physical Education, Krolowej Jadwigi 27/39, 61-871 Poznan, Poland;
- IRCCS Istituto Ortopedico Galeazzi, Lab Experimental Biochemistry & Molecular Biology, Via Riccardo Galeazzi, 4, 20161 Milano, Italy
| | - Ewa Ziemann
- Department of Athletics, Strength and Conditioning, Poznan University of Physical Education, Krolowej Jadwigi 27/39, 61-871 Poznan, Poland;
- Correspondence:
| |
Collapse
|
19
|
Iron Status in Elderly Women Impacts Myostatin, Adiponectin and Osteocalcin Levels Induced by Nordic Walking Training. Nutrients 2020; 12:nu12041129. [PMID: 32316589 PMCID: PMC7231223 DOI: 10.3390/nu12041129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 01/26/2023] Open
Abstract
Impaired iron metabolism is associated with increased risk of many morbidities. Exercise was shown to have a beneficial role; however, the mechanism is not well understood. The purpose of this study was to assess the relationship between exerkines and iron metabolism in elderly women before and after 12 weeks of Nordic Walking (NW) training. Exerkines like myostatin, adiponectin, and osteocalcin have been shown to have several positive effects on metabolism. Thirty-six post-menopausal women (66 ± 5 years old, mean ± SD) were randomly assigned to a NW intervention group (n = 18; body mass, 68.8 ± 11.37 kg; fat, 23.43 ± 7.5 kg; free fat mass, 45.37 ± 5.92 kg) or a control group (n = 18; body mass, 68.34 ± 11.81 kg; fat, 23.61 ± 10.03 kg; free fat mass, 44.73 ± 3.9 kg). The training was performed three times a week for 12 weeks, with the intensity adjusted to 70% of the individual maximum ability. Before and one day after the 12-weeks intervention, performance indices were assessed using a senior fitness test. Blood samples (5 mL) were obtained from the participants between 7 and 8 AM, following an overnight fast, at baseline and one day immediately after the 12-week training program. A significant and large time × group interaction was observed for iron (NW: 98.6 ± 26.68 to 76.1 ± 15.31; CON: 100.6 ± 25.37 to 99.1 ± 27.2; p = 0.01; ηp2 = 0.21), myostatin (NW: 4.42 ± 1.97 to 3.83 ± 1.52; CON: 4.11 ± 0.95 to 4.84 ± 1.19; p = 0.00; ηp2 = 0.62), adiponectin (NW: 12.0 ± 9.46 to 14.6 ± 10.64; CON: 12.8 ± 8.99 to 11.9 ± 8.53; p = 0.00; ηp2 = 0.58), and osteocalcin (NW: 38.9 ± 26.04 to 41.6 ± 25.09; CON: 37.1 ± 33.2 to 37.2 ± 32.29; p = 0.03; ηp2 = 0.13). Furthermore, we have observed the correlations: basal ferritin levels were inversely correlated with changes in myostatin (r = −0.51, p = 0.05), change in adiponectin, and change in serum iron (r = −0.45, p = 0.05), basal iron, and osteocalcin after training (r = -0.55, p = 0.04). These findings indicate that iron modulates NW training-induced changes in exerkine levels.
Collapse
|
20
|
Recovery of muscle mass and muscle oxidative phenotype following disuse does not require GSK-3 inactivation. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165740. [PMID: 32087280 DOI: 10.1016/j.bbadis.2020.165740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/24/2020] [Accepted: 02/18/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Physical inactivity contributes to muscle wasting and reductions in mitochondrial oxidative phenotype (OXPHEN), reducing physical performance and quality of life during aging and in chronic disease. Previously, it was shown that inactivation of glycogen synthase kinase (GSK)-3β stimulates muscle protein accretion, myogenesis, and mitochondrial biogenesis. Additionally, GSK-3β is inactivated during recovery of disuse-induced muscle atrophy. AIM Therefore, we hypothesize that GSK-3 inhibition is required for reloading-induced recovery of skeletal muscle mass and OXPHEN. METHODS Wild-type (WT) and whole-body constitutively active (C.A.) Ser21/9 GSK-3α/β knock-in mice were subjected to a 14-day hind-limb suspension/14-day reloading protocol. Soleus muscle mass, fiber cross-sectional area (CSA), OXPHEN (abundance of sub-units of oxidative phosphorylation (OXPHOS) complexes and fiber-type composition), as well as expression levels of their main regulators (respectively protein synthesis/degradation, myogenesis and peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α) signaling) were monitored. RESULTS Subtle but consistent differences suggesting suppression of protein turnover signaling and decreased expression of several OXPHOS sub-units and PGC-1α signaling constituents were observed at baseline in C.A. GSK-3 versus WT mice. Although soleus mass recovery during reloading occurred more rapidly in C.A. GSK-3 mice, this was not accompanied by a parallel increased CSA. The OXPHEN response to reloading was not distinct between C.A. GSK-3 and WT mice. No consistent or significant differences in reloading-induced changes in the regulatory steps of protein turnover, myogenesis or muscle OXPHEN were observed in C.A. GSK-3 compared to WT muscle. CONCLUSION This study indicates that GSK-3 inactivation is dispensable for reloading-induced recovery of muscle mass and OXPHEN.
Collapse
|
21
|
Watanabe H, Enoki Y, Maruyama T. Sarcopenia in Chronic Kidney Disease: Factors, Mechanisms, and Therapeutic Interventions. Biol Pharm Bull 2020; 42:1437-1445. [PMID: 31474705 DOI: 10.1248/bpb.b19-00513] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic kidney disease (CKD), a chronic catabolic condition, is characterized by muscle wasting and decreased muscle endurance. Many insights into the molecular mechanisms of muscle wasting in CKD have been obtained. A persistent imbalance between protein degradation and synthesis in muscle causes muscle wasting. During muscle wasting, high levels of reactive oxygen species (ROS) and inflammatory cytokines are detected in muscle. These increased ROS and inflammatory cytokine levels induce the expression of myostatin. The myostatin binding to its receptor activin A receptor type IIB stimulates the expression of atrogenes such as atrogin-1 and muscle ring factor 1, members of the muscle-specific ubiquitin ligase family. Impaired mitochondrial function also contributes to reducing muscle endurance. The increased protein-bound uremic toxin, parathyroid hormone, glucocorticoid, and angiotensin II levels that are observed in CKD all have a negative effect on muscle mass and endurance. Among the protein-bound uremic toxins, indoxyl sulfate, an indole-containing compound has the potential to induce muscle atrophy by stimulating ROS-mediated myostatin and atrogenes expression. Indoxyl sulfate also impairs mitochondrial function. Some potential therapeutic approaches based on the muscle wasting mechanisms in CKD are currently in the testing stages.
Collapse
Affiliation(s)
- Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | - Yuki Enoki
- Division of Pharmacodynamics, Keio University Faculty of Pharmacy
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| |
Collapse
|
22
|
Duan Y, Zheng C, Zhong Y, Song B, Yan Z, Kong X, Deng J, Li F, Yin Y. Beta-hydroxy beta-methyl butyrate decreases muscle protein degradation via increased Akt/FoxO3a signaling and mitochondrial biogenesis in weanling piglets after lipopolysaccharide challenge. Food Funct 2019; 10:5152-5165. [PMID: 31373594 DOI: 10.1039/c9fo00769e] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The aim of this study was to investigate the effects of dietary β-hydroxy-β-methylbutyrate (HMB) on lipopolysaccharide (LPS)-induced muscle atrophy and to investigate the mechanisms involved. Sixty pigs (21 ± 2 days old, 5.86 ± 0.18 kg body weight) were used in a 2 × 3 factorial design and the main factors included diet (0, 0.60%, or 1.20% HMB) and immunological challenge (LPS or saline). After 15 d of treatment with LPS and/or HMB, growth performance, blood parameters, and muscle protein degradation rate were measured. The results showed that in LPS-injected pigs, 0.60% HMB supplementation increased the average daily gain and average daily feed intake and decreased the feed : gain ratio (P < 0.05), with a concurrent increase of lean percentage. Moreover, 0.60% HMB supplementation decreased the serum concentrations of blood urea nitrogen, IL-1β, and TNF-α and the rate of protein degradation as well as cell apoptosis in selected muscles (P < 0.05). In addition, dietary HMB supplementation (0.60%) regulated the expression of genes involved in mitochondrial biogenesis and increased the phosphorylation of Akt and Forkhead Box O3a (FoxO3a) in selected muscles, accompanied by decreased protein expression of muscle RING finger 1 and muscle atrophy F-box. These results indicate that HMB may exert protective effects against LPS-induced muscle atrophy by normalizing the Akt/FoxO3a axis that regulates ubiquitin proteolysis and by improving mitochondrial biogenesis.
Collapse
Affiliation(s)
- Yehui Duan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences; Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production; Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Skeletal muscle atrophy is a common side effect of most human diseases. Muscle loss is not only detrimental for the quality of life but it also dramatically impairs physiological processes of the organism and decreases the efficiency of medical treatments. While hypothesized for years, the existence of an atrophying programme common to all pathologies is still incompletely solved despite the discovery of several actors and key regulators of muscle atrophy. More than a decade ago, the discovery of a set of genes, whose expression at the mRNA levels were similarly altered in different catabolic situations, opened the way of a new concept: the presence of atrogenes, i.e. atrophy-related genes. Importantly, the atrogenes are referred as such on the basis of their mRNA content in atrophying muscles, the regulation at the protein level being sometimes more complicate to elucidate. It should be noticed that the atrogenes are markers of atrophy and that their implication as active inducers of atrophy is still an open question for most of them. While the atrogene family has grown over the years, it has mostly been incremented based on data coming from rodent models. Whether the rodent atrogenes are valid for humans still remain to be established. An "atrogene" was originally defined as a gene systematically up- or down-regulated in several catabolic situations. Even if recent works often restrict this notion to the up-regulation of a limited number of proteolytic enzymes, it is important to keep in mind the big picture view. In this review, we provide an update of the validated and potential rodent atrogenes and the metabolic pathways they belong, and based on recent work, their relevance in human physio-pathological situations. We also propose a more precise definition of the atrogenes that integrates rapid recovery when catabolic stimuli are stopped or replaced by anabolic ones.
Collapse
Affiliation(s)
- Daniel Taillandier
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France.
| | - Cécile Polge
- Université Clermont Auvergne, INRA, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000, Clermont-Ferrand, France
| |
Collapse
|
24
|
Protective Effect of Pyropia yezoensis Peptide on Dexamethasone-Induced Myotube Atrophy in C2C12 Myotubes. Mar Drugs 2019; 17:md17050284. [PMID: 31083497 PMCID: PMC6563069 DOI: 10.3390/md17050284] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023] Open
Abstract
Dexamethasone (DEX), a synthetic glucocorticoid, causes skeletal muscle atrophy. This study examined the protective effects of Pyropia yezoensis peptide (PYP15) against DEX-induced myotube atrophy and its association with insulin-like growth factor-I (IGF-I) and the Akt/mammalian target of rapamycin (mTOR)-forkhead box O (FoxO) signaling pathway. To elucidate the molecular mechanisms underlying the effects of PYP15 on DEX-induced myotube atrophy, C2C12 myotubes were treated for 24 h with 100 μM DEX in the presence or absence of 500 ng/mL PYP15. Cell viability assays revealed no PYP15 toxicity in C2C12 myotubes. PYP15 activated the insulin-like growth factor-I receptor (IGF-IR) and Akt-mTORC1 signaling pathway in DEX-induced myotube atrophy. In addition, PYP15 markedly downregulated the nuclear translocation of transcription factors FoxO1 and FoxO3a, and inhibited 20S proteasome activity. Furthermore, PYP15 inhibited the autophagy-lysosomal pathway in DEX-stimulated myotube atrophy. Our findings suggest that PYP15 treatment protected against myotube atrophy by regulating IGF-I and the Akt-mTORC1-FoxO signaling pathway in skeletal muscle. Therefore, PYP15 treatment appears to exert protective effects against skeletal muscle atrophy.
Collapse
|
25
|
Preau S, Ambler M, Sigurta A, Kleyman A, Dyson A, Hill NE, Boulanger E, Singer M. Protein recycling and limb muscle recovery after critical illness in slow- and fast-twitch limb muscle. Am J Physiol Regul Integr Comp Physiol 2019; 316:R584-R593. [PMID: 30789789 DOI: 10.1152/ajpregu.00221.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
An impaired capacity of muscle to regenerate after critical illness results in long-term functional disability. We previously described in a long-term rat peritonitis model that gastrocnemius displays near-normal histology whereas soleus demonstrates a necrotizing phenotype. We thus investigated the link between the necrotizing phenotype of critical illness myopathy and proteasome activity in these two limb muscles. We studied male Wistar rats that underwent an intraperitoneal injection of the fungal cell wall constituent zymosan or n-saline as a sham-treated control. Rats (n = 74) were killed at 2, 7, and 14 days postintervention with gastrocnemius and soleus muscle removed and studied ex vivo. Zymosan-treated animals displayed an initial reduction of body weight but a persistent decrease in mass of both lower hindlimb muscles. Zymosan increased chymotrypsin- and trypsin-like proteasome activities in gastrocnemius at days 2 and 7 but in soleus at day 2 only. Activated caspases-3 and -9, polyubiquitin proteins, and 14-kDa fragments of myofibrillar actin (proteasome substrates) remained persistently increased from day 2 to day 14 in soleus but not in gastrocnemius. These results suggest that a relative proteasome deficiency in soleus is associated with a necrotizing phenotype during long-term critical illness. Rescuing proteasome clearance may offer a potential therapeutic option to prevent long-term functional disability in critically ill patients.
Collapse
Affiliation(s)
- Sebastien Preau
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London , London , United Kingdom.,Université de Lille, Centre Hospitalier et Universitaire de Lille, INSERM, Lille Inflammation Research International Center, Lille, France
| | - Michael Ambler
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London , London , United Kingdom
| | - Anna Sigurta
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London , London , United Kingdom
| | - Anna Kleyman
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London , London , United Kingdom
| | - Alex Dyson
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London , London , United Kingdom
| | - Neil E Hill
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London , London , United Kingdom
| | - Eric Boulanger
- Université de Lille, Centre Hospitalier et Universitaire de Lille, INSERM, Lille Inflammation Research International Center, Lille, France
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London , London , United Kingdom
| |
Collapse
|
26
|
Tumor-Derived Ligands Trigger Tumor Growth and Host Wasting via Differential MEK Activation. Dev Cell 2019; 48:277-286.e6. [PMID: 30639055 DOI: 10.1016/j.devcel.2018.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/21/2018] [Accepted: 12/04/2018] [Indexed: 12/19/2022]
Abstract
Interactions between tumors and host tissues play essential roles in tumor-induced systemic wasting and cancer cachexia, including muscle wasting and lipid loss. However, the pathogenic molecular mechanisms of wasting are still poorly understood. Using a fly model of tumor-induced organ wasting, we observed aberrant MEK activation in both tumors and host tissues of flies bearing gut-yki3SA tumors. We found that host MEK activation results in muscle wasting and lipid loss, while tumor MEK activation is required for tumor growth. Strikingly, host MEK suppression alone is sufficient to abolish the wasting phenotypes without affecting tumor growth. We further uncovered that yki3SA tumors produce the vein (vn) ligand to trigger autonomous Egfr/MEK-induced tumor growth and produce the PDGF- and VEGF-related factor 1 (Pvf1) ligand to non-autonomously activate host Pvr/MEK signaling and wasting. Altogether, our results demonstrate the essential roles and molecular mechanisms of differential MEK activation in tumor-induced host wasting.
Collapse
|
27
|
Fappi A, Neves JDC, Kawasaki KA, Bacelar L, Sanches LN, P. da Silva F, Larina‐Neto R, Chadi G, Zanoteli E. Omega-3 multiple effects increasing glucocorticoid-induced muscle atrophy: autophagic, AMPK and UPS mechanisms. Physiol Rep 2019; 7:e13966. [PMID: 30648357 PMCID: PMC6333722 DOI: 10.14814/phy2.13966] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/23/2022] Open
Abstract
Muscle atrophy occurs in many conditions, including use of glucocorticoids. N-3 (omega-3) is widely consumed due its healthy properties; however, concomitant use with glucocorticoids can increase its side effects. We evaluated the influences of N-3 on glucocorticoid atrophy considering IGF-1, Myostatin, MEK/ERK, AMPK pathways besides the ubiquitin-proteasome system (UPS) and autophagic/lysosomal systems. Sixty animals constituted six groups: CT, N-3 (EPA 100 mg/kg/day for 40 days), DEXA 1.25 (DEXA 1.25 mg/kg/day for 10 days), DEXA 1.25 + N3 (EPA for 40 days + DEXA 1.25 mg/kg/day for the last 10 days), DEXA 2.5 (DEXA 2.5 mg/kg/day for 10 days), and DEXA 2.5 + N3 (EPA for 40 days + DEXA 2.5 mg/kg/day for 10 days). Results: N-3 associated with DEXA increases atrophy (fibers 1 and 2A), FOXO3a, P-SMAD2/3, Atrogin-1/MAFbx (mRNA) expression, and autophagic protein markers (LC3II, LC3II/LC3I, LAMP-1 and acid phosphatase). Additionally, N-3 supplementation alone decreased P-FOXO3a, PGC1-alpha, and type 1 muscle fiber area. Conclusion: N-3 supplementation increases muscle atrophy caused by DEXA in an autophagic, AMPK and UPS process.
Collapse
Affiliation(s)
- Alan Fappi
- Department of NeurologyFaculdade de Medicina FMUSPUniversidade de Sao PauloSP, Brazil
| | - Juliana de C. Neves
- Department of NeurologyFaculdade de Medicina FMUSPUniversidade de Sao PauloSP, Brazil
| | - Karine A. Kawasaki
- Department of NeurologyFaculdade de Medicina FMUSPUniversidade de Sao PauloSP, Brazil
| | - Luana Bacelar
- Department of NeurologyFaculdade de Medicina FMUSPUniversidade de Sao PauloSP, Brazil
| | - Leandro N. Sanches
- Department of NeurologyFaculdade de Medicina FMUSPUniversidade de Sao PauloSP, Brazil
| | - Felipe P. da Silva
- Department of NeurologyFaculdade de Medicina FMUSPUniversidade de Sao PauloSP, Brazil
| | - Rubens Larina‐Neto
- Department of NeurologyFaculdade de Medicina FMUSPUniversidade de Sao PauloSP, Brazil
| | - Gerson Chadi
- Department of NeurologyFaculdade de Medicina FMUSPUniversidade de Sao PauloSP, Brazil
| | - Edmar Zanoteli
- Department of NeurologyFaculdade de Medicina FMUSPUniversidade de Sao PauloSP, Brazil
| |
Collapse
|
28
|
|
29
|
Protein synthesis signaling in skeletal muscle is refractory to whey protein ingestion during a severe energy deficit evoked by prolonged exercise and caloric restriction. Int J Obes (Lond) 2018; 43:872-882. [PMID: 30242237 DOI: 10.1038/s41366-018-0174-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/24/2018] [Accepted: 06/24/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Exercise and protein ingestion preserve muscle mass during moderate energy deficits. OBJECTIVE To determine the molecular mechanisms by which exercise and protein ingestion may spare muscle mass during severe energy deficit (5500 kcal/day). DESIGN Fifteen overweight, but otherwise healthy men, underwent a pre-test (PRE), caloric restriction (3.2 kcals/kg body weight/day) + exercise (45 min one-arm cranking + 8 h walking) for 4 days (CRE), followed by a control diet (CD) for 3 days, with a caloric content similar to pre-intervention while exercise was reduced to less than 10,000 steps per day. During CRE, participants ingested either whey protein (PRO, n = 8) or sucrose (SU, n = 7) (0.8 g/kg body weight/day). Muscle biopsies were obtained from the trained and untrained deltoid, and vastus lateralis. RESULTS Following CRE and CD, serum concentrations of leptin, insulin, and testosterone were reduced, whereas cortisol and the catabolic index (cortisol/total testosterone) increased. The Akt/mTor/p70S6K pathway and total eIF2α were unchanged, while total 4E-BP1 and Thr37/464E-BP1 were higher. After CRE, plasma BCAA and EAA were elevated, with a greater response in PRO group, and total GSK3β, pSer9GSK3β, pSer51eIF2α, and pSer51eIF2α/total eIF2α were reduced, with a greater response of pSer9GSK3β in the PRO group. The changes in signaling were associated with the changes in leptin, insulin, amino acids, cortisol, cortisol/total testosterone, and lean mass. CONCLUSIONS During severe energy deficit, pSer9GSK3β levels are reduced and human skeletal muscle becomes refractory to the anabolic effects of whey protein ingestion, regardless of contractile activity. These effects are associated with the changes in lean mass and serum insulin, testosterone, and cortisol concentrations.
Collapse
|
30
|
Woodworth-Hobbs ME, Perry BD, Rahnert JA, Hudson MB, Zheng B, Russ Price S. Docosahexaenoic acid counteracts palmitate-induced endoplasmic reticulum stress in C2C12 myotubes: Impact on muscle atrophy. Physiol Rep 2018; 5. [PMID: 29199180 PMCID: PMC5727283 DOI: 10.14814/phy2.13530] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 11/08/2017] [Indexed: 12/21/2022] Open
Abstract
Lipid accumulation in skeletal muscle results in dysregulation of protein metabolism and muscle atrophy. We previously reported that treating C2C12 myotubes with palmitate (PA), a saturated fatty acid, increases the overall rate of proteolysis via activation of the ubiquitin-proteasome and autophagy systems; co-treatment with the omega-3 polyunsaturated fatty acid docosahexaenoic acid (DHA) prevents the PA-induced responses. Others have reported that PA induces endoplasmic reticulum (ER) stress which initiates the unfolded protein response (UPR), a collective group of responses that can lead to activation of caspase-mediated proteolysis and autophagy. Presently, we tested the hypothesis that DHA protects against PA-induced ER stress/UPR and its atrophy-related responses in muscle cells. C2C12 myotubes were treated with 500 μmol/L PA and/or 100 μmol/L DHA for 24 h. Proteins and mRNA associated with ER stress/UPR, autophagy, and caspase-3 activation were evaluated. PA robustly increased the phosphorylation of protein kinase R (PKR)-like ER kinase (PERK) and eukaryotic initiation factor 2α (eIF2α). It also increased the mRNAs encoding activating transcription factor 4 (ATF4), spliced X-box binding protein 1 (XBP1s), C/EBP homologous protein (CHOP), and autophagy-related 5 (Atg5) as well as the protein levels of the PERK target nuclear factor erythroid 2-related factor (Nrf2), CHOP, and cleaved (i.e., activated) caspase-3. Co-treatment with DHA prevented all of the PA-induced responses. Our results indicate that DHA prevents PA-induced muscle cell atrophy, in part, by preventing ER stress/UPR, a process that leads to activation of caspase-mediated proteolysis and an increase in expression of autophagy-related genes.
Collapse
Affiliation(s)
- Myra E Woodworth-Hobbs
- Nutrition and Health Sciences Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, Georgia.,Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - Ben D Perry
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia.,Atlanta VA Medical Center, Decatur, Georgia
| | - Jill A Rahnert
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia.,Atlanta VA Medical Center, Decatur, Georgia
| | - Matthew B Hudson
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia.,University of Delaware, Department of Kinesiology and Applied Physiology, Newark, Delaware
| | - Bin Zheng
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia
| | - S Russ Price
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia.,Atlanta VA Medical Center, Decatur, Georgia.,Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| |
Collapse
|
31
|
Xie Y, Perry BD, Espinoza D, Zhang P, Price SR. Glucocorticoid-induced CREB activation and myostatin expression in C2C12 myotubes involves phosphodiesterase-3/4 signaling. Biochem Biophys Res Commun 2018; 503:1409-1414. [PMID: 30025893 PMCID: PMC6173943 DOI: 10.1016/j.bbrc.2018.07.056] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 11/02/2022]
Abstract
Muscle atrophy in metabolic conditions like chronic kidney disease (CKD) and diabetes are associated with glucocorticoid production, dysfunctional insulin/Akt/FoxO3 signaling and increased myostatin expression. We recently found that CREB, a transcription factor proposed to regulate myostatin expression, is highly phosphorylated in some wasting conditions. Based on a novel Akt-PDE3/4 signaling paradigm, we hypothesized that reduced Akt signaling contributes to CREB activation and myostatin expression. C2C12 myotubes were incubated with dexamethasone (Dex), an atrophy-inducing synthetic glucocorticoid. Akt/CREB signaling and myostatin expression were evaluated by immunoblot and qPCR analyses. Inhibitors of Akt, phosphodiesterase (PDE)-3/4, and protein kinase A (PKA) signaling were used to test our hypothesis. Incubating myotubes with Dex for 3-24 h inhibited Akt phosphorylation and enhanced CREB phosphorylation as well as myostatin mRNA and protein. Inhibition of PI3K/Akt signaling with LY294002 similarly increased CREB phosphorylation. Isobutyl-methylxanthine (IBMX, a pan PDE inhibitor), milrinone (PDE3 inhibitor) and rolipram (PDE4 inhibitor) augmented CREB phosphorylation and myostatin expression. Inhibition of protein kinase A by PKI reverted Dex- or IBMX-induced CREB phosphorylation and myostatin expression. Our study provides evidence supporting a newly identified mechanism by which a glucocorticoid-related reduction in Akt signaling contributes to myostatin expression via CREB activation.
Collapse
Affiliation(s)
- Yang Xie
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA; Department of Nephrology, Xiangya Hospital and Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, PR China; Department of Nephrology, Beijing Hospital, Beijing 100730, PR China
| | - Ben D Perry
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA; Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA; School of Science and Health, Western Sydney University, Campbelltown NSW 2560, Australia
| | - Daniel Espinoza
- Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - Peng Zhang
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA; Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - S Russ Price
- Department of Medicine, Renal Division, Emory University, Atlanta, GA 30322, USA; Research Service Line, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA; Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
32
|
Ferbeyre G. Aberrant signaling and senescence associated protein degradation. Exp Gerontol 2018; 107:50-54. [DOI: 10.1016/j.exger.2017.06.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 11/17/2022]
|
33
|
Mota R, Parry TL, Yates CC, Qiang Z, Eaton SC, Mwiza JM, Tulasi D, Schisler JC, Patterson C, Zaglia T, Sandri M, Willis MS. Increasing Cardiomyocyte Atrogin-1 Reduces Aging-Associated Fibrosis and Regulates Remodeling in Vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1676-1692. [PMID: 29758183 DOI: 10.1016/j.ajpath.2018.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 03/10/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
Abstract
The muscle-specific ubiquitin ligase atrogin-1 (MAFbx) has been identified as a critical regulator of pathologic and physiological cardiac hypertrophy; it regulates these processes by ubiquitinating transcription factors [nuclear factor of activated T-cells and forkhead box O (FoxO) 1/3]. However, the role of atrogin-1 in regulating transcription factors in aging has not previously been described. Atrogin-1 cardiomyocyte-specific transgenic (Tg+) adult mice (α-major histocompatibility complex promoter driven) have normal cardiac function and size. Herein, we demonstrate that 18-month-old atrogin-1 Tg+ hearts exhibit significantly increased anterior wall thickness without functional impairment versus wild-type mice. Histologic analysis at 18 months revealed atrogin-1 Tg+ mice had significantly less fibrosis and significantly greater nuclei and cardiomyocyte cross-sectional analysis. Furthermore, by real-time quantitative PCR, atrogin-1 Tg+ had increased Col 6a4, 6a5, 6a6, matrix metalloproteinase 8 (Mmp8), and Mmp9 mRNA, suggesting a role for atrogin-1 in regulating collagen deposits and MMP-8 and MMP-9. Because atrogin-1 Tg+ mice exhibited significantly less collagen deposition and protein levels, enhanced Mmp8 and Mmp9 mRNA may offer one mechanism by which collagen levels are kept in check in the aged atrogin-1 Tg+ heart. In addition, atrogin-1 Tg+ hearts showed enhanced FoxO1/3 activity. The present study shows a novel link between atrogin-1-mediated regulation of FoxO1/3 activity and reduced collagen deposition and fibrosis in the aged heart. Therefore, targeting FoxO1/3 activity via the muscle-specific atrogin-1 ubiquitin ligase may offer a muscle-specific method to modulate aging-related cardiac fibrosis.
Collapse
Affiliation(s)
- Roberto Mota
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Traci L Parry
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Cecelia C Yates
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zhaoyan Qiang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina; Department of Pharmacology, Tianjin Medical University, Tianjin, China
| | - Samuel C Eaton
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina
| | - Jean Marie Mwiza
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Deepthi Tulasi
- Department of Biology, University of North Carolina, Chapel Hill, North Carolina
| | - Jonathan C Schisler
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina; Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina
| | - Cam Patterson
- Presbyterian Hospital/Weill-Cornell Medical Center, New York, New York
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy
| | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Dulbecco Telethon Institute, Padova, Italy
| | - Monte S Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina; Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina; Indiana Center for Musculoskeletal Health and Department of Pathology and Laboratory Medicine, University of Indiana School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
34
|
Perry BD, Rahnert JA, Xie Y, Zheng B, Woodworth-Hobbs ME, Price SR. Palmitate-induced ER stress and inhibition of protein synthesis in cultured myotubes does not require Toll-like receptor 4. PLoS One 2018; 13:e0191313. [PMID: 29329354 PMCID: PMC5766250 DOI: 10.1371/journal.pone.0191313] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 01/01/2018] [Indexed: 12/13/2022] Open
Abstract
Saturated fatty acids, such as palmitate, are elevated in metabolically dysfunctional conditions like type 2 diabetes mellitus. Palmitate has been shown to impair insulin sensitivity and suppress protein synthesis while upregulating proteolytic systems in skeletal muscle. Increased sarco/endoplasmic reticulum (ER) stress and subsequent activation of the unfolded protein response may contribute to the palmitate-induced impairment of muscle protein synthesis. In some cell types, ER stress occurs through activation of the Toll-like receptor 4 (TLR4). Given the link between ER stress and suppression of protein synthesis, we investigated whether palmitate induces markers of ER stress and protein synthesis by activating TLR4 in cultured mouse C2C12 myotubes. Myotubes were treated with vehicle, a TLR4-specific ligand (lipopolysaccharides), palmitate, or a combination of palmitate plus a TLR4-specific inhibitor (TAK-242). Inflammatory indicators of TLR4 activation (IL-6 and TNFα) and markers of ER stress were measured, and protein synthesis was assessed using puromycin incorporation. Palmitate substantially increased the levels of IL-6, TNF-α, CHOP, XBP1s, and ATF 4 mRNAs and augmented the levels of CHOP, XBP1s, phospho-PERK and phospho-eIF2α proteins. The TLR4 antagonist attenuated both acute palmitate and LPS-induced increases in IL-6 and TNFα, but did not reduce ER stress signaling with either 6 h or 24 h palmitate treatment. Similarly, treating myotubes with palmitate for 6 h caused a 43% decline in protein synthesis consistent with an increase in phospho-eIF2α, and the TLR4 antagonist did not alter these responses. These results suggest that palmitate does not induce ER stress through TLR4 in muscle, and that palmitate impairs protein synthesis in skeletal muscle in part by induction of ER stress.
Collapse
Affiliation(s)
- Ben D. Perry
- Department of Medicine, Renal Division, Emory University, Atlanta, GA, United States of America
| | - Jill A. Rahnert
- Department of Medicine, Renal Division, Emory University, Atlanta, GA, United States of America
| | - Yang Xie
- Department of Medicine, Renal Division, Emory University, Atlanta, GA, United States of America
| | - Bin Zheng
- Department of Medicine, Renal Division, Emory University, Atlanta, GA, United States of America
| | - Myra E. Woodworth-Hobbs
- Department of Medicine, Renal Division, Emory University, Atlanta, GA, United States of America
- Center for the Study of Human Health, Emory College of Arts and Sciences, Emory University, Atlanta, GA, United States of America
| | - S. Russ Price
- Department of Medicine, Renal Division, Emory University, Atlanta, GA, United States of America
- Atlanta VA Medical Center, Decatur, GA, United States of America
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC, United States of America
- * E-mail:
| |
Collapse
|
35
|
van Doeselaar S, Burgering BMT. FOXOs Maintaining the Equilibrium for Better or for Worse. Curr Top Dev Biol 2018; 127:49-103. [PMID: 29433740 DOI: 10.1016/bs.ctdb.2017.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A paradigm shift is emerging within the FOXO field and accumulating evidence indicates that we need to reappreciate the role of FOXOs, at least in cancer development. Here, we discuss the possibility that FOXOs are both tumor suppressors as well as promoters of tumor progression. This is mostly dependent on the biological context. Critical to this dichotomous role is the notion that FOXOs are central in preserving cellular homeostasis in redox control, genomic stability, and protein turnover. From this perspective, a paradoxical role in both suppressing and enhancing tumor progression can be reconciled. As many small molecules targeting the PI3K pathway are developed by big pharmaceutical companies and/or are in clinical trial, we will discuss what the consequences may be for the context-dependent role of FOXOs in tumor development in treatment options based on active PI3K signaling in tumors.
Collapse
Affiliation(s)
- Sabina van Doeselaar
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Marino JS, Ruban C, Blanchette CM. The Association of COPD Exacerbations with New Onset Type 2 Diabetes among Medicare Patients. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2017; 5:183-193. [PMID: 35620780 PMCID: PMC9090466 DOI: 10.36469/9810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Objective: Chronic obstructive pulmonary disease (COPD) is highly prevalent in the elderly population and typically reduces overall quality of life. Exacerbations of COPD are commonly treated with corticosteroids, a class of drug known to cause insulin resistance. The objective of this study was to assess the rate of exacerbations requiring emergency room visits, hospitalizations or any medical encounter (a combination of emergency room and hospitalizations) between COPD patients who did and did not develop type 2 diabetes. Research Design and Methods: A case-control study of COPD patients from the 2011-2012 Medicare 5% sample Limited Data Set (LDS) was conducted. Beneficiaries with at least 1 year of continuous enrollment and evidence of > 2 COPD-related claims (>1 primary diagnosis) were included in the study. Cases were defined as a beneficiary with a new claim for type 2 diabetes, whereas controls lacked evidence of type 2 diabetes (beneficiaries with evidence of non-incident type 2 diabetes were excluded). Results: Of 27 456 COPD beneficiaries, 1274 developed incident type 2 diabetes (4.6%). After matching, 2536 beneficiaries were assigned as cases (n = 1268) and controls (n = 1268). Cases in the emergency room (1.97 claims per person) (p = <0.001) and hospitalizations (2.02 claims per person) (p = <0.001) had a higher rate of exacerbations. Conclusion: Our findings suggest that patients that were hospitalized and visited the emergency room for COPD exacerbations had a greater likelihood of type 2 diabetes. Type 2 diabetes may be associated with exposure to corticosteroids as a result of the treatment for exacerbations. Future work should investigate the risk for type 2 diabetes in COPD patients treated with corticosteroids.
Collapse
Affiliation(s)
- Joseph S Marino
- Health Informatics and Outcomes Research Academy, Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina, Charlotte, NC, USA
| | - Cynthiya Ruban
- Health Informatics and Outcomes Research Academy, Department of Public Health Sciences, University of North Carolina, Charlotte, NC, USA
| | - Christopher M Blanchette
- Health Informatics and Outcomes Research Academy, Department of Public Health Sciences, University of North Carolina, Charlotte, NC, USA
| |
Collapse
|
37
|
Bartley JM, Pan SJ, Keilich SR, Hopkins JW, Al-Naggar IM, Kuchel GA, Haynes L. Aging augments the impact of influenza respiratory tract infection on mobility impairments, muscle-localized inflammation, and muscle atrophy. Aging (Albany NY) 2017; 8:620-35. [PMID: 26856410 PMCID: PMC4925818 DOI: 10.18632/aging.100882] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 01/20/2016] [Indexed: 12/16/2022]
Abstract
Although the influenza virus only infects the respiratory system, myalgias are commonly experienced during infection. In addition to a greater risk of hospitalization and death, older adults are more likely to develop disability following influenza infection; however, this relationship is understudied. We hypothesized that upon challenge with influenza, aging would be associated with functional impairments, as well as upregulation of skeletal muscle inflammatory and atrophy genes. Infected young and aged mice demonstrated decreased mobility and altered gait kinetics. These declines were more prominent in hind limbs and in aged mice. Skeletal muscle expression of genes involved in inflammation, as well as muscle atrophy and proteolysis, increased during influenza infection with an elevated and prolonged peak in aged mice. Infection also decreased expression of positive regulators of muscle mass and myogenesis components to a greater degree in aged mice. Gene expression correlated to influenza-induced body mass loss, although evidence did not support direct muscle infection. Overall, influenza leads to mobility impairments with induction of inflammatory and muscle degradation genes and downregulation of positive regulators of muscle. These effects are augmented and prolonged with aging, providing a molecular link between influenza infection, decreased resilience and increased risk of disability in the elderly.
Collapse
Affiliation(s)
- Jenna M Bartley
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT 06030, USA.,Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Sarah J Pan
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Spencer R Keilich
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jacob W Hopkins
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT 06030, USA.,Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Iman M Al-Naggar
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Laura Haynes
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT 06030, USA.,Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
38
|
|
39
|
Li F, Li X, Peng X, Sun L, Jia S, Wang P, Ma S, Zhao H, Yu Q, Huo H. Ginsenoside Rg1 prevents starvation-induced muscle protein degradation via regulation of AKT/mTOR/FoxO signaling in C2C12 myotubes. Exp Ther Med 2017; 14:1241-1247. [PMID: 28781621 DOI: 10.3892/etm.2017.4615] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/19/2016] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle atrophy is often caused by catabolic conditions including fasting, disuse, aging and chronic diseases, such as chronic obstructive pulmonary disease. Atrophy occurs when the protein degradation rate exceeds the rate of protein synthesis. Therefore, maintaining a balance between the synthesis and degradation of protein in muscle cells is a major way to prevent skeletal muscle atrophy. Ginsenoside Rg1 (Rg1) is a primary active ingredient in Panax ginseng, which is considered to be one of the most valuable herbs in traditional Chinese medicine. In the current study, Rg1 was observed to inhibit the expression of MuRF-1 and atrogin-1 in C2C12 muscle cells in a starvation model. Rg1 also activated the phosphorylation of mammalian target of rapamycin (mTOR), protein kinase B (AKT), and forkhead transcription factor O, subtypes 1 and 3a. This phosphorylation was inhibited by LY294002, a phosphatidylinositol 3-kinase inhibitor. These data suggest that Rg1 may participate in the regulation of the balance between protein synthesis and degradation, and that the function of Rg1 is associated with the AKT/mTOR/FoxO signaling pathway.
Collapse
Affiliation(s)
- Fengyu Li
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Xiaoxue Li
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Xuewei Peng
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Lili Sun
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Shengnan Jia
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Ping Wang
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Shuang Ma
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Hongyan Zhao
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Qingmiao Yu
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Hongliang Huo
- Laboratory of Molecular and Cellular Physiology, School of Life Science, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| |
Collapse
|
40
|
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: 6.7] [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
|
41
|
Mao Y, Kuo SW, Chen L, Heckman CJ, Jiang MC. The essential and downstream common proteins of amyotrophic lateral sclerosis: A protein-protein interaction network analysis. PLoS One 2017; 12:e0172246. [PMID: 28282387 PMCID: PMC5345759 DOI: 10.1371/journal.pone.0172246] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 02/01/2017] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastative neurodegenerative disease characterized by selective loss of motoneurons. While several breakthroughs have been made in identifying ALS genetic defects, the detailed molecular mechanisms are still unclear. These genetic defects involve in numerous biological processes, which converge to a common destiny: motoneuron degeneration. In addition, the common comorbid Frontotemporal Dementia (FTD) further complicates the investigation of ALS etiology. In this study, we aimed to explore the protein-protein interaction network built on known ALS-causative genes to identify essential proteins and common downstream proteins between classical ALS and ALS+FTD (classical ALS + ALS/FTD) groups. The results suggest that classical ALS and ALS+FTD share similar essential protein set (VCP, FUS, TDP-43 and hnRNPA1) but have distinctive functional enrichment profiles. Thus, disruptions to these essential proteins might cause motoneuron susceptible to cellular stresses and eventually vulnerable to proteinopathies. Moreover, we identified a common downstream protein, ubiquitin-C, extensively interconnected with ALS-causative proteins (22 out of 24) which was not linked to ALS previously. Our in silico approach provides the computational background for identifying ALS therapeutic targets, and points out the potential downstream common ground of ALS-causative mutations.
Collapse
Affiliation(s)
- Yimin Mao
- Applied Science Institute, Jiangxi University of Science and Technology, Jiangxi, China
- Department of Physiology, Northwestern University, Chicago, Illinois, United States of America
| | - Su-Wei Kuo
- Department of Physiology, Northwestern University, Chicago, Illinois, United States of America
| | - Le Chen
- Applied Science Institute, Jiangxi University of Science and Technology, Jiangxi, China
| | - C. J. Heckman
- Department of Physiology, Northwestern University, Chicago, Illinois, United States of America
- Department of Physical Medicine and Rehabilitation, Northwestern University, Chicago, Illinois, United States of America
- Department of Physical Therapy and Human Movement Sciences, Northwestern University, Chicago, Illinois, United States of America
| | - M. C. Jiang
- Department of Physiology, Northwestern University, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
42
|
Zha Y, Qian Q. Protein Nutrition and Malnutrition in CKD and ESRD. Nutrients 2017; 9:nu9030208. [PMID: 28264439 PMCID: PMC5372871 DOI: 10.3390/nu9030208] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/23/2017] [Indexed: 01/28/2023] Open
Abstract
Elevated protein catabolism and protein malnutrition are common in patients with chronic kidney disease (CKD) and end-stage renal disease (ESRD). The underlying etiology includes, but is not limited to, metabolic acidosis intestinal dysbiosis; systemic inflammation with activation of complements, endothelin-1 and renin-angiotensin-aldosterone (RAAS) axis; anabolic hormone resistance; energy expenditure elevation; and uremic toxin accumulation. All of these derangements can further worsen kidney function, leading to poor patient outcomes. Many of these CKD-related derangements can be prevented and substantially reversed, representing an area of great potential to improve CKD and ESRD care. This review integrates known information and recent advances in the area of protein nutrition and malnutrition in CKD and ESRD. Management recommendations are summarized. Thorough understanding the pathogenesis and etiology of protein malnutrition in CKD and ESRD patients will undoubtedly facilitate the design and development of more effective strategies to optimize protein nutrition and improve outcomes.
Collapse
Affiliation(s)
- Yan Zha
- Department of Nephrology, Guizhou Provincial People's Hospital, Guizhou 550002, China.
| | - Qi Qian
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
43
|
Bonifacio A, Sanvee GM, Brecht K, Kratschmar DV, Odermatt A, Bouitbir J, Krähenbühl S. IGF-1 prevents simvastatin-induced myotoxicity in C2C12 myotubes. Arch Toxicol 2016; 91:2223-2234. [PMID: 27734117 DOI: 10.1007/s00204-016-1871-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/06/2016] [Indexed: 12/18/2022]
Abstract
Statins are generally well tolerated, but treatment with these drugs may be associated with myopathy. The mechanisms of statin-associated myopathy are not completely understood. Statins inhibit AKT phosphorylation by an unclear mechanism, whereas insulin-like growth factor (IGF-1) activates the IGF-1/AKT signaling pathway and promotes muscle growth. The aims of the study were to investigate mechanisms of impaired AKT phosphorylation by simvastatin and to assess effects of IGF-1 on simvastatin-induced myotoxicity in C2C12 myotubes. C2C12 mouse myotubes were exposed to 10 μM simvastatin and/or 10 ng/mL IGF-1 for 18 h. Simvastatin inhibited the IGF-1/AKT signaling pathway, resulting in increased breakdown of myofibrillar proteins, impaired protein synthesis and increased apoptosis. Simvastatin inhibited AKT S473 phosphorylation, indicating reduced activity of mTORC2. In addition, simvastatin impaired stimulation of AKT T308 phosphorylation by IGF-1, indicating reduced activation of the IGF-1R/PI3K pathway by IGF-1. Nevertheless, simvastatin-induced myotoxicity could be at least partially prevented by IGF-1. The protective effects of IGF-1 were mediated by activation of the IGF-1R/AKT signaling cascade. Treatment with IGF-1 also suppressed muscle atrophy markers, restored protein synthesis and inhibited apoptosis. These results were confirmed by normalization of myotube morphology and protein content of C2C12 cells exposed to simvastatin and treated with IGF-1. In conclusion, impaired activity of AKT can be explained by reduced function of mTORC2 and of the IGF-1R/PI3K pathway. IGF-1 can prevent simvastatin-associated cytotoxicity and metabolic effects on C2C12 cells. The study gives insight into mechanisms of simvastatin-associated myotoxicity and provides potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Annalisa Bonifacio
- Division of Clinical Pharmacology and Toxicology, University Hospital, 4031, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Gerda M Sanvee
- Division of Clinical Pharmacology and Toxicology, University Hospital, 4031, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Karin Brecht
- Division of Clinical Pharmacology and Toxicology, University Hospital, 4031, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Denise V Kratschmar
- Division of Molecular and Systems Toxicology, University of Basel, Basel, Switzerland.,Swiss Centre of Applied Human Toxicology, Basel, Switzerland
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, University of Basel, Basel, Switzerland.,Swiss Centre of Applied Human Toxicology, Basel, Switzerland
| | - Jamal Bouitbir
- Division of Clinical Pharmacology and Toxicology, University Hospital, 4031, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland.,Swiss Centre of Applied Human Toxicology, Basel, Switzerland
| | - Stephan Krähenbühl
- Division of Clinical Pharmacology and Toxicology, University Hospital, 4031, Basel, Switzerland. .,Department of Biomedicine, University of Basel, Basel, Switzerland. .,Swiss Centre of Applied Human Toxicology, Basel, Switzerland.
| |
Collapse
|
44
|
Quan-Jun Y, Yan H, Yong-Long H, Li-Li W, Jie L, Jin-Lu H, Jin L, Peng-Guo C, Run G, Cheng G. Selumetinib Attenuates Skeletal Muscle Wasting in Murine Cachexia Model through ERK Inhibition and AKT Activation. Mol Cancer Ther 2016; 16:334-343. [PMID: 27599525 DOI: 10.1158/1535-7163.mct-16-0324] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/09/2016] [Accepted: 08/22/2016] [Indexed: 11/16/2022]
Abstract
Cancer cachexia is a multifactorial syndrome affecting the skeletal muscle. Previous clinical trials showed that treatment with MEK inhibitor selumetinib resulted in skeletal muscle anabolism. However, it is conflicting that MAPK/ERK pathway controls the mass of the skeletal muscle. The current study investigated the therapeutic effect and mechanisms of selumetinib in amelioration of cancer cachexia. The classical cancer cachexia model was established via transplantation of CT26 colon adenocarcinoma cells into BALB/c mice. The effect of selumetinib on body weight, tumor growth, skeletal muscle, food intake, serum proinflammatory cytokines, E3 ligases, and MEK/ERK-related pathways was analyzed. Two independent experiments showed that 30 mg/kg/d selumetinib prevented the loss of body weight in murine cachexia mice. Muscle wasting was attenuated and the expression of E3 ligases, MuRF1 and Fbx32, was inhibited following selumetinib treatment of the gastrocnemius muscle. Furthermore, selumetinib efficiently reduced tumor burden without influencing the cancer cell proliferation, cumulative food intake, and serum cytokines. These results indicated that the role of selumetinib in attenuating muscle wasting was independent of cancer burden. Detailed analysis of the mechanism revealed AKT and mTOR were activated, while ERK, FoxO3a, and GSK3β were inhibited in the selumetinib -treated cachexia group. These indicated that selumetinib effectively prevented skeletal muscle wasting in cancer cachexia model through ERK inhibition and AKT activation in gastrocnemius muscle via cross-inhibition. The study not only elucidated the mechanism of MEK/ERK inhibition in skeletal muscle anabolism, but also validated selumetinib therapy as an effective intervention against cancer cachexia. Mol Cancer Ther; 16(2); 334-43. ©2016 AACR.
Collapse
Affiliation(s)
- Yang Quan-Jun
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Huo Yan
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Han Yong-Long
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Wan Li-Li
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Li Jie
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Huang Jin-Lu
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Lu Jin
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Chen Peng-Guo
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Gan Run
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China
| | - Guo Cheng
- Department of Pharmacy, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, P.R. China.
| |
Collapse
|
45
|
Rahnert JA, Zheng B, Hudson MB, Woodworth-Hobbs ME, Price SR. Glucocorticoids Alter CRTC-CREB Signaling in Muscle Cells: Impact on PGC-1α Expression and Atrophy Markers. PLoS One 2016; 11:e0159181. [PMID: 27404111 PMCID: PMC4942104 DOI: 10.1371/journal.pone.0159181] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 06/28/2016] [Indexed: 01/21/2023] Open
Abstract
Muscle wasting associated with chronic diseases has been linked to decreased expression of PGC-1α and overexpression of PGC-1α counters muscle loss. CREB, in conjunction with the CREB-regulated transcription coactivator (CRTC2), is a positive modulator of PGC-1α transcription. We previously reported that PGC-1α expression is decreased in skeletal muscle of diabetic rats despite a high level of CREB phosphorylation (i.e., activation), suggesting that CRTC2-CREB signaling may be dysregulated. In this study, the relationship between CREB/CRTC signaling and PGC-1α expression was examined in L6 myotubes treated with dexamethasone (Dex, 48h) to induce atrophy. Dex decreased PGC-1α mRNA and protein as well as the levels of CRTC1 and CRTC2 in the nucleus. Dex also altered the nuclear levels of two known regulators of CRTC2 localization; the amount of calcinuerin catalytic A subunit (CnA) was decreased whereas SIK was increased. To assess PGC-1α transcription, muscle cells were transfected with a PGC-1α luciferase reporter plasmid (PGC-1α-Luc). Dex suppressed PGC-1α luciferase activity while both isobutylmethylxanthine (IBMX) and over-expression of CRTC1 or CRTC2 increased PGC-1α-Luc activity. Mutation of the CRE binding site from PGC-1α-Luc reporter attenuated the responses to both IBMX and the CRTC proteins. Consistent with the reporter gene results, overexpression of CRTC2 produced an increase in CRTC2 in the nucleus and in PGC-1α mRNA and PGC-1α protein. Overexpression of CRTC2 was not sufficient to prevent the decrease in PGC-1α mRNA or protein by Dex. In summary, these data suggest that attenuated CREB/CRTC signaling contributes to the decrease in PGC-1α expression during atrophy.
Collapse
Affiliation(s)
- Jill A. Rahnert
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Bin Zheng
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Matthew B. Hudson
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Myra E. Woodworth-Hobbs
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - S. Russ Price
- Renal Division, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States of America
- * E-mail:
| |
Collapse
|
46
|
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: 2.8] [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
|
47
|
Kuo T, Liu PH, Chen TC, Lee RA, New J, Zhang D, Lei C, Chau A, Tang Y, Cheung E, Wang JC. Transcriptional regulation of FoxO3 gene by glucocorticoids in murine myotubes. Am J Physiol Endocrinol Metab 2016; 310:E572-85. [PMID: 26758684 PMCID: PMC4824139 DOI: 10.1152/ajpendo.00214.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 01/06/2016] [Indexed: 01/08/2023]
Abstract
Glucocorticoids and FoxO3 exert similar metabolic effects in skeletal muscle. FoxO3 gene expression was increased by dexamethasone (Dex), a synthetic glucocorticoid, both in vitro and in vivo. In C2C12 myotubes the increased expression is due to, at least in part, the elevated rate of FoxO3 gene transcription. In the mouse FoxO3 gene, we identified three glucocorticoid receptor (GR) binding regions (GBRs): one being upstream of the transcription start site, -17kbGBR; and two in introns, +45kbGBR and +71kbGBR. Together, these three GBRs contain four 15-bp glucocorticoid response elements (GREs). Micrococcal nuclease (MNase) assay revealed that Dex treatment increased the sensitivity to MNase in the GRE of +45kbGBR and +71kbGBR upon 30- and 60-min Dex treatment, respectively. Conversely, Dex treatment did not affect the chromatin structure near the -17kbGBR, in which the GRE is located in the linker region. Dex treatment also increased histone H3 and/or H4 acetylation in genomic regions near all three GBRs. Moreover, using chromatin conformation capture (3C) assay, we showed that Dex treatment increased the interaction between the -17kbGBR and two genomic regions: one located around +500 bp and the other around +73 kb. Finally, the transcriptional coregulator p300 was recruited to all three GBRs upon Dex treatment. The reduction of p300 expression decreased FoxO3 gene expression and Dex-stimulated interaction between distinct genomic regions of FoxO3 gene identified by 3C. Overall, our results demonstrate that glucocorticoids activated FoxO3 gene transcription through multiple GREs by chromatin structural change and DNA looping.
Collapse
Affiliation(s)
- Taiyi Kuo
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, California; and Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Patty H Liu
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Tzu-Chieh Chen
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Rebecca A Lee
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, California; and Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Jenny New
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Danyun Zhang
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Cassandra Lei
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Andy Chau
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Yicheng Tang
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Edna Cheung
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| | - Jen-Chywan Wang
- Endocrinology Graduate Program, University of California Berkeley, Berkeley, California; and Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California
| |
Collapse
|
48
|
Furukawa K, Kikusato M, Kamizono T, Toyomizu M. Time-course changes in muscle protein degradation in heat-stressed chickens: Possible involvement of corticosterone and mitochondrial reactive oxygen species generation in induction of the ubiquitin-proteasome system. Gen Comp Endocrinol 2016; 228:105-110. [PMID: 26883687 DOI: 10.1016/j.ygcen.2016.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 02/04/2016] [Accepted: 02/07/2016] [Indexed: 12/22/2022]
Abstract
Heat stress (HS) induces muscle protein degradation as well as production of mitochondrial reactive oxygen species (ROS). In the present study, to improve our understanding of how protein degradation is induced by HS treatment in birds, a time course analysis of changes in the circulating levels of glucocorticoid and N(τ)-methylhistidine, muscle proteolysis-related gene expression, and mitochondrial ROS generation, was conducted. At 25 days of age, chickens were exposed to HS conditions (33 °C) for 0, 0.5, 1 or 3 days. While no alteration in plasma N(τ)-methylhistidine concentration relative to that of the control group was observed in the 0.5 day HS group, the concentration was significantly higher in the 3-d HS treatment group. Plasma corticosterone concentrations increased in response to 0.5-d HS treatment, but subsequently returned to near-normal values. HS treatment for 0.5 days did not change the levels of μ-calpain, cathepsin B, or proteasome C2 subunit mRNA, but increased the levels of mRNA encoding atrogin-1 (P<0.05) and its transcription factor, forkhead box O3 (P=0.09). Under these hyperthermic conditions, mitochondrial superoxide production was significantly increased than that of thermoneutral control. Here, we show that HS-induced muscle protein degradation may be due to the activation of ubiquitination by atrogin-1, and that this process may involve mitochondrial ROS production as well as corticosterone secretion.
Collapse
Affiliation(s)
- Kyohei Furukawa
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori-Amamiyamachi, Aoba-ku, Miyagi, Sendai 981-8555, Japan
| | - Motoi Kikusato
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori-Amamiyamachi, Aoba-ku, Miyagi, Sendai 981-8555, Japan.
| | - Tomomi Kamizono
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori-Amamiyamachi, Aoba-ku, Miyagi, Sendai 981-8555, Japan
| | - Masaaki Toyomizu
- Animal Nutrition, Life Sciences, Graduate School of Agricultural Science, Tohoku University, 1-1 Tsutsumidori-Amamiyamachi, Aoba-ku, Miyagi, Sendai 981-8555, Japan
| |
Collapse
|
49
|
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: 2.6] [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
|
50
|
Recent advances in the pathophysiology and management of protein-energy wasting in chronic kidney disease. RENAL REPLACEMENT THERAPY 2016. [DOI: 10.1186/s41100-016-0015-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|