1
|
Ji LL. Nuclear factor κB signaling revisited: Its role in skeletal muscle and exercise. Free Radic Biol Med 2025; 232:158-170. [PMID: 40010515 DOI: 10.1016/j.freeradbiomed.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
Nuclear factor (NF) κB as a redox sensitive, anti-apoptotic and pro-inflammatory signaling molecule has been studied extensively for more than three decades. Its role in inducing antioxidant enzymes, defending against extracellular and intracellular stress and maintaining redox homeostasis in skeletal muscle has also been recognized. New research continues to explore the polytropic nature of NFκB in cellular function, especially its crosstalk with other important signaling pathways. Understanding of the broad impact of these functions has significant implications in health and disease of skeletal muscle as an organ designed for contraction and mobility. Two important aspects of muscle wellbeing, i.e., disease and aging, are not discussed in this review. This review will provide an update on the new findings related to NFκB involvement in multiple signaling pathways and refresh our knowledge of its activation in skeletal muscle with a special reference to physical exercise.
Collapse
Affiliation(s)
- Li Li Ji
- The Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota Twin Cities, USA.
| |
Collapse
|
2
|
Noguchi R, Yanagihara K, Iino Y, Komatsu T, Kubo T, Ono T, Osaki J, Adachi Y, Iwata S, Shiota Y, Seyama T, Kondo T. Establishment and characterization of novel cancer cachexia-inducing cell line, Aku60GC, of scirrhous gastric cancer. Hum Cell 2025; 38:82. [PMID: 40178664 DOI: 10.1007/s13577-025-01208-9] [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: 12/29/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
Cancer cachexia is a pathological state characterized by severe weight loss, skeletal muscle depletion, and adipose tissue reduction. Cancer cachexia is observed in gastric cancer (GC) with a higher incidence over 80%. Approximately 80% patients with advanced GC including scirrhous gastric cancer (SGC), which has the worst prognosis among all GC, are affected with cachexia. The exact pathophysiology in SGC cancer cachexia remains elusive, and therapeutic approaches for the cancer cachexia have not been established. Patient-derived cancer cachexia models are promising for elucidating the underlying mechanisms of disease progression and developing novel treatments, none of which originate from SGC. Therefore, we established a novel cancer cachexia-inducing cell line, designated Aku60GC, through stepwise selection of a patient-derived SGC cell line, HSC-60. Subcutaneous implantation of the Aku60GC cells into nude mice resulted in weight loss, muscle atrophy, and adipose tissue depletion with high reproducibility, accompanied by elevation of the circulating cytokines IL-8 and IL-18. Compared to parental HSC-60 cells, Aku60GC cells exhibited additional genomic changes, such as AKT2 and CCNE1 gains, a somatic mutation of RUNX1, and accelerated growth. Thus, our results demonstrate that the Aku60GC cell line is a valuable resource for research on cancer cachexia in SGC.
Collapse
Affiliation(s)
- Rei Noguchi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kazuyoshi Yanagihara
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
- Department of Life Sciences, Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima-shi, Hiroshima, 731-0153, Japan.
- Biospecimen Laboratories, Inc., 1-5-10-105 Nakamagome, Ohta-ku, Tokyo, 143-0027, Japan.
| | - Yuki Iino
- Exploratory Oncology and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Teruo Komatsu
- Exploratory Oncology and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Takanori Kubo
- Department of Life Sciences, Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima-shi, Hiroshima, 731-0153, Japan
| | - Takuya Ono
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Julia Osaki
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yuki Adachi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shuhei Iwata
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yomogi Shiota
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Toshio Seyama
- Department of Life Sciences, Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima-shi, Hiroshima, 731-0153, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
3
|
Cheon YH, Lee CH, Chung CH, Kim JY, Lee MS. Vigeo Promotes Myotube Differentiation and Protects Dexamethasone-Induced Skeletal Muscle Atrophy via Regulating the Protein Degradation, AKT/mTOR, and AMPK/Sirt-1/PGC1α Signaling Pathway In Vitro and In Vivo. Nutrients 2024; 16:2687. [PMID: 39203823 PMCID: PMC11357481 DOI: 10.3390/nu16162687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Sarcopenia, a condition caused by an imbalance between muscle growth and loss, can severely affect the quality of life of elderly patients with metabolic, inflammatory, and cancer diseases. Vigeo, a nuruk-fermented extract of three plants (Eleutherococcus senticosus Maxim (ESM), Achyranthes japonica (Miq.) Nakai (AJN), and Atractylodes japonica Koidzumi (AJK)) has been reported to have anti-osteoporotic effects. However, evidence of the effects of Vigeo on muscle atrophy is not available. Here, in the in vivo model of dexamethasone (Dex)-induced muscle atrophy, Vigeo treatment significantly reversed Dex-induced decreases in calf muscle volume, gastrocnemius (GA) muscle weight, and histological cross-section area. In addition, in mRNA and protein analyses isolated from GA muscle, we observed that Vigeo significantly protected against Dex-induced mouse muscle atrophy by inhibiting protein degradation regulated by atrogin and MuRF-1. Moreover, we demonstrated that Vigeo significantly promoted C2C12 cell line differentiation, as evidenced by the increased width and length of myotubes, and the increased number of fused myotubes with three or more nuclei. Vigeo alleviated the formation of myotubes compared to the control group. Vigeo also significantly increased the mRNA and protein expression of myosin heavy chain (MyHC), MyoD, and myogenin compared to that in the control. Vigeo treatment significantly reduced the mRNA and protein expression of muscle degradation markers atrogin-1 and muscle RING Finger 1 (MuRF-1) in the C2C12 cell line in vitro. Vigeo also activated the AMP-activated protein kinase (AMPK)/silent information regulator 1 (Sirt-1)/peroxisome proliferator-activated receptor-γ co-activator-1α (PGC1α) mitochondrial biogenesis pathway and the Akt/mTOR protein synthesis signaling pathway in Dex-induced myotube atrophy. These findings suggest that Vigeo may have protective effects against Dex-induced muscle atrophy. Therefore, we propose Vigeo as a supplement or potential therapeutic agent to prevent or treat sarcopenia accompanied by muscle atrophy and degeneration.
Collapse
Affiliation(s)
- Yoon-Hee Cheon
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (Y.-H.C.); (C.-H.L.); (C.-H.C.)
| | - Chang-Hoon Lee
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (Y.-H.C.); (C.-H.L.); (C.-H.C.)
- Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan 54538, Republic of Korea
| | - Chong-Hyuk Chung
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (Y.-H.C.); (C.-H.L.); (C.-H.C.)
- Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan 54538, Republic of Korea
| | - Ju-Young Kim
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (Y.-H.C.); (C.-H.L.); (C.-H.C.)
| | - Myeung-Su Lee
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Republic of Korea; (Y.-H.C.); (C.-H.L.); (C.-H.C.)
- Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan 54538, Republic of Korea
| |
Collapse
|
4
|
Carrara E, Soliveri L, Poloni S, Bozzetto M, Campiglio CE. Effects of high-frequency mechanical stimuli on flow related vascular cell biology. Int J Artif Organs 2024; 47:590-601. [PMID: 39166431 PMCID: PMC11487902 DOI: 10.1177/03913988241268105] [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: 04/30/2024] [Accepted: 07/12/2024] [Indexed: 08/23/2024]
Abstract
Mechanical forces related to blood pressure and flow patterns play a crucial role in vascular homeostasis. Perturbations in vascular stresses and strain resulting from changes in hemodynamic may occur in pathological conditions, leading to vascular dysfunction as well as in vascular prosthesis, arteriovenous shunt for hemodialysis and in mechanical circulation support. Turbulent-like blood flows can induce high-frequency vibrations of the vessel wall, and this stimulus has recently gained attention as potential contributors to vascular pathologies, such as development of intimal hyperplasia in arteriovenous fistula for hemodialysis. However, the biological response of vascular cells to this stimulus remains incompletely understood. This review provides an analysis of the existing literature concerning the impact of high-frequency stimuli on vascular cell morphology, function, and gene expression. Morphological and functional investigations reveal that vascular cells stimulated at frequencies higher than the normal heart rate exhibit alterations in cell shape, alignment, and proliferation, potentially leading to vessel remodeling. Furthermore, vibrations modulate endothelial and smooth muscle cells gene expression, affecting pathways related to inflammation, oxidative stress, and muscle hypertrophy. Understanding the effects of high-frequency vibrations on vascular cells is essential for unraveling the mechanisms underlying vascular diseases and identifying potential therapeutic targets. Nevertheless, there are still gaps in our understanding of the molecular pathways governing these cellular responses. Further research is necessary to elucidate these mechanisms and their therapeutic implications for vascular diseases.
Collapse
Affiliation(s)
- Elena Carrara
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Luca Soliveri
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Sofia Poloni
- Department of Engineering and Applied Sciences, University of Bergamo, Dalmine, Italy
| | - Michela Bozzetto
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Chiara Emma Campiglio
- Department of Management, Information and Production Engineering, University of Bergamo, Dalmine, Italy
| |
Collapse
|
5
|
Kim MJ, Lee JM, Min K, Choi YS. Xenogeneic transplantation of mitochondria induces muscle regeneration in an in vivo rat model of dexamethasone-induced atrophy. J Muscle Res Cell Motil 2024; 45:53-68. [PMID: 36802005 DOI: 10.1007/s10974-023-09643-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 01/16/2023] [Indexed: 02/21/2023]
Abstract
Muscle atrophy significantly impairs health and quality of life; however, there is still no cure. Recently, the possibility of regeneration in muscle atrophic cells was suggested through mitochondrial transfer. Therefore, we attempted to prove the efficacy of mitochondrial transplantation in animal models. To this end, we prepared intact mitochondria from umbilical cord-derived mesenchymal stem cells maintaining their membrane potential. To examine the efficacy of mitochondrial transplantation on muscle regeneration, we measured muscle mass, cross-sectional area of muscle fiber, and changes in muscle-specific protein. In addition, changes in the signaling mechanisms related to muscle atrophy were evaluated. As a result, in mitochondrial transplantation, the muscle mass increased by 1.5-fold and the lactate concentration decreased by 2.5-fold at 1 week in dexamethasone-induced atrophic muscles. In addition, a 2.3-fold increase in the expression of desmin protein, a muscle regeneration marker, showed a significant recovery in MT 5 µg group. Importantly, the muscle-specific ubiquitin E3-ligases MAFbx and MuRF-1 were significantly decreased through AMPK-mediated Akt-FoxO signaling pathway by mitochondrial transplantation compared with the saline group, reaching a level similar to that in the control. Based on these results, mitochondrial transplantation may have therapeutic applications in the treatment of atrophic muscle disorders.
Collapse
Affiliation(s)
- Mi Jin Kim
- Department of Biotechnology, CHA University, 13488, Seongnam, Korea
| | - Ji Min Lee
- Department of Biotechnology, CHA University, 13488, Seongnam, Korea
| | - Kyunghoon Min
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University School of Medicine, 13496, Seongnam, Korea
| | - Yong-Soo Choi
- Department of Biotechnology, CHA University, 13488, Seongnam, Korea.
| |
Collapse
|
6
|
Han P, Yuan C, Chen X, Hu Y, Hu X, Xu Z, Guo Q. Metabolic signatures and potential biomarkers of sarcopenia in suburb-dwelling older Chinese: based on untargeted GC-MS and LC-MS. Skelet Muscle 2024; 14:4. [PMID: 38454497 PMCID: PMC10921582 DOI: 10.1186/s13395-024-00337-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Untargeted metabolomics can be used to expand our understanding of the pathogenesis of sarcopenia. However, the metabolic signatures of sarcopenia patients have not been thoroughly investigated. Herein, we explored metabolites associated with sarcopenia by untargeted gas chromatography (GC)/liquid chromatography (LC)-mass spectrometry (MS) and identified possible diagnostic markers. METHODS Forty-eight elderly subjects with sarcopenia were age and sex matched with 48 elderly subjects without sarcopenia. We first used untargeted GC/LC-MS to analyze the plasma of these participants and then combined it with a large number of multivariate statistical analyses to analyze the data. Finally, based on a multidimensional analysis of the metabolites, the most critical metabolites were considered to be biomarkers of sarcopenia. RESULTS According to variable importance in the project (VIP > 1) and the p-value of t-test (p < 0.05), a total of 55 metabolites by GC-MS and 85 metabolites by LC-MS were identified between sarcopenia subjects and normal controls, and these were mostly lipids and lipid-like molecules. Among the top 20 metabolites, seven phosphatidylcholines, seven lysophosphatidylcholines (LysoPCs), phosphatidylinositol, sphingomyelin, palmitamide, L-2-amino-3-oxobutanoic acid, and palmitic acid were downregulated in the sarcopenia group; only ethylamine was upregulated. Among that, three metabolites of LysoPC(17:0), L-2-amino-3-oxobutanoic acid, and palmitic acid showed very good prediction capacity with AUCs of 0.887 (95% CI = 0.817-0.957), 0.836 (95% CI = 0.751-0.921), and 0.805 (95% CI = 0.717-0.893), respectively. CONCLUSIONS These findings show that metabonomic analysis has great potential to be applied to sarcopenia. The identified metabolites could be potential biomarkers and could be used to study sarcopenia pathomechanisms.
Collapse
Affiliation(s)
- Peipei Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
- Jiangwan Hospital of Shanghai Hongkou District, Shanghai University of Medicine and Health Science Affiliated First Rehabilitation Hospital, Shanghai, China
| | - Chunhua Yuan
- Comprehensive Surgical Rehabilitation Ward, Shanghai Health Rehabilitation Hospital, Shanghai, China
| | - Xiaoyu Chen
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
| | - Yuanqing Hu
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
| | - Xiaodan Hu
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
| | - Zhangtao Xu
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China
| | - Qi Guo
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China.
- College of Rehabilitation Sciences, Pudong New Area, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai, 201318, China.
| |
Collapse
|
7
|
Petry ÉR, Dresch DDF, Carvalho C, Medeiros PC, Rosa TG, de Oliveira CM, Martins LAM, Guma FCR, Marroni NP, Wannmacher CMD. Oral glutamine supplementation relieves muscle loss in immobilized rats, altering p38MAPK and FOXO3a signaling pathways. Nutrition 2024; 118:112273. [PMID: 38096603 DOI: 10.1016/j.nut.2023.112273] [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: 08/22/2023] [Revised: 10/05/2023] [Accepted: 10/22/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Skeletal muscle synthesizes, stores, and releases body L-glutamine (GLN). Muscle atrophy due to disabling diseases triggers the activation of proteolytic and pro-apoptotic cell signaling, thus impairing the body's capacity to manage GLN content. This situation has a poor therapeutic prognosis. OBJECTIVE Evaluating if oral GLN supplementation can attenuate muscle wasting mediated by elevated plasma cortisol and activation of caspase-3, p38MAPK, and FOXO3a signaling pathways in soleus and gastrocnemius muscles of rats submitted to 14-day bilateral hindlimbs immobilization. METHODS Animals were randomly distributed into six groups: non-immobilized rats (Control), control orally supplemented with GLN (1 g kg-1) in solution with L-alanine (ALA: 0.61 g kg-1; GLN+ALA), control orally supplemented with dipeptide L-alanyl-L-glutamine (DIP; 1.49 g kg-1), hindlimbs immobilized rats (IMOB), IMOB orally GLN+ALA supplemented (GLN+ALA-IMOB), and IMOB orally DIP supplemented (DIP-IMOB). Plasma and muscle GLN concentration, plasma cortisol level, muscle caspase-3 activity, muscle p38MAPK and FOXO3a protein content (total and phosphorylated forms), and muscle cross-sectional area (CSA) were measured. RESULTS Compared to controls, IMOB rats presented: a) increased plasma cortisol levels; b) decreased plasma and muscle GLN concentration; c) increased muscle caspase-3 activity; d) increased total and phosphorylated p38MAPK protein content; e) increased FOXO3a and decreased phosphorylated FOXO3a protein content; f) reduced muscle weight and CSA befitting to atrophy. Oral supplementation with GLN+ALA and DIP was able to significantly attenuate these effects. CONCLUSIONS These findings attest that oral GLN supplementation in GLN+ALA solution or DIP forms attenuates rats' skeletal muscle mass wasting caused by disuse-mediated muscle atrophy.
Collapse
Affiliation(s)
- Éder Ricardo Petry
- Department of Cellular and Molecular Physiology, College of Medicine, Penn State University, Hershey, Pennsylvania, USA; Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Diego de Freitas Dresch
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Clarice Carvalho
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Patricia Calçada Medeiros
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Tatiana Gomes Rosa
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Faculdades Integradas de Taquara (FACCAT), Taquara, Rio Grande do Sul, Brazil
| | - Cleverson Morais de Oliveira
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Leo Anderson Meira Martins
- Laboratory of Endocrine and Tumor Molecular Biology, Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul, Brazil; Post-Graduate Program in Biological Sciences: Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Fátima Costa Rodrigues Guma
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Norma Possas Marroni
- Post-Graduate Program in Biological Sciences: Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil; Post-Graduate Program in Medicine: Medical Sciences, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil; Laboratory of Pulmonological Sciences: Inflammation, Experimental Research Center, Clinical Hospital of Porto Alegre (HCPA), UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Clóvis Milton Duval Wannmacher
- Post-Graduate Program in Biological Sciences: Biochemistry, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Department of Biochemistry, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
8
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
9
|
Sudharma AA, Siginam S, Husain GM, Mullapudi SV, Ismail A. ATROPHIC REMODELING OF THE HEART DURING VITAMIN D DEFICIENCY AND INSUFFICIENCY IN A RAT MODEL. J Nutr Biochem 2023:109382. [PMID: 37209952 DOI: 10.1016/j.jnutbio.2023.109382] [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: 12/05/2022] [Revised: 05/01/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Vitamin D deficiency (VDD) is associated with skeletal muscle wasting and impaired cardiac function in humans and animals. However, the molecular events that cause cardiac dysfunction in VDD are poorly understood, and therefore, therapeutic approaches are limited. In the present study, we investigated the effects of VDD on heart function with an emphasis on signaling pathways that regulate anabolism/catabolism in cardiac muscle. A Vitamin D deficient or insufficient rat model was employed. Heart electrical activity was measured by electrocardiography. Gene expression was monitored by qPCR, while protein expression was assessed by western blotting. Catalytic activities of the proteasome, lysosomal cathepsin activity, and apoptotic caspases were measured by fluorimetry. Vitamin D insufficiency and deficiency led to cardiac arrhythmia, a decrease in heart weight, and an increase in apoptosis and interstitial fibrosis. Ex-vivo cultures of atria revealed an increase in total protein degradation and a decrease in de-novo protein synthesis. The catalytic activities of the major proteolytic systems: ubiquitin-proteasome system, autophagy-lysosome, and calpains were upregulated in the heart of VDD and insufficient rats. In contrast, the mTOR pathway that regulates protein synthesis was suppressed. These catabolic events were exacerbated by a decrease in the expression of myosin heavy chain and troponin genes, as well as decreased expression and activities of metabolic enzymes. These latter changes occurred despite the activation of the energy sensor, AMPK. Our results provide, compelling evidence for cardiac atrophy in Vitamin D deficient rats. Unlike the skeletal muscle, the heart responded to VDD by activating all three proteolytic systems.
Collapse
Affiliation(s)
| | | | - Gulam M Husain
- Department of Pharmacology, National Research Institute of Unani Medicine for Skin Disorders, Hyderabad, India
| | | | - Ayesha Ismail
- Department of Endocrinology, National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
10
|
Unveiling the Role of the Proton Gateway, Uncoupling Proteins (UCPs), in Cancer Cachexia. Cancers (Basel) 2023; 15:cancers15051407. [PMID: 36900198 PMCID: PMC10000250 DOI: 10.3390/cancers15051407] [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: 12/07/2022] [Revised: 01/30/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Uncoupling proteins (UCPs) are identified as carriers of proton ions between the mitochondrial inner membrane and the mitochondrial matrix. ATP is mainly generated through oxidative phosphorylation in mitochondria. The proton gradient is generated across the inner mitochondrial membrane and the mitochondrial matrix, which facilitates a smooth transfer of electrons across ETC complexes. Until now, it was thought that the role of UCPs was to break the electron transport chain and thereby inhibit the synthesis of ATP. UCPs allow protons to pass from the inner mitochondrial membrane to the mitochondrial matrix and decrease the proton gradient across the membrane, which results in decreased ATP synthesis and increased production of heat by mitochondria. In recent years, the role of UCPs in other physiological processes has been deciphered. In this review, we first highlighted the different types of UCPs and their precise location across the body. Second, we summarized the role of UCPs in different diseases, mainly metabolic disorders such as obesity and diabetes, cardiovascular complications, cancer, wasting syndrome, neurodegenerative diseases, and kidney complications. Based on our findings, we conclude that UCPs play a major role in maintaining energy homeostasis, mitochondrial functions, ROS production, and apoptosis. Finally, our findings reveal that mitochondrial uncoupling by UCPs may treat many diseases, and extensive clinical studies are required to meet the unmet need of certain diseases.
Collapse
|
11
|
Lipoxin A4 attenuated dexamethasone-induced muscle atrophy via activation of PGC-1α/Nrf2/TFAM pathway. J Physiol Biochem 2023; 79:107-115. [PMID: 36125698 PMCID: PMC9905194 DOI: 10.1007/s13105-022-00925-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 09/07/2022] [Indexed: 02/08/2023]
Abstract
Prolonged dexamethasone (DEX) administration causes skeletal muscle atrophy through induction of both oxidative stress and mitochondrial dysfunction. Lipoxin A4 (LXA4) is a recognized antioxidant but its effect against DEX-induced muscle atrophy has not been studied yet. This study aimed to assess the potential ameliorating effect of LXA4 on DEX-induced muscle atrophy and investigate the possible involvement of the mitochondrial dynamics pathway and the redox state in this effect. Forty male rats were divided into four groups; normal control, LXA4-treated, DEX-treated, and LXA4 plus DEX-treated. At the end of the experiment, LXA4 counteracted the effect of DEX on different parameters including muscle weight, muscle strength, serum creatine kinase activity, malondialdehyde and protein carbonyl contents, Na/K-ATPase and citrate synthase activities, mitochondrial transmembrane potential, mitochondrial transcription factor (TFAM), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and nuclear factor erythroid 2-related factor 2 (Nrf2). These findings signify the promising therapeutic effect of LXA4 against DEX-induced skeletal muscle atrophy and indicate the possible involvement of LXA4-induced mitochondrial activation in addition to its well-known antioxidant effects.
Collapse
|
12
|
Chen K, Gao P, Li Z, Dai A, Yang M, Chen S, Su J, Deng Z, Li L. Forkhead Box O Signaling Pathway in Skeletal Muscle Atrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1648-1657. [PMID: 36174679 DOI: 10.1016/j.ajpath.2022.09.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/01/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Skeletal muscle atrophy is the consequence of protein degradation exceeding protein synthesis because of disease, aging, and physical inactivity. Patients with skeletal muscle atrophy have decreased muscle mass and fiber cross-sectional area, and experience reduced survival quality and motor function. The forkhead box O (FOXO) signaling pathway plays an important role in the pathogenesis of skeletal muscle atrophy by regulating E3 ubiquitin ligases and some autophagy factors. However, the mechanism of FOXO signaling pathway leading to skeletal muscle atrophy is still unclear. The development of treatment strategies for skeletal muscle atrophy has been a thorny clinical problem. FOXO-targeted therapy to treat skeletal muscle atrophy is a promising approach, and an increasing number of relevant studies have been reported. This article reviews the mechanism and therapeutic targets of the FOXO signaling pathway mediating skeletal muscle atrophy, and provides ideas for the clinical treatment of this condition.
Collapse
Affiliation(s)
- Kun Chen
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Peng Gao
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Zongchao Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Aonan Dai
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Ming Yang
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Siyu Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Jingyue Su
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; School of Medicine, Guangxi University of Chinese Medicine, Nanning, China.
| | - Liangjun Li
- Department of Orthopaedics, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.
| |
Collapse
|
13
|
Lee JA, Lee SH, Shin MR, Park HJ, Roh SS. Gardeniae Fructus Extract Alleviates Dexamethasone-Induced Muscle Atrophy in Mice. J Med Food 2022; 25:882-891. [PMID: 36084316 DOI: 10.1089/jmf.2022.k.0038] [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: 11/13/2022] Open
Abstract
Muscle atrophy (MA) is a case in which protein degeneration occurs excessively due to an imbalance between protein synthesis and breakdown, and is characterized by decreased muscle mass and weakened muscle strength. Despite mounting concern about MA, the number of patients with MA is increasing every year. The aim of the present study was to assess the impact of Gardeniae Fructus (GF) hot water extract on dexamethasone (DEX)-induced MA in mice. C57BL/6N mice were grouped (n = 8) as follows: Normal mice (Normal), MA mice were treated with distilled water (Control), MA mice were treated with GF 100 mg/kg (GF100), MA mice were treated with GF 200 mg/kg (GF200). For 10 days, DEX (25 mg/kg body weight, i.p.) injection was used to induce MA, and GF was administered. GF treatment restored the muscle weight decreased due to MA, and in particular, the weights of EDL+TA and Sol were significantly increased in the GF200 group. Also, it was confirmed that the swimming time was improved in the GF200 group. In addition, the expression of NADPH oxidase related to oxidative stress was significantly reduced, and protective (insulin-like growth factor I/phosphoinositide 3-kinase/protein kinase B pathway) and catabolic (AMP-activated kinase [AMPK]/sirtuin 1 [SIRT1]/proliferator-activated receptor-gamma coactivator-1α (PGC-1α)-forkhead box O (FOXO) pathway) pathways were significantly modulated. These results demonstrate that GF regulates muscle protein synthesis and catabolic pathways, and in particular, it is judged to improve MA by regulating the proteolytic AMPK/SIRT1/PGC-1α-FOXO pathway.
Collapse
Affiliation(s)
- Jin A Lee
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| | - Se Hui Lee
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| | - Mi-Rae Shin
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| | - Hae-Jin Park
- DHU Bio Convergence Testing Center, Gyeongsan, Korea
| | - Seong-Soo Roh
- Department of Herbology, College of Korean Medicine, Daegu Haany University, Daegu, Korea
| |
Collapse
|
14
|
Elbialy ZI, Gamal S, Al-Hawary II, Shukry M, Salah AS, Aboshosha AA, Assar DH. Exploring the impacts of different fasting and refeeding regimes on Nile tilapia (Oreochromis niloticus L.): growth performance, histopathological study, and expression levels of some muscle growth-related genes. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:973-989. [PMID: 35781858 PMCID: PMC9385825 DOI: 10.1007/s10695-022-01094-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023]
Abstract
The current study investigated how different fasting and refeeding regimes would impact Nile tilapia growth performance, histopathological examination, and gene expression of myostatin, myogenin, GH, IGF-1, and NPYa. Nile tilapia fish (n = 120) were randomly allocated into four groups, including the control group fed on a basal diet for 6 weeks (F6), group A starved for 1 week and then refed for 5 weeks (S1F5), group B starved for 2 weeks and then refed for 4 weeks (S2F4), while group C starved for 4 weeks and then refed for 2 weeks (S4F2). Fasting provoked a decrease in body weight coincided with more extended starvation periods. Also, it induced muscle and liver histological alterations; the severity was correlated with the length of fasting periods. Gene expression levels of GH, MSTN, MYOG, and NPYa were significantly increased, while IGF1 was markedly depressed in fasted fish compared to the control group. Interestingly, refeeding after well-planned short fasting period (S1F5) modulated the histopathological alterations. To some extent, these changes were restored after refeeding. Restored IGF-I and opposing fasting expression profiles of the genes mentioned above thus recovered weights almost like the control group and achieved satisfactory growth compensation. Conversely, refeeding following more extended fasting periods failed to restore body weight. In conclusion, refeeding after fasting can induce a compensatory response. Still, the restoration capacity is dependent on the length of fasting and refeeding periods through exhibiting differential morphological structure and expressions pattern for muscle and growth-related genes.
Collapse
Affiliation(s)
- Zizy I. Elbialy
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Shrouk Gamal
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Ibrahim I. Al-Hawary
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Abdallah S. Salah
- Department of Aquaculture, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
- Institute of Aquaculture, Faculty of Natural Sciences, University of Stirling, Stirling, FK9 4LA UK
| | - Ali A. Aboshosha
- Department of Genetics, Faculty of Agriculture, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| | - Doaa H. Assar
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516 Egypt
| |
Collapse
|
15
|
Extracellular polysaccharides purified (Polycan) from Aureobasidium pullulans SM‑2001 improves pathophysiology of dystrophin-deficient mdx mice. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00245-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Background
Duchenne muscular dystrophy is a hereditary muscular disease involving degeneration (i.e. atrophy and loss of muscle fibres) of skeletal muscles, including the diaphragm, and progressively severe functional decline. A previous study shows Polycan, a type of β-glucan derived from the black yeast Aureobasidium pullulans (SM-2001), promotes osteogenicity and bone loss, and possesses anti-inflammatory activity to induce inflammatory cytokines in human immune and cancer cells.
Objective
In this study, we evaluated changes in exercise load behaviour measurements and changes in muscle-related physiological indicators following oral administration of Polycan in mdx mice, an experimental animal model of Duchenne muscular dystrophy.
Result
In mdx mice, Polycan prevented weight loss and thickness of skeletal muscle. In addition, by monitoring increases in running time of mice on treadmills and performing a grip strength test, we confirmed reduced muscle function was recovered to some extent after administering Polycan to mdx mice. In addition, we confirmed that Polycan significantly altered mRNA expression in a concentration-dependent manner, whereby myogenic transcription factors (MyoD, Myf5 and Myogenin) increased and FoxO3α, MuRF1 and Atrogin-1 decreased. We aimed to investigate the mechanism of action in Polycan on energy metabolism of p-AMPK, SIRT1 and PGC1α with apoptosis expression levels as factors related to signalling pathways. Expression ratios of cleaved-caspase-3/caspase-3 and Bax/Bcl-2 in the Polycan extract-administered group increased compared with the control group.
Conclusion
These results demonstrate that Polycan can improve and protect muscle atrophy by preventing apoptosis via pathway regulation related to myogenic transcription factors and energy metabolism in mdx mice.
Collapse
|
16
|
Nagendra AH, Najar MA, Bose B, Shenoy PS. High concentration of sodium fluoride in drinking water induce hypertrophy versus atrophy in mouse skeletal muscle via modulation of sarcomeric proteins. JOURNAL OF HAZARDOUS MATERIALS 2022; 432:128654. [PMID: 35286933 DOI: 10.1016/j.jhazmat.2022.128654] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 06/14/2023]
Abstract
Fluoride at high doses is a well-known toxic agent for the musculoskeletal system, primarily in bone and cartilage cells. Research on fluoride toxicity concerning particularly on the skeletal muscle is scanty. We hypothesized that during skeletal fluorosis, along with bone, muscle is also affected, so we have evaluated the effects of Sodium fluoride (NaF) on mouse skeletal muscles. Sodium fluoride (80 ppm) was administered to 5-week-old C57BL6 mice drinking water for 15 and 60 days, respectively. We carried out histology, primary culture, molecular and proteomic analysis of fluoride administered mouse skeletal muscles. Results indicated an increase in the muscle mass (hypertrophy) in vivo and myotubes ex vivo by activating the IGF1/PI3/Akt/mTOR signalling pathway due to short term NaF exposure. The long-term exposure of mice to NaF caused loss of muscle proteins leading to muscle atrophy due to activation of the ubiquitin-proteasome pathway. Differentially expressed proteins were characterized and mapped using a proteomic approach. Moreover, the factors responsible for protein synthesis and PI3/Akt/mTOR pathway were upregulated, leading to muscle hypertrophy during the short term NaF exposure. Long term exposure to NaF resulted in down-regulation of metabolic pathways. Elevated myostatin resulted in the up-regulation of the muscle-specific E3 ligases-MuRF1, promoting the ubiquitination and proteasome-mediated degradation of critical sarcomeric proteins.
Collapse
Affiliation(s)
- Apoorva H Nagendra
- Stem cells and Regenerative medicine centre, Yenepoya research centre, Yenepoya Deemed to be University, University Road, Derlakatte, Mangalore 575018, Karnataka, India
| | - Mohd Altaf Najar
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Bipasha Bose
- Stem cells and Regenerative medicine centre, Yenepoya research centre, Yenepoya Deemed to be University, University Road, Derlakatte, Mangalore 575018, Karnataka, India.
| | - P Sudheer Shenoy
- Stem cells and Regenerative medicine centre, Yenepoya research centre, Yenepoya Deemed to be University, University Road, Derlakatte, Mangalore 575018, Karnataka, India.
| |
Collapse
|
17
|
Shin JE, Jeon SH, Lee SJ, Choung SY. The Administration of Panax Ginseng Berry Extract Attenuates High-Fat-Diet-Induced Sarcopenic Obesity in C57BL/6 Mice. Nutrients 2022; 14:nu14091747. [PMID: 35565712 PMCID: PMC9099595 DOI: 10.3390/nu14091747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/14/2022] [Accepted: 04/20/2022] [Indexed: 01/05/2023] Open
Abstract
Sarcopenia and obesity are serious health problems that are highly related to several metabolic diseases. Sarcopenic obesity, a combined state of sarcopenia and obesity, results in higher risks of metabolic diseases and even mortality than sarcopenia or obesity alone. Therefore, the development of therapeutic agents for sarcopenic obesity is crucial. C57BL/6 mice were fed with a high-fat diet (HFD) for 9 weeks. Then, mice were administered with Panax ginseng berry extract (GBE) for an additional 4 weeks, with continuous HFD intake. GBE significantly decreased the food efficiency ratio, serum lipid and insulin levels, adipose tissue weights, and adipocyte size. It significantly increased the grip strength, muscle masses, and myofiber cross-sectional area. It deactivated the protein kinase C (PKC) theta and zeta, resulting in activation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, which is known to regulate muscle synthesis and degradation. Furthermore, it inhibited the production of inflammatory cytokines in the muscle tissue. GBE attenuated both obesity and sarcopenia. Thus, GBE is a potential agent to prevent or treat sarcopenic obesity.
Collapse
Affiliation(s)
- Ji-Eun Shin
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea;
| | - So-Hyun Jeon
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | | | - Se-Young Choung
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea;
- Department of Biomedical and Pharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
- Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Correspondence: ; Tel.: +82-2-961-9198; Fax: +82-2-961-0372
| |
Collapse
|
18
|
Xu M, Liu X, Bao P, Wang YJ, Lu J, Liu YJ. H2S Protects Against Immobilization-Induced Muscle Atrophy via Reducing Oxidative Stress and Inflammation. Front Physiol 2022; 13:844539. [PMID: 35464091 PMCID: PMC9019569 DOI: 10.3389/fphys.2022.844539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation and oxidative stress are major triggers of the imbalance between protein synthesis and degradation during the pathogenesis of immobilization-induced muscle atrophy. This study aimed to elucidate the effects of hydrogen sulfide (H2S), a gas transmitter with potent anti-inflammatory and antioxidant properties, on immobilization-induced muscle atrophy. Mice were allocated to control and immobilization (IM) groups, which were treated with slow (GYY4137) or rapid (NaHS) H2S releasing donors for 14 days. The results showed that both GYY4137 and NaHS treatment reduced the IM-induced muscle loss, and increased muscle mass. The IM-induced expressions of Muscle RING finger 1 (MuRF1) and atrogin-1, two muscle-specific E3 ubiquitin ligases, were decreased by administration of GYY4137 or NaHS. Both GYY4137 and NaHS treatments alleviated the IM-induced muscle fibrosis, as evidenced by decreases in collagen deposition and levels of tissue fibrosis biomarkers. Moreover, administration of GYY4137 or NaHS alleviated the IM-induced infiltration of CD45 + leukocytes, meanwhile inhibited the expressions of the pro-inflammatory biomarkers in skeletal muscles. It was found that administration of either GYY4137 or NaHS significantly attenuated immobilization-induced oxidative stress as indicated by decreased H2O2 levels and 8-hydroxy-2′-deoxyguanosine (8-OHdG) immunoreactivity, as well as increased total antioxidant capacity (T-AOC), nuclear factor erythroid-2-related factor 2 (NRF2) and NRF2 downstream anti-oxidant targets levels in skeletal muscles. Collectively, the present study demonstrated that treatment with either slow or rapid H2S releasing donors protected mice against immobilization-induced muscle fibrosis and atrophy. The beneficial effects of H2S on immobilization-induced skeletal muscle atrophy might be due to both the anti-inflammatory and anti-oxidant properties of H2S.
Collapse
Affiliation(s)
- Miaomiao Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xiaoguang Liu
- Guangzhou Sport University Sports and Health, Guangzhou Sport University, Guangzhou, China
| | - Peng Bao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yan Jie Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jianqiang Lu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- *Correspondence: Jianqiang Lu, ; Yu Jian Liu,
| | - Yu Jian Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- *Correspondence: Jianqiang Lu, ; Yu Jian Liu,
| |
Collapse
|
19
|
Diphlorethohydroxycarmalol Derived from Ishige okamurae Improves Behavioral and Physiological Responses of Muscle Atrophy Induced by Dexamethasone in an In-Vivo Model. Pharmaceutics 2022; 14:pharmaceutics14040719. [PMID: 35456553 PMCID: PMC9026865 DOI: 10.3390/pharmaceutics14040719] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023] Open
Abstract
Muscle atrophy refers to the loss of skeletal muscle mass, myofiber size, and related physical functions such as walking speed or grip strength caused by aging or a lack of physical activity due to injury or illness and can also be attributed to excessive exposure to corticosteroids. Ishige okamurae (IO) and its active component, diphlorethohydroxycarmalol (DPHC), have been known to improve glucose homeostasis by controlling the contraction of skeletal muscles. Based on this idea, we hypothesized that the effects of DPHC and IO extract on muscle metabolism are associated with their role in improving muscle physical function. This study assessed the effects of DPHC or IO extract on muscle behavioral responses with their metabolic properties in muscle atrophy induced by glucocorticoids and dexamethasone (DEX) in vivo. In addition to the improvement in muscle behavioral response by DPHC or IO extract, the loss of muscle fiber and the related metabolic properties by DEX exposure in the gastrocnemius and soleus of calf muscle was prevented. These findings suggest that IO extract and its active component DPHC can potentially prevent muscle atrophy caused by exposure to corticosteroids and could be used to treat reverse skeletal atrophy.
Collapse
|
20
|
Endurance exercise training under normal diet conditions activates skeletal muscle protein synthesis and inhibits protein degradation signaling except MuRF1. SPORT SCIENCES FOR HEALTH 2022. [DOI: 10.1007/s11332-021-00888-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
21
|
Brooks DS, Vishal K, Bawa S, Alder A, Geisbrecht ER. Integration of proteomic and genetic approaches to assess developmental muscle atrophy. J Exp Biol 2021; 224:272703. [PMID: 34647571 DOI: 10.1242/jeb.242698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 10/07/2021] [Indexed: 01/04/2023]
Abstract
Muscle atrophy, or a decline in muscle protein mass, is a significant problem in the aging population and in numerous disease states. Unraveling molecular signals that trigger and promote atrophy may lead to a better understanding of treatment options; however, there is no single cause of atrophy identified to date. To gain insight into this problem, we chose to investigate changes in protein profiles during muscle atrophy in Manduca sexta and Drosophila melanogaster. The use of insect models provides an interesting parallel to probe atrophic mechanisms as these organisms undergo a normal developmental atrophy process during the pupal transition stage. Leveraging the inherent advantages of each model organism, we first defined protein signature changes during M. sexta intersegmental muscle (ISM) atrophy and then used genetic approaches to confirm their functional importance in the D. melanogaster dorsal internal oblique muscles (DIOMs). Our data reveal an upregulation of proteasome and peptidase components and a general downregulation of proteins that regulate actin filament formation. Surprisingly, thick filament proteins that comprise the A-band are increased in abundance, providing support for the ordered destruction of myofibrillar components during developmental atrophy. We also uncovered the actin filament regulator ciboulot (Cib) as a novel regulator of muscle atrophy. These insights provide a framework towards a better understanding of global changes that occur during atrophy and may eventually lead to therapeutic targets.
Collapse
Affiliation(s)
- David S Brooks
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Kumar Vishal
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Simranjot Bawa
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Adrienne Alder
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| | - Erika R Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
22
|
Ryu B, Je JG, Jeon YJ, Yang HW. Zebrafish Model for Studying Dexamethasone-Induced Muscle Atrophy and Preventive Effect of Maca ( Lepidium meyenii). Cells 2021; 10:cells10112879. [PMID: 34831102 PMCID: PMC8616435 DOI: 10.3390/cells10112879] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/13/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023] Open
Abstract
Loss of myofibers during muscle atrophy affects functional capacity and quality of life. Dexamethasone, an inducer of rapid atrophy of skeletal myofibers, has been studied as a glucocorticoid receptor in muscle atrophy or motor neurodegeneration. In this study, we examined dexamethasone-induced muscle atrophy using zebrafish (Danio rerio), a vertebrate model, and assessed whether administration of Lepidium meyenii (maca) as a dietary supplement can prevent muscle atrophy. Changes in skeletal myofibers in zebrafish were evaluated after exposure to dexamethasone for different periods and at different concentrations. Under optimized conditions, zebrafish pre-fed with maca for 3 days were exposed to 0.01% dexamethasone for 1 h/day for 7 days. Thereafter, myofiber loss, damaged muscle contractile proteins, and abnormal exploratory behavior due to the structural and functional impairment of skeletal muscle associated with muscle atrophy were investigated using hematoxylin-eosin, immunofluorescence staining, and behavioral analyses. Our findings suggest that dexamethasone induces muscle atrophy in zebrafish, inhibiting exploratory behavior by inducing myofiber loss, inhibiting muscle contraction, and causing changes in endurance and velocity. Thus, the zebrafish model can be used to screen pharmaceutical agents and to study muscle atrophy. Furthermore, maca is a potential dietary supplement to prevent muscle atrophy, as it protects muscle fibers.
Collapse
Affiliation(s)
- Bomi Ryu
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (B.R.); (J.-G.J.)
- Healthy Seafood Research Center, Jeju National University, Jeju 63243, Korea
| | - Jun-Geon Je
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (B.R.); (J.-G.J.)
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (B.R.); (J.-G.J.)
- Healthy Seafood Research Center, Jeju National University, Jeju 63243, Korea
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
- Correspondence: (Y.-J.J.); (H.-W.Y.); Tel.: +82-64-754-3475 (Y.-J.J.)
| | - Hye-Won Yang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea; (B.R.); (J.-G.J.)
- Correspondence: (Y.-J.J.); (H.-W.Y.); Tel.: +82-64-754-3475 (Y.-J.J.)
| |
Collapse
|
23
|
Dieckol Reduces Muscle Atrophy by Modulating Angiotensin Type II Type 1 Receptor and NADPH Oxidase in Spontaneously Hypertensive Rats. Antioxidants (Basel) 2021; 10:antiox10101561. [PMID: 34679696 PMCID: PMC8533257 DOI: 10.3390/antiox10101561] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 12/17/2022] Open
Abstract
The renin-angiotensin system is involved in the development of hypertension and sarcopenia. Increased levels of angiotensin II (Ang II) lead to upregulation of Ang II type 1 receptor (AT1R), which results in increasing reactive oxygen species (ROS) by NAD(P)H oxidase (Nox). Increased ROS led to increased helper T17 (Th17) and decreased regulatory T (Treg) cells through HIF-1α. Increased Th17 secretes more IL-17, leading to increased NF-κB and muscle atrophy. We evaluated the effect of Ecklonia cava extracts (ECE) and dieckol (DK) on attenuating muscle atrophy by decreasing AT1R and NOX activity in spontaneous hypertensive rats (SHRs). The serum levels of Ang II and expression of AT1R in the muscle were higher in SHRs than in normotensive animals of Wistar-Kyoto rats (2.4 and 1.8 times higher than WKY, respectively). The expression of AT1R decreased by ECE or DK (0.62 and 0.84 times lower than SHR, respectively). In SHRs, the expression of Nox 1, 2, and 4 were increased (1.2-1.15 times higher than WKY) but were decreased by the administration of ECE (0.8-0.9 times lower than SHR) or DK (0.7-0.9 times lower than SHR). The Nox activity was increased in SHRs (2.3 times more than WKY) and it was decreased by ECE (0.9 times lower than SHRs) and DK (0.9 times lower than SHRs). The expression of HIF-1α, a marker of Th17 (RORγt), and cytokine secreted by Th17 (IL-17) was increased in SHRs and was decreased by ECE or DK. The marker of Treg (Foxp3) and cytokine secreted from Treg cells (IL-10) was decreased in SHRs and was increased by ECE or DK. The expression of NF-κB/IL-1β/TNF-α and MuRF-1/MAFbx/atrogin-1 was increased in SHRs and these were decreased by ECE or DK. The cross-sectional area of muscle fiber was decreased in SHRs (0.7 times lower than WKY) and was increased by ECE (1.3 times greater than SHR) or DK (1.5 times greater than SHR). In conclusion, ECE or DK leads to a decreased expression of AT1R and Nox activity which modulates Th17/Treg balance and consequently, decreased muscle atrophy.
Collapse
|
24
|
Memme JM, Slavin M, Moradi N, Hood DA. Mitochondrial Bioenergetics and Turnover during Chronic Muscle Disuse. Int J Mol Sci 2021; 22:ijms22105179. [PMID: 34068411 PMCID: PMC8153634 DOI: 10.3390/ijms22105179] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
Periods of muscle disuse promote marked mitochondrial alterations that contribute to the impaired metabolic health and degree of atrophy in the muscle. Thus, understanding the molecular underpinnings of muscle mitochondrial decline with prolonged inactivity is of considerable interest. There are translational applications to patients subjected to limb immobilization following injury, illness-induced bed rest, neuropathies, and even microgravity. Studies in these patients, as well as on various pre-clinical rodent models have elucidated the pathways involved in mitochondrial quality control, such as mitochondrial biogenesis, mitophagy, fission and fusion, and the corresponding mitochondrial derangements that underlie the muscle atrophy that ensues from inactivity. Defective organelles display altered respiratory function concurrent with increased accumulation of reactive oxygen species, which exacerbate myofiber atrophy via degradative pathways. The preservation of muscle quality and function is critical for maintaining mobility throughout the lifespan, and for the prevention of inactivity-related diseases. Exercise training is effective in preserving muscle mass by promoting favourable mitochondrial adaptations that offset the mitochondrial dysfunction, which contributes to the declines in muscle and whole-body metabolic health. This highlights the need for further investigation of the mechanisms in which mitochondria contribute to disuse-induced atrophy, as well as the specific molecular targets that can be exploited therapeutically.
Collapse
Affiliation(s)
| | | | | | - David A. Hood
- Correspondence: ; Tel.: +1-(416)-736-2100 (ext. 66640)
| |
Collapse
|
25
|
Lee MK, Choi YH, Nam TJ. Pyropia yezoensis protein protects against TNF‑α‑induced myotube atrophy in C2C12 myotubes via the NF‑κB signaling pathway. Mol Med Rep 2021; 24:486. [PMID: 33955507 PMCID: PMC8127067 DOI: 10.3892/mmr.2021.12125] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 04/12/2021] [Indexed: 12/25/2022] Open
Abstract
The protein extracted from red algae Pyropia yezoensis has various biological activities, including anti-inflammatory, anticancer, antioxidant, and antiobesity properties. However, the effects of P. yezoensis protein (PYCP) on tumor necrosis factor-α (TNF-α)-induced muscle atrophy are unknown. Therefore, the present study investigated the protective effects and related mechanisms of PYCP against TNF-α-induced myotube atrophy in C2C12 myotubes. Treatment with TNF-α (20 ng/ml) for 48 h significantly reduced myotube viability and diameter and increased intracellular reactive oxygen species levels; these effects were significantly reversed in a dose-dependent manner following treatment with 25–100 µg/ml PYCP. PYCP inhibited the expression of TNF receptor-1 in TNF-α-induced myotubes. In addition, PYCP markedly downregulated the nuclear translocation of nuclear factor-κB (NF-κB) by inhibiting the phosphorylation of inhibitor of κB. Furthermore, PYCP treatment suppressed 20S proteasome activity, IL-6 production, and the expression of the E3 ubiquitin ligases, atrogin-1/muscle atrophy F-box and muscle RING-finger protein-1. Finally, PYCP treatment increased the protein expression levels of myoblast determination protein 1 and myogenin in TNF-α-induced myotubes. The present findings indicate that PYCP may protect against TNF-α-induced myotube atrophy by inhibiting the proinflammatory NF-κB pathway.
Collapse
Affiliation(s)
- Min-Kyeong Lee
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Youn Hee Choi
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| | - Taek-Jeong Nam
- Institute of Fisheries Sciences, Pukyong National University, Busan 46041, Republic of Korea
| |
Collapse
|
26
|
Tebbe AW, Hanson J, Weiss WP. Effects of metabolizable protein concentration, amino acid profile, and fiber source on the messenger RNA expression of skeletal muscle in peripartum dairy cows. J Dairy Sci 2021; 104:7888-7901. [PMID: 33814155 DOI: 10.3168/jds.2021-20176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/25/2021] [Indexed: 01/11/2023]
Abstract
After parturition, dairy cows mobilize AA from skeletal muscle to meet metabolizable protein (MP) requirements. High mobilization may compromise cow health and longer-term milk production. Postpartum diets with higher MP concentrations, improved AA profiles, or MP increased at the expense of forages rather than nonforage fiber sources may attenuate muscle catabolism; however, the molecular mechanisms responsible need investigation. We evaluated mRNA expression in the longissimus dorsi of cows fed postpartum diets differing in MP concentration, AA profile, and fiber source. From 0 to 25 d after parturition, 40 multiparous cows received the following diets: (1) 13% deficient in MP (D-MP), (2) adequate in MP using primarily soy protein (A-MP), (3) adequate in MP using blends of proteins and individual AA to improve the AA profile (Blend), or (4) similar to Blend except additional protein replaced forage (Blend-fNDF). Biopsies were taken approximately -5, 7, and 25 d relative to parturition. Greater dietary MP concentration (D-MP vs. A-MP and Blend) decreased expression of genes related to protein synthesis (MTOR, RPS6KB1) and degradation (FOXO1), inflammation (IFNG, TLR4), and endoplasmic reticulum (ER) stress (HSPA5, DDIT) and increased genes associated with lipogenesis (PPARG) and glucose oxidation (LDH, MB). In Blend versus A-MP (i.e., effect of AA profile), expression related to apoptosis (CASP8) and inflammation (TNFA) decreased and genes associated with cell cycle progression (E2F1) and fast-twitch glycolytic muscle fiber type (MYH4) increased. Less forage (Blend-fNDF vs. Blend) decreased genes associated with lipogenesis (PPARG, ACACA) and ER stress (BCL2, DDIT3, EIF2AK3, PPP1R15A) and increased genes associated with inflammation (TNF), inhibition of myogenesis (MSTN), and autophagy (PEBP1). In summary and based on mRNA expression, increasing MP supply may attenuate muscle turnover and ER stress. However, an unbalanced AA supply reduced cell cycle progression and protein synthesis. Lower energy supplies may reduce cell growth and cause autophagy.
Collapse
Affiliation(s)
- Alexander W Tebbe
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster 44691
| | - Juliette Hanson
- Food Animal Health Research Program, The Ohio State University, Wooster 44691
| | - William P Weiss
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster 44691.
| |
Collapse
|
27
|
Jang YC, Rodriguez K, Lustgarten MS, Muller FL, Bhattacharya A, Pierce A, Choi JJ, Lee NH, Chaudhuri A, Richardson AG, Van Remmen H. Superoxide-mediated oxidative stress accelerates skeletal muscle atrophy by synchronous activation of proteolytic systems. GeroScience 2020; 42:1579-1591. [PMID: 32451848 PMCID: PMC7732940 DOI: 10.1007/s11357-020-00200-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/06/2020] [Indexed: 12/25/2022] Open
Abstract
The maintenance of skeletal muscle mass depends on the overall balance between the rates of protein synthesis and degradation. Thus, age-related muscle atrophy and function, commonly known as sarcopenia, may result from decreased protein synthesis, increased proteolysis, or simultaneous changes in both processes governed by complex multifactorial mechanisms. Growing evidence implicates oxidative stress and reactive oxygen species (ROS) as an essential regulator of proteolysis. Our previous studies have shown that genetic deletion of CuZn superoxide dismutase (CuZnSOD, Sod1) in mice leads to elevated oxidative stress, muscle atrophy and weakness, and an acceleration in age-related phenotypes associated with sarcopenia. The goal of this study is to determine whether oxidative stress directly influences the acceleration of proteolysis in skeletal muscle of Sod1-/- mice as a function of age. Compared to control, Sod1-/- muscle showed a significant elevation in protein carbonyls and 3-nitrotyrosine levels, suggesting high oxidative and nitrosative protein modifications were present. In addition, age-dependent muscle atrophy in Sod1-/- muscle was accompanied by an upregulation of the cysteine proteases, calpain, and caspase-3, which are known to play a key role in the initial breakdown of sarcomeres during atrophic conditions. Furthermore, an increase in oxidative stress-induced muscle atrophy was also strongly coupled with simultaneous activation of two major proteolytic systems, the ubiquitin-proteasome and lysosomal autophagy pathways. Collectively, our data suggest that chronic oxidative stress in Sod1-/- mice accelerates age-dependent muscle atrophy by enhancing coordinated activation of the proteolytic systems, thereby resulting in overall protein degradation.
Collapse
Affiliation(s)
- Young C Jang
- School of Biological Sciences and Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Karl Rodriguez
- Sam & Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michael S Lustgarten
- Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Florian L Muller
- MD Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Arunabh Bhattacharya
- School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX, USA
| | | | - Jeongmoon J Choi
- School of Biological Sciences and Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nan Hee Lee
- School of Biological Sciences and Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Arlan G Richardson
- Reynolds Oklahoma Center on Aging, Oklahoma Health Science Center, Oklahoma City, OK, USA
| | | |
Collapse
|
28
|
Kang P, Huang X, Wan Z, Liang T, Wang Y, Li X, Zhang J, Zhu H, Liu Y. Kinetics of changes in gene and microRNA expression related with muscle inflammation and protein degradation following LPS-challenge in weaned piglets. Innate Immun 2020; 27:23-30. [PMID: 33232194 PMCID: PMC7780359 DOI: 10.1177/1753425920971032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
To test the dynamic changes of the expression of genes and microRNA in the gastrocnemius muscle after LPS challenge, 36 piglets were assigned to a control group (slaughtered 0 h after saline injection) and LPS groups (slaughtered at 1 h, 2 h, 4 h, 8 h, and 12 h after LPS treatment, respectively). After LPS treatment, the mRNA expression of IL-1β, IL-6, and TNF-α reached maximal levels at 1 h, 2 h, and 1 h, respectively (P < 0.05), and mRNA expression of TLR4, NODs, muscle-specific ring finger 1, and muscle atrophy F-box peaked at 12 h (P < 0.05). Moreover, the expression of miR-122, miR-135a, and miR-370 reduced at 1 h, 1 h, and 2 h, respectively (P < 0.05), and miR-34a, miR-224, miR-132, and miR-145 reached maximum expression levels at 1 h, 1 h, 2 h, and 4 h, respectively (P < 0.05). These results suggested that mRNA expression of pro-inflammatory cytokines was elevated in the early stage, mRNA expression of genes related to TLR4 and NODs signaling pathways and protein degradation increased in the later phase, and the expression of microRNA related to muscle inflammation and protein degradation changed in the early stage after LPS injection.
Collapse
Affiliation(s)
- Ping Kang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, 74615Wuhan Polytechnic University, China
| | - Xingfa Huang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, 74615Wuhan Polytechnic University, China
| | - Zhicheng Wan
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, 74615Wuhan Polytechnic University, China
| | - Tianzeng Liang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, 74615Wuhan Polytechnic University, China
| | - Yang Wang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, 74615Wuhan Polytechnic University, China
| | - Xiangen Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, 74615Wuhan Polytechnic University, China
| | - Jing Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, 74615Wuhan Polytechnic University, China
| | - Huiling Zhu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, 74615Wuhan Polytechnic University, China
| | - Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, 74615Wuhan Polytechnic University, China
| |
Collapse
|
29
|
Webster JM, Kempen LJAP, Hardy RS, Langen RCJ. Inflammation and Skeletal Muscle Wasting During Cachexia. Front Physiol 2020; 11:597675. [PMID: 33329046 PMCID: PMC7710765 DOI: 10.3389/fphys.2020.597675] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022] Open
Abstract
Cachexia is the involuntary loss of muscle and adipose tissue that strongly affects mortality and treatment efficacy in patients with cancer or chronic inflammatory disease. Currently, no specific treatments or interventions are available for patients developing this disorder. Given the well-documented involvement of pro-inflammatory cytokines in muscle and fat metabolism in physiological responses and in the pathophysiology of chronic inflammatory disease and cancer, considerable interest has revolved around their role in mediating cachexia. This has been supported by association studies that report increased levels of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) in some, but not all, cancers and in chronic inflammatory diseases such as chronic obstructive pulmonary disease (COPD) and rheumatoid arthritis (RA). In addition, preclinical studies including animal disease models have provided a substantial body of evidence implicating a causal contribution of systemic inflammation to cachexia. The presence of inflammatory cytokines can affect skeletal muscle through several direct mechanisms, relying on activation of the corresponding receptor expressed by muscle, and resulting in inhibition of muscle protein synthesis (MPS), elevation of catabolic activity through the ubiquitin-proteasomal system (UPS) and autophagy, and impairment of myogenesis. Additionally, systemic inflammatory mediators indirectly contribute to muscle wasting through dysregulation of tissue and organ systems, including GCs via the hypothalamus-pituitary-adrenal (HPA) axis, the digestive system leading to anorexia-cachexia, and alterations in liver and adipocyte behavior, which subsequently impact on muscle. Finally, myokines secreted by skeletal muscle itself in response to inflammation have been implicated as autocrine and endocrine mediators of cachexia, as well as potential modulators of this debilitating condition. While inflammation has been shown to play a pivotal role in cachexia development, further understanding how these cytokines contribute to disease progression is required to reveal biomarkers or diagnostic tools to help identify at risk patients, or enable the design of targeted therapies to prevent or delay the progression of cachexia.
Collapse
Affiliation(s)
- Justine M. Webster
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Laura J. A. P. Kempen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Rowan S. Hardy
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Institute for Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Arthritis Research UK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham, United Kingdom
| | - Ramon C. J. Langen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
30
|
Gonzalez-Ruiz C, Cordero-Anguiano P, Morales-Guadarrama A, Mondragón-Lozano R, Sánchez-Torres S, Salgado-Ceballos H, Villarreal F, Meaney E, Ceballos G, Nájera N. (-)-Epicatechin reduces muscle waste after complete spinal cord transection in a murine model: role of ubiquitin-proteasome system. Mol Biol Rep 2020; 47:8975-8985. [PMID: 33151476 DOI: 10.1007/s11033-020-05954-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/28/2020] [Indexed: 12/17/2022]
Abstract
The skeletal muscle mass reduces 30-60% after spinal cord injury, this is mostly due to protein degradation through ubiquitin-proteasome system. In this work, we propose that the flavanol (-)-epicatechin, due its widespread biological effects on muscle health, can prevent muscle mass decrease after spinal cord injury. Thirty-six female Long Evans rats were randomized into 5 groups: (1) Spinal cord injury 7 days, (2) Spinal cord injury + (-)-epicatechin 7 days, (3) Spinal cord injury 30 days, (4) Spinal cord injury + (-)-epicatechin 30 days and (5) Sham (Only laminectomy). Hind limb perimeter, muscle cross section area, fiber cross section area and ubiquitin-proteasome system protein expression together with total protein ubiquitination were assessed. At 30 days Spinal cord injury group lost 49.52 ± 2.023% of muscle cross section area (-)-epicatechin treated group lost only 24.28 ± 15.45% being a significant difference. Ubiquitin-proteasome markers showed significant changes. FOXO1a increased in spinal cord injury group vs Sham (-)-epicatechin reduced this increase. In spinal cord injury group MAFbx increased significantly vs Sham but decrease in (-)-epicatechin treatment group at 30 days. At 7 and 30 days MuRF1 increased in the spinal cord injury and decreased in the (-)-epicatechin group. The global protein ubiquitination increases after spinal cord injury, epicatechin treatment induce a significant decrease in protein ubiquitination. These results suggest that (-)-epicatechin reduces the muscle waste after spinal cord injury through down regulation of the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Cristian Gonzalez-Ruiz
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico
| | - Paola Cordero-Anguiano
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico
| | - Axayacatl Morales-Guadarrama
- Centro Nacional de Investigación en Imagenología e Instrumentación Médica, Departamento de Ingeniería Eléctrica, Universidad Autónoma Metropolitana Iztapalapa, Mexico, Mexico
| | - Rodrigo Mondragón-Lozano
- Consejo Nacional de Ciencia y Tecnología, Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico, Mexico
| | - Stephanie Sánchez-Torres
- División de Ciencias Biológicas y de la Salud, Posgrado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Mexico, Mexico
| | - Hermelinda Salgado-Ceballos
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico, Mexico
| | | | - Eduardo Meaney
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico
| | - Guillermo Ceballos
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico.
| | - Nayelli Nájera
- Laboratorio de Investigación Integral Cardiometabólica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico, Mexico.
| |
Collapse
|
31
|
Ji LL, Yeo D, Kang C, Zhang T. The role of mitochondria in redox signaling of muscle homeostasis. JOURNAL OF SPORT AND HEALTH SCIENCE 2020; 9:386-393. [PMID: 32780692 PMCID: PMC7498629 DOI: 10.1016/j.jshs.2020.01.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/20/2019] [Accepted: 12/12/2019] [Indexed: 05/07/2023]
Abstract
In the past, contraction-induced production of reactive oxygen species (ROS) has been implicated in oxidative stress to skeletal muscle. As research advances, clear evidence has revealed a more complete role of ROS under both physiologic and pathologic conditions. Central to the role of ROS is the redox signaling pathways that control exercise-induced major physiologic and cellular responses and adaptations, such as mitochondrial biogenesis, mitophagy, mitochondrial morphologic dynamics, antioxidant defense, and inflammation. The current review focuses on how muscle contraction and immobilization may activate or inhibit redox signalings and their impact on muscle mitochondrial homeostasis and physiologic implications.
Collapse
Affiliation(s)
- Li Li Ji
- The Laboratory of Physiological Hygiene and Exercise Science, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA.
| | - Dongwook Yeo
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Chounghun Kang
- Department of Physical Education, Inha University, Incheon 22212, Republic of Korea
| | - Tianou Zhang
- Department of Kinesiology, Health and Nutrition, University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
32
|
Sharma B, Dutt V, Kaur N, Mittal A, Dabur R. Tinospora cordifolia protects from skeletal muscle atrophy by alleviating oxidative stress and inflammation induced by sciatic denervation. JOURNAL OF ETHNOPHARMACOLOGY 2020; 254:112720. [PMID: 32114167 DOI: 10.1016/j.jep.2020.112720] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 12/09/2019] [Accepted: 02/24/2020] [Indexed: 06/10/2023]
Abstract
ETHANOPHARMACOLOGICAL RELEVANCE Tinospora cordifolia (TC) is widely being used as immunomodulatory and re-juvenile drug and well described in Indian Ayurveda system of medicine. Rejuvenation also means the fine tuning of the skeletal muscles. Skeletal muscle related disorder, i.e. atrophy is major problem which arise due to cachexia, sarcopenia and immobilization. However, despite of the great efforts, there is scarcity of FDA approved drugs in the market to treat skeletal muscle atrophy. AIM OF THE STUDY The current study was aimed to explore the in-vitro and in-vivo efficacy and mechanism of TC in myogenic differentiation and skeletal muscle atrophy to establish the possibility of its usage to counteract skeletal muscle atrophy. MATERIALS AND METHODS C2C12 cell lines were used to determine myogenic potential and anti-atrophic effects of T. cordifolia water extract (TCE). Its in-vitro efficacy was re-validated in vivo by supplementation of TCE at a dose of 200 mg/kg/p.o. for 30 days in denervated mice model of skeletal muscle atrophy. Effects of TCE administration on levels of oxidative stress, inflammatory markers and proteolysis were determined. RESULTS TCE supplementation displayed increased lymphocyte proliferation and induced myogenic differentiation of C2C12 myoblasts by significantly increasing myocytes length and thickness, in comparison to control (p < 0.05). TCE supplementation decreased oxidative stress and inflammatory response by significantly modulating activities of catalase, glutathione peroxidase, lipid peroxidase, superoxide dismutase and β-glucuronidase (p < 0.05). It increased MF-20c expression and ameliorated degradation of muscle protein by down-regulating MuRF-1 and calpain activity. CONCLUSION TCE supplementation promotes myogenic differentiation in C2C12 cell lines and prevents denervation induced skeletal muscle atrophy by antagonizing the proteolytic systems (calpain and UPS) and maintaining the oxidative defense mechanism of the cell. Hence, TCE can be used as a protective agent against muscle atrophy.
Collapse
Affiliation(s)
- Bhawana Sharma
- Clinical Biochemistry Research Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Vikas Dutt
- Skeletal Muscle Lab, Department of Biochemistry, University College, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Nirmaljeet Kaur
- Skeletal Muscle Lab, Department of Biochemistry, University College, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Ashwani Mittal
- Skeletal Muscle Lab, Department of Biochemistry, University College, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Rajesh Dabur
- Clinical Biochemistry Research Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| |
Collapse
|
33
|
Abstract
Significance: Regular contractile activity plays a critical role in maintaining skeletal muscle morphological integrity and physiological function. If the muscle is forced to stop contraction, such as during limb immobilization (IM), the IGF/Akt/mTOR signaling pathway that normally stimulates protein synthesis and inhibits proteolysis will be suppressed, whereas the FoxO-controlled catabolic pathways such as ubiquitin-proteolysis and autophagy/mitophagy will be activated and dominate, resulting in muscle fiber atrophy. Recent Advances: Mitochondria occupy a central position in the regulation of both protein synthesis and degradation through several redox-sensitive pathways, including peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), mitochondrial fusion and fission proteins, mitophagy, and sirtuins. Prolonged IM downregulates PGC-1α due to AMPK (5'-AMP-activated protein kinase) and FoxO activation, thus decreasing mitochondrial biogenesis and causing oxidative damage. Decrease of mitochondrial inner membrane potential and increase of mitochondrial fission can trigger cascades of mitophagy leading to loss of mitochondrial homeostasis (mitostasis), inflammation, and apoptosis. The phenotypic outcomes of these disorders are compromised muscle function and fiber atrophy. Critical Issues: Given the molecular mechanism of the pathogenesis, it is imperative that the integrity of intracellular signaling be restored to prevent the deterioration. So far, overexpression of PGC-1α via transgene and in vivo DNA transfection has been found to be effective in ameliorating mitostasis and reduces IM-induced muscle atrophy. Nutritional supplementation of select amino acids and phytochemicals also provides mechanistic and practical insights into the prevention of muscle disuse atrophy. Future Directions: In light of the importance of mitochondria in regulating the various critical signaling pathways, future work should focus on exploring new epigenetic strategies to restore mitostasis and redox balance.
Collapse
Affiliation(s)
- Li Li Ji
- The Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
| | - Dongwook Yeo
- The Laboratory of Physiological Hygiene and Exercise Science, School of Kinesiology, University of Minnesota Twin Cities, Minneapolis, Minnesota, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Chounghun Kang
- Departmet Physical Education, Inha University, Incheon, South Korea
| |
Collapse
|
34
|
Sharma B, Dabur R. Role of Pro-inflammatory Cytokines in Regulation of Skeletal Muscle Metabolism: A Systematic Review. Curr Med Chem 2020; 27:2161-2188. [DOI: 10.2174/0929867326666181129095309] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 12/18/2022]
Abstract
Background:
Metabolic pathways perturbations lead to skeletal muscular atrophy in the
cachexia and sarcopenia due to increased catabolism. Pro-inflammatory cytokines induce the catabolic
pathways that impair the muscle integrity and function. Hence, this review primarily concentrates
on the effects of pro-inflammatory cytokines in regulation of skeletal muscle metabolism.
Objective:
This review will discuss the role of pro-inflammatory cytokines in skeletal muscles during
muscle wasting conditions. Moreover, the coordination among the pro-inflammatory cytokines
and their regulated molecular signaling pathways which increase the protein degradation will be
discussed.
Results:
During normal conditions, pro-inflammatory cytokines are required to balance anabolism
and catabolism and to maintain normal myogenesis process. However, during muscle wasting their
enhanced expression leads to marked destructive metabolism in the skeletal muscles. Proinflammatory
cytokines primarily exert their effects by increasing the expression of calpains and E3
ligases as well as of Nf-κB, required for protein breakdown and local inflammation. Proinflammatory
cytokines also locally suppress the IGF-1and insulin functions, hence increase the
FoxO activation and decrease the Akt function, the central point of carbohydrates lipid and protein
metabolism.
Conclusion:
Current advancements have revealed that the muscle mass loss during skeletal muscular
atrophy is multifactorial. Despite great efforts, not even a single FDA approved drug is available
in the market. It indicates the well-organized coordination among the pro-inflammatory cytokines
that need to be further understood and explored.
Collapse
Affiliation(s)
- Bhawana Sharma
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana-124001, India
| | - Rajesh Dabur
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana-124001, India
| |
Collapse
|
35
|
Chemerin: A Potential Regulator of Inflammation and Metabolism for Chronic Obstructive Pulmonary Disease and Pulmonary Rehabilitation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4574509. [PMID: 32337250 PMCID: PMC7166297 DOI: 10.1155/2020/4574509] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/05/2020] [Accepted: 03/23/2020] [Indexed: 01/09/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) features chronic inflammatory reactions of both intra- and extrapulmonary nature. Moreover, COPD is associated with abnormal glucose and lipid metabolism in patients, which influences the prognosis and chronicity of this disease. Abnormal glucose and lipid metabolism are also closely related to inflammation processes. Further insights into the interactions of inflammation and glucose and lipid metabolism might therefore inspire novel therapeutic interventions to promote lung rehabilitation. Chemerin, as a recently discovered adipokine, has been shown to play a role in inflammatory response and glucose and lipid metabolism in many diseases (including COPD). Chemerin recruits inflammatory cells to sites of inflammation during the early stages of COPD, leading to endothelial barrier dysfunction, early vascular remodeling, and angiogenesis. Moreover, it supports the recruitment of antigen-presenting cells that guide immune cells as part of the body's inflammatory responses. Chemerin also regulates metabolism via activation of its cognate receptors. Glucose homeostasis is affected via effects on insulin secretion and sensitivity, and lipid metabolism is changed by increased transformation of preadipocytes to mature adipocytes through chemerin-binding receptors. Controlling chemerin signaling may be a promising approach to improve various aspects of COPD-related dysfunction. Importantly, several studies indicate that chemerin expression in vivo is influenced by exercise. Although available evidence is still limited, therapeutic alterations of chemerin activity may be a promising target of therapeutic approaches aimed at the rehabilitation of COPD patients based on exercises. In conclusion, chemerin plays an essential role in COPD, especially in the inflammatory responses and metabolism, and has a potential to become a target for, and a biomarker of, curative mechanisms underlying exercise-mediated lung rehabilitation.
Collapse
|
36
|
Gogulothu R, Nagar D, Gopalakrishnan S, Garlapati VR, Kallamadi PR, Ismail A. Disrupted expression of genes essential for skeletal muscle fibre integrity and energy metabolism in Vitamin D deficient rats. J Steroid Biochem Mol Biol 2020; 197:105525. [PMID: 31705962 DOI: 10.1016/j.jsbmb.2019.105525] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/16/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022]
Abstract
Vitamin D, a secosteroid that regulates mineral homeostasis via its actions in intestine, bone, kidneys and parathyroid glands, has many other target tissues, including skeletal muscle. In the present study, we used rats to examine if diet-induced vitamin D deficiency or insufficiency altered protein synthesis in muscle via the mTOR pathway, and impaired skeletal muscle quality by changing expression of genes needed for its function. Vitamin D deficiency resulted in reduced levels of phosphorylated mTOR, and suppressed mTOR-dependent phosphorylation of 4E-BP1 and p70-S6K, implying a decrease in activity of the protein synthesis machinery. These changes were coupled with up regulation of genes that are negative regulators of muscle growth (Fbxo32 & Trim63), leading to a net loss of skeletal muscle mass. Vitamin D deficiency or insufficiency also led to a decrease in expression of both myosin and actin-associated proteins (Myh1, Myh2, Myh7, Tnnc1& Tnnt1), which are essential for generation of the mechanical force needed for muscle contraction. We also detected a decrease in expression of glycolytic and oxidative enzyme genes (Hk2, Pfkm, Cs, Pdk4 & βHad) and transcriptional coactivator genes (Ppargc-1α & Ppargc-1β) which indicate a low oxidative capacity of skeletal muscle in the vitamin D deficient state. Furthermore, decreased citrate synthase activity corroborates a decrease in mitochondrial density and aerobic capacity of the muscle. In conclusion, our study demonstrates that chronic vitamin D deficiency or insufficiency reduced the size of skeletal muscle fibres, altered their composition, and decreased their oxidative potential. Most of the changes observed were reversible, either partially or completely, by restoring vitamin D to the diet of the deficient rats.
Collapse
Affiliation(s)
- Ramesh Gogulothu
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | - Devika Nagar
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | | | - Venkat R Garlapati
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India
| | | | - Ayesha Ismail
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, India.
| |
Collapse
|
37
|
Molina Peña ME, Sánchez CM, Rodríguez-Triviño CY. Physiopathological mechanisms of diaphragmatic dysfunction associated with mechanical ventilation. ACTA ACUST UNITED AC 2020; 67:195-203. [PMID: 31982168 DOI: 10.1016/j.redar.2019.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/06/2019] [Accepted: 12/16/2019] [Indexed: 02/07/2023]
Abstract
Ventilator-induced diaphragm dysfunction (VIDD) is the loss of diaphragmatic muscle strength'related to of mechanical ventilation, noticed during the first day or 48hours after initiating controlled mechanical ventilation. This alteration has been related to disruption on the insulin growth factor/phosphoinositol 3-kinase/kinase B protein pathway (IGF/PI3K/AKT), in addition to an overexpression of FOXO, expression of NF-kB signaling, increase function of muscular ubiquitin ligase and activation of caspasa-3. VIDD has a negative impact on quality of life, duration of mechanical ventilation, and hospitalization stance and cost. More studies are necessary to understated the process and impact of VIDD. This is a narrative review of non-systematic literature, aiming to explain the molecular pathways involved in VIDD.
Collapse
Affiliation(s)
- M E Molina Peña
- Semillero de Fisiología Pr ctica aplicada, Grupo Navarra Medicina, Departamento de Ciencias Fisiológicas, Facultad de Ciencias de la Salud, Fundación Universitaria Navarra-UNINAVARRA, Neiva, Huila, Colombia.
| | - C M Sánchez
- Semillero de Fisiología Pr ctica aplicada, Grupo Navarra Medicina, Departamento de Ciencias Fisiológicas, Facultad de Ciencias de la Salud, Fundación Universitaria Navarra-UNINAVARRA, Neiva, Huila, Colombia
| | - C Y Rodríguez-Triviño
- Grupo Navarra Medicina, Departamento de Ciencias Fisiológicas, Facultad de Ciencias de la Salud, Fundación Universitaria Navarra-UNINAVARRA, Neiva, Huila, Colombia; Grupo Cuidar, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| |
Collapse
|
38
|
Lavin KM, Perkins RK, Jemiolo B, Raue U, Trappe SW, Trappe TA. Effects of aging and lifelong aerobic exercise on basal and exercise-induced inflammation. J Appl Physiol (1985) 2020; 128:87-99. [PMID: 31751180 PMCID: PMC6985808 DOI: 10.1152/japplphysiol.00495.2019] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/07/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022] Open
Abstract
Age-associated chronic basal inflammation compromises muscle mass and adaptability, but exercise training may exert an anti-inflammatory effect. This investigation assessed basal and exercise-induced inflammation in three cohorts of men: young exercisers [YE; n = 10 men; 25 ± 1 yr; maximal oxygen consumption (V̇o2max), 53 ± 3 mL·kg-1·min-1; quadriceps area, 78 ± 3 cm2; means ± SE], old healthy nonexercisers (OH; n = 10; 75 ± 1 yr; V̇o2max, 22 ± 1 mL·kg-1·min-1; quadriceps area, 56 ± 3 cm2), and lifelong exercisers with an aerobic training history of 53 ± 1 yr (LLE; n = 21; 74 ± 1 yr; V̇o2max, 34 ± 1 mL·kg-1·min-1; quadriceps area, 67 ± 2 cm2). Resting serum IL-6, TNF-α, C-reactive protein, and IGF-1 levels were measured. Vastus lateralis muscle biopsies were obtained at rest (basal) and 4 h after an acute exercise challenge (3 × 10 repetitions, 70% 1-repetition maximum) to assess gene expression of cytokines [IL-6, TNF-α, IL-1β, IL-10, IL-4, interleukin-1 receptor antagonist (IL-1Ra), and transforming growth factor-β (TGF-β)], chemokines [IL-8 and monocyte chemoattractant protein-1 (MCP-1)], cyclooxygenase enzymes [cyclooxygenase-1 and -2 (COX-1 and COX-2, respectively), prostaglandin E2 synthases [microsomal prostaglandin E synthase 1 (mPGES-1) and cytosolic prostaglandin E2 synthase (cPGES)] and receptors [prostaglandin E2 receptor EP3 and EP4 subtypes (EP3 and EP4, respectively), and macrophage markers [cluster of differentiation 16b (CD16b) and CD163], as well as basal macrophage abundance (CD68+ cells). Aging led to higher (P ≤ 0.05) circulating IL-6 and skeletal muscle COX-1, mPGES-1, and CD163 expression. However, LLE had significantly lower serum IL-6 levels (P ≤ 0.05 vs. OH) and a predominantly anti-inflammatory muscle profile [higher IL-10 (P ≤ 0.05 vs. YE), TNF-α, TGF-β, and EP4 levels (P ≤ 0.05 vs. OH)]. In OH only, acute exercise increased expression of proinflammatory factors TNF-α, TGF-β, and IL-8 (P ≤ 0.05). LLE had postexercise gene expression similar to YE, except lower IL-10 (P ≤ 0.10), mPGES-1, and EP3 expression (P ≤ 0.05). Thus, although aging led to a proinflammatory profile within blood and muscle, lifelong exercise partially prevented this and generally preserved the acute inflammatory response to exercise seen in young exercising men. Lifelong exercise may positively impact muscle health throughout aging by promoting anti-inflammation in skeletal muscle.NEW & NOTEWORTHY This study assessed a unique population of lifelong aerobic exercising men and demonstrated that their activity status exerts an anti-inflammatory effect in skeletal muscle and circulation. Furthermore, we provide evidence that the inflammatory response to acute exercise is dysregulated by aging but preserved with lifelong exercise, which might improve skeletal muscle resilience to unaccustomed loading and adaptability into late life.
Collapse
Affiliation(s)
- Kaleen M Lavin
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| | - Ryan K Perkins
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| | - Bozena Jemiolo
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| | - Ulrika Raue
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| | - Scott W Trappe
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| | - Todd A Trappe
- Human Performance Laboratory, Ball State University, Muncie, Indiana
| |
Collapse
|
39
|
Ji LL, Yeo D. Cellular mechanism of immobilization-induced muscle atrophy: A mini review. SPORTS MEDICINE AND HEALTH SCIENCE 2019; 1:19-23. [PMID: 35782462 PMCID: PMC9219315 DOI: 10.1016/j.smhs.2019.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
It is well-established that regular contraction maintains morphological and functional integrity of skeletal muscle, whereas rigorous exercise training can upregulate muscle metabolic and contractile function. However, when muscles stop contraction, such as during immobilization (IM) and denervation, withdrawal of IGF/Akt/mTOR signaling allows FoxO-controlled protein degradation pathways to dominate. Mitochondria play an important role in regulating both protein synthesis and degradation via several redox sensitive signaling pathways such as mitochondrial biogenesis, fusion and fission dynamics, ubiquitin-proteolysis, autophagy/mitophagy, and apoptosis. During prolonged IM, downregulation of PGC-1α and increased mitochondrial oxidative damage facilitate fission protein and inflammatory cytokine production and activate mitophagic process, leading to a vicious cycle of protein degradation. This “mitostasis theory of muscle atrophy” is the opposite pathway of hormesis, which defines enhanced muscle function with contractile overload. The demonstration that PGC-1α overexpression via transgene or in vivo DNA transfection can successfully restore mitochondrial homeostasis and reverse myocyte atrophy supports such a proposition. Understanding the mechanism governing mitostasis can be instrumental to the treatment of muscle atrophy associated with bedrest, cancer cachexia and sarcopenia.
Collapse
Affiliation(s)
- Li Li Ji
- Corresponding author. 111 Cooke Hall, 1900 University Avenue SE, Minneapolis, MN, 55455, USA.
| | | |
Collapse
|
40
|
Ucci S, Renzini A, Russi V, Mangialardo C, Cammarata I, Cavioli G, Santaguida MG, Virili C, Centanni M, Adamo S, Moresi V, Verga-Falzacappa C. Thyroid Hormone Protects from Fasting-Induced Skeletal Muscle Atrophy by Promoting Metabolic Adaptation. Int J Mol Sci 2019; 20:ijms20225754. [PMID: 31731814 PMCID: PMC6888244 DOI: 10.3390/ijms20225754] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 02/07/2023] Open
Abstract
Thyroid hormones regulate a wide range of cellular responses, via non-genomic and genomic actions, depending on cell-specific thyroid hormone transporters, co-repressors, or co-activators. Skeletal muscle has been identified as a direct target of thyroid hormone T3, where it regulates stem cell proliferation and differentiation, as well as myofiber metabolism. However, the effects of T3 in muscle-wasting conditions have not been yet addressed. Being T3 primarily responsible for the regulation of metabolism, we challenged mice with fasting and found that T3 counteracted starvation-induced muscle atrophy. Interestingly, T3 did not prevent the activation of the main catabolic pathways, i.e., the ubiquitin-proteasome or the autophagy-lysosomal systems, nor did it stimulate de novo muscle synthesis in starved muscles. Transcriptome analyses revealed that T3 mainly affected the metabolic processes in starved muscle. Further analyses of myofiber metabolism revealed that T3 prevented the starvation-mediated metabolic shift, thus preserving skeletal muscle mass. Our study elucidated new T3 functions in regulating skeletal muscle homeostasis and metabolism in pathological conditions, opening to new potential therapeutic approaches for the treatment of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Sarassunta Ucci
- Pasteur Institute, 00161 Rome, Italy; (S.U.); (V.R.); (C.M.); (I.C.); (C.V.-F.)
| | - Alessandra Renzini
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, 00161 Rome, Italy; (A.R.); (G.C.); (S.A.)
| | - Valentina Russi
- Pasteur Institute, 00161 Rome, Italy; (S.U.); (V.R.); (C.M.); (I.C.); (C.V.-F.)
| | - Claudia Mangialardo
- Pasteur Institute, 00161 Rome, Italy; (S.U.); (V.R.); (C.M.); (I.C.); (C.V.-F.)
| | - Ilenia Cammarata
- Pasteur Institute, 00161 Rome, Italy; (S.U.); (V.R.); (C.M.); (I.C.); (C.V.-F.)
| | - Giorgia Cavioli
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, 00161 Rome, Italy; (A.R.); (G.C.); (S.A.)
| | - Maria Giulia Santaguida
- Department of Medico-Surgical Sciences and Biotechnologies Sapienza University of Rome, 04100 Latina, Italy; (M.G.S.); (C.V.); (M.C.)
| | - Camilla Virili
- Department of Medico-Surgical Sciences and Biotechnologies Sapienza University of Rome, 04100 Latina, Italy; (M.G.S.); (C.V.); (M.C.)
| | - Marco Centanni
- Department of Medico-Surgical Sciences and Biotechnologies Sapienza University of Rome, 04100 Latina, Italy; (M.G.S.); (C.V.); (M.C.)
| | - Sergio Adamo
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, 00161 Rome, Italy; (A.R.); (G.C.); (S.A.)
| | - Viviana Moresi
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, 00161 Rome, Italy; (A.R.); (G.C.); (S.A.)
- Correspondence:
| | - Cecilia Verga-Falzacappa
- Pasteur Institute, 00161 Rome, Italy; (S.U.); (V.R.); (C.M.); (I.C.); (C.V.-F.)
- Department of Medico-Surgical Sciences and Biotechnologies Sapienza University of Rome, 04100 Latina, Italy; (M.G.S.); (C.V.); (M.C.)
| |
Collapse
|
41
|
Lee SJ, Bae JH, Lee H, Lee H, Park J, Kang JS, Bae GU. Ginsenoside Rg3 upregulates myotube formation and mitochondrial function, thereby protecting myotube atrophy induced by tumor necrosis factor-alpha. JOURNAL OF ETHNOPHARMACOLOGY 2019; 242:112054. [PMID: 31271820 DOI: 10.1016/j.jep.2019.112054] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/26/2019] [Accepted: 06/30/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginsenoside Rg3 from Panax ginseng has reported to have multiple pharmacological activities including anti-diabetics, anti-inflammation and anti-cancer. However, the effect of ginsenoside Rg3 on myogenic differentiation and muscle atrophy is unknown. AIM TO THE STUDY In this study, we investigated the myogenic effect and underlying molecular mechanisms of ginsenoside Rg3 on myotube atrophy induced by tumor necrosis factor-α (TNF-α). MATERIALS AND METHODS C2C12 myoblasts were induced to differentiate for one day followed by the treatment of TNF-α along with vehicle or ginsenoside Rg3 for additional 2 days and subjected to immunoblotting, immunocytochemistry, quantitative RT-PCR and biochemical analysis for mitochondrial function. RESULTS Ginsenoside Rg3 promotes myogenic differentiation and multinucleated myotube formation through Akt activation in a dose-dependent manner, without any cytotoxicity. Ginsenoside Rg3 treatment restores myotube formation and increases myotube diameters under TNF-α-treated conditions. Ginsenoside Rg3 enhances Akt/mTOR (mammalian target of rapamycin) signaling that in turn stimulates muscle-specific gene expression such as myosin heavy chain (MHC) and Myogenin, and suppresses the expression of muscle-specific ubiquitin ligases. In addition, ginsenoside Rg3 in TNF-α-treated myotubes significantly inhibits the production of mitochondrial ROS and restores mitochondrial membrane potential (MMP) and ATP contents. Furthermore, ginsenoside Rg3 upregulates the activities and expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α) and the mitochondrial biogenetic transcription factors, nuclear respiratory factor-1 (NRF1) and mitochondrial transcription factor A (Tfam) in TNF-α-induced myotube atrophy. CONCLUSIONS This study provides a mechanistic insight into the effect of ginsenoside Rg3 on myogenic differentiation and myotube atrophy, suggesting that ginsenoside Rg3 has a promising potential as a therapeutic or neutraceutical remedy to intervene muscle weakness and atrophy.
Collapse
Affiliation(s)
- Sang-Jin Lee
- Research Institute of Pharmaceutical Science, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Ju Hyun Bae
- Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| | - Hani Lee
- Research Institute of Pharmaceutical Science, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Hyunji Lee
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| | - Jongsun Park
- Department of Pharmacology, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, 35015, Republic of Korea.
| | - Jong-Sun Kang
- Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| | - Gyu-Un Bae
- Research Institute of Pharmaceutical Science, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
42
|
Nuell S, Illera-Domínguez VR, Carmona G, Alomar X, Padullés JM, Lloret M, Cadefau JA. Hypertrophic muscle changes and sprint performance enhancement during a sprint-based training macrocycle in national-level sprinters. Eur J Sport Sci 2019; 20:793-802. [PMID: 31526116 DOI: 10.1080/17461391.2019.1668063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Abstract This study aimed to analyse changes in sprint performance, muscle volumes (MVs) and sprint mechanical parameters (SMPs) in national-level sprinters performing a 5-month indoor sprint-based training macrocycle (SBTM). Twelve well-trained sprinters were tested on three different occasions throughout the SBTM. Testing procedures included: sprint performance over 10m, 40m, 80m, 150m, and 300m; MRI of thighs, to compute MVs of quadriceps, hamstrings and adductors; and a 40m sprint using a radar gun to assess SMPs such as theoretical maximal horizontal force, theoretical maximal horizontal velocity (V0), maximal power and index of force application (DRF). Improvements in sprint performance of between 4% and 7% (ES = 0.46-1.11, P < 0.01) were accompanied by increments in: quadriceps of 6% (ES = 0.41, P < 0.01), hamstrings of 10% (ES = 0.62, P < 0.01), adductors of 12% (ES = 0.87, P < 0.01), V0 of 5% (ES = 0.40, P < 0.01) and DRF of 7% (ES = 0.91, P < 0.01). In conclusion, during the SBTM after the off-season, moderate hypertrophic changes occur in sprinters. Moreover, the greater increase in hamstrings and adductors, compared with quadriceps, might be related to the prominent role of these muscle groups in sprinting. Furthermore, the SBTM was likely effective at developing sprint performance in sprinters, thereby endorsing the idea that sprint-specific training is crucial for highly trained individuals. Finally, our results support the notion that V0 or the "velocity-oriented" force-velocity profile is determinant of performance in sprinters.
Collapse
Affiliation(s)
- Sergi Nuell
- Institut Nacional d'Educació Física de Catalunya (INEFC), Universitat de Barcelona (UB), Barcelona, Spain
| | - Víctor R Illera-Domínguez
- Institut Nacional d'Educació Física de Catalunya (INEFC), Universitat de Barcelona (UB), Barcelona, Spain
| | - Gerard Carmona
- Sports Sciences Department, FC Barcelona, Barcelona, Spain.,Tecnocampus, Escola Superior de Ciències de la Salut, Universitat Pompeu Fabra, Mataró, Spain
| | | | - Josep Maria Padullés
- Institut Nacional d'Educació Física de Catalunya (INEFC), Universitat de Barcelona (UB), Barcelona, Spain
| | - Mario Lloret
- Institut Nacional d'Educació Física de Catalunya (INEFC), Universitat de Barcelona (UB), Barcelona, Spain
| | - Joan Aureli Cadefau
- Institut Nacional d'Educació Física de Catalunya (INEFC), Universitat de Barcelona (UB), Barcelona, Spain.,Departament de Biomedicina, Universitat de Barcelona (UB), Barcelona, Spain
| |
Collapse
|
43
|
Abstract
It is well established that mitochondria play a critical role in the metabolic and physiological adaptation of skeletal muscle to enhanced contractile activity. Several redox-sensitive signaling pathways such as PGC-1α, AMPK, IGF/Akt/mTOR, SIRT, NFκB, and FoxO are involved with extensive crosstalk to regulate vital cellular functions such as mitochondrial biogenesis, mitochondrial fusion and fission dynamics, autophagy/mitophagy, and apoptosis under altered demand and stress. However, when muscles cease contraction, such as during immobilization and denervation, mitochondria undergo a series of detrimental changes characterized by downregulation of PGC-1α and antioxidant defense, increased ROS generation, activated FoxO, NFκB, and inflammation, enhanced ubiquitination, and finally mitophagy and apoptotic cascades. The phenotypic outcome of the discord of mitochondrial homeostasis is elevated proteolysis and muscle atrophy. The demonstration that PGC-1α overexpression via transgene or in vivo DNA transfection can restore mitochondrial homeostasis and reverse myocyte atrophy supports the "mitostasis theory of muscle atrophy".
Collapse
Affiliation(s)
- Li Li Ji
- The Laboratory of Physiological Hygiene and Exercise Science, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
| | - Dongwook Yeo
- The Laboratory of Physiological Hygiene and Exercise Science, University of Minnesota Twin Cities, Minneapolis, MN, 55455, USA
| |
Collapse
|
44
|
Anderson LB, Latour CD, Khader O, Massey BH, Cobb B, Pond AL. Ether-a-go-go related gene-1a potassium channel abundance varies within specific skeletal muscle fiber type. Eur J Transl Myol 2019; 29:8402. [PMID: 31579487 PMCID: PMC6767934 DOI: 10.4081/ejtm.2019.8402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/28/2019] [Indexed: 01/24/2023] Open
Abstract
The ERG1A K+ channel, which is partially responsible for repolarization of the cardiac action potential, has also been reported in skeletal muscle where it modulates ubiquitin proteolysis. Because ERG1A protein appears variably expressed in muscles composed of mixed fiber types, we hypothesized that its abundance in skeletal muscle might differ with fiber type. Indeed, skeletal muscle fibers vary in speed of contraction (fast or slow), which is mainly determined by myosin heavy chain (MyHC) isoform content, but a sarcolemmal K+ channel might also modulate contraction speed. To test our hypothesis, we cryo-sectioned Soleus (SOL), Extensor Digitorum Longus (EDL), and Gastrocnemius muscles from five rats. These muscles were chosen because the SOL and EDL contain an abundance of slow- and fast-twitch fibers, respectively, while the Gastrocnemius has a more heterogeneous composition. The muscle sections were co-immunostained for the ERG1A protein and either the fast- or slow-twitch MyHC to identify fiber type. ERG1A fluorescence was then measured in the sarcolemma of each fiber type and compared. The data reveal that the ERG1A protein is more abundant in the fibers of the SOL than in the EDL muscles, suggesting ERG1A may be more abundant in the slow than the fast fibers, and this was confirmed with immunoblot. However, because of the homogeneity of fiber type within these muscles, it was not possible to get enough data from both fiber types within a single muscle to compare ERG1A composition within fiber type. However, immunohistochemistry of sections from the fiber type heterogeneous Gastrocnemius muscle reveals that slow fibers had, on average, a 17.2% greater ERG1A fluorescence intensity than fast fibers (p<0.03). Further, immunoblot reveals that ERG1A protein is 41.6% more abundant (p=0.051) in old than in young rat Gastrocnemius muscle. We postulate that this membrane bound voltage-gated channel may affect membrane characteristics, the duration of the action potential generated, and/or the speed of contraction. Indeed, ERG1A protein is more abundant in aged and atrophic skeletal muscle, both of which exhibit slower rates of contraction.
Collapse
Affiliation(s)
- Luke B. Anderson
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL
| | - Chase D. Latour
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO
| | - Omar Khader
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL
| | | | - Brittan Cobb
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL
| | - Amber L. Pond
- Department of Anatomy, Southern Illinois University School of Medicine, Carbondale, IL
| |
Collapse
|
45
|
Dzik KP, Kaczor JJ. Mechanisms of vitamin D on skeletal muscle function: oxidative stress, energy metabolism and anabolic state. Eur J Appl Physiol 2019; 119:825-839. [PMID: 30830277 PMCID: PMC6422984 DOI: 10.1007/s00421-019-04104-x] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/13/2019] [Indexed: 02/08/2023]
Abstract
PURPOSE This review provides a current perspective on the mechanism of vitamin D on skeletal muscle function with the emphasis on oxidative stress, muscle anabolic state and muscle energy metabolism. It focuses on several aspects related to cellular and molecular physiology such as VDR as the trigger point of vitamin D action, oxidative stress as a consequence of vitamin D deficiency. METHOD The interaction between vitamin D deficiency and mitochondrial function as well as skeletal muscle atrophy signalling pathways have been studied and clarified in the last years. To the best of our knowledge, we summarize key knowledge and knowledge gaps regarding the mechanism(s) of action of vitamin D in skeletal muscle. RESULT Vitamin D deficiency is associated with oxidative stress in skeletal muscle that influences the mitochondrial function and affects the development of skeletal muscle atrophy. Namely, vitamin D deficiency decreases oxygen consumption rate and induces disruption of mitochondrial function. These deleterious consequences on muscle may be associated through the vitamin D receptor (VDR) action. Moreover, vitamin D deficiency may contribute to the development of muscle atrophy. The possible signalling pathway triggering the expression of Atrogin-1 involves Src-ERK1/2-Akt- FOXO causing protein degradation. CONCLUSION Based on the current knowledge we propose that vitamin D deficiency results from the loss of VDR function and it could be partly responsible for the development of neurodegenerative diseases in human beings.
Collapse
Affiliation(s)
- Katarzyna Patrycja Dzik
- Department of Neurobiology of Muscle, Gdansk University of Physical Education and Sport, Kazimierza Gorskiego 1, 80-336, Gdansk, Poland
| | - Jan Jacek Kaczor
- Department of Neurobiology of Muscle, Gdansk University of Physical Education and Sport, Kazimierza Gorskiego 1, 80-336, Gdansk, Poland.
| |
Collapse
|
46
|
Yoshihara T, Natsume T, Tsuzuki T, Chang SW, Kakigi R, Sugiura T, Naito H. Sex differences in forkhead box O3a signaling response to hindlimb unloading in rat soleus muscle. J Physiol Sci 2019; 69:235-244. [PMID: 30259391 PMCID: PMC10716962 DOI: 10.1007/s12576-018-0640-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/16/2018] [Indexed: 12/28/2022]
Abstract
We tested the hypothesis that there are sex differences in hindlimb unloading-induced activation of the forkhead box subfamily O3a (FoxO3a) signaling pathway in rat soleus muscle. Age-matched male and female Wistar rats were subjected to hindlimb unloading, and the soleus muscle was removed before or 1 or 7 days after unloading. Female rats showed greater percent changes in relative soleus muscle weight than males. FoxO3a phosphorylation was lower in females than in males and was associated with higher levels of protein ubiquitination 7 days after unloading. Heat shock protein 72 (Hsp72) levels were lower in female rats and increased in males during unloading. Female rats showed slightly higher myostatin levels, which showed a non-significant decline in male rats following unloading. Thus, males and females show different responses to the FoxO3a/ubiquitin-proteasome pathway following hindlimb unloading in rat soleus muscle, which may be associated with differences in Hsp72 expression and myostatin signaling.
Collapse
Affiliation(s)
- Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan.
| | - Toshiharu Natsume
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| | - Takamasa Tsuzuki
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
- Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya, Aichi, 468-8503, Japan
| | - Shuo-Wen Chang
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| | - Ryo Kakigi
- Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Takao Sugiura
- Faculty of Education, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8513, Japan
| | - Hisashi Naito
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hirakagakuendai, Inzai, Chiba, 270-1695, Japan
| |
Collapse
|
47
|
Schilling BK, Schusterman MA, Kim DY, Repko AJ, Klett KC, Christ GJ, Marra KG. Adipose-derived stem cells delay muscle atrophy after peripheral nerve injury in the rodent model. Muscle Nerve 2019; 59:603-610. [PMID: 30681163 DOI: 10.1002/mus.26432] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 01/12/2019] [Accepted: 01/20/2019] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Injuries to peripheral nerves cause distal muscle atrophy. The effects of adipose-derived stem cell (ASC) injections into a muscle after injury were examined. METHODS A 1.5 cm defect in the rat sciatic nerve was created, resulting in gastrocnemius muscle atrophy. The nerve defect was repaired with autograft; DiR-labeled ASCs were injected into the gastrocnemius immediately postoperatively. Quantitation of gross musculature and muscle fiber area, cell survival, fibrosis, lipid deposition, inflammation, and reconstructive responses were investigated. RESULTS ASCs were identified in the muscle at 6 weeks, where injections showed increased muscle mass percentage retained, larger average fiber area, and less overall lipid content accumulated throughout the musculature. Muscles having received ASCs showed increased presence of interlukin-10 and Ki67, and decreased inducible nitric oxide synthase (iNOS). DISCUSSION This investigation is suggestive that an ASC injection into denervated muscle post-operatively is able to delay the onset of atrophy. Muscle Nerve 59:603-603, 2019.
Collapse
Affiliation(s)
- Benjamin K Schilling
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - M Asher Schusterman
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Deok-Yeol Kim
- Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexander J Repko
- Department of Biology, School of Arts & Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katarina C Klett
- Department of Chemical Engineering, School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - George J Christ
- Department of Biomedical Engineering and Orthopaedic Surgery, University of Virginia, Charlottesville, VA, USA
| | - Kacey G Marra
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Plastic Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
48
|
Nascimento CM, Ingles M, Salvador-Pascual A, Cominetti MR, Gomez-Cabrera MC, Viña J. Sarcopenia, frailty and their prevention by exercise. Free Radic Biol Med 2019; 132:42-49. [PMID: 30176345 DOI: 10.1016/j.freeradbiomed.2018.08.035] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/08/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022]
Abstract
Sarcopenia is a major component of the frailty syndrome, both being considered as strong predictors of morbidity, disability, and death in older people. In this review, we explore the definitions of sarcopenia and frailty and summarize the current knowledge on their relationship with oxidative stress and the possible therapeutic interventions to prevent or treat them, including exercise-based interventions and multimodal strategies. We highlight the relevance of the impairment of the nervous system and of the anabolic response (protein synthesis) in muscle aging leading to frailty and sarcopenia. We also discuss the importance of malnutrition and physical inactivity in these geriatric syndromes. Finally, we propose multimodal interventions, including exercise programs and nutritional supplementation, as the strategies to prevent and treat both sarcopenia and frailty.
Collapse
Affiliation(s)
- C M Nascimento
- Laboratorio de Biologia do Envelhecimento (LABEN), Departamento de Gerontologia UFSCar, Rod. Washington Luis, km 235, São Carlos, SP, Brazil
| | - M Ingles
- Freshage Research Group. Department of Physiotherapy. University of Valencia, CIBERFES, INCLIVA, Spain
| | - A Salvador-Pascual
- Freshage Research Group. Department of Physiology. University of Valencia, CIBERFES, INCLIVA, Spain
| | - M R Cominetti
- Laboratorio de Biologia do Envelhecimento (LABEN), Departamento de Gerontologia UFSCar, Rod. Washington Luis, km 235, São Carlos, SP, Brazil
| | - M C Gomez-Cabrera
- Freshage Research Group. Department of Physiology. University of Valencia, CIBERFES, INCLIVA, Spain.
| | - J Viña
- Freshage Research Group. Department of Physiology. University of Valencia, CIBERFES, INCLIVA, Spain
| |
Collapse
|
49
|
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: 1.8] [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
|
50
|
Loss of SMYD1 Results in Perinatal Lethality via Selective Defects within Myotonic Muscle Descendants. Diseases 2018; 7:diseases7010001. [PMID: 30577454 PMCID: PMC6473627 DOI: 10.3390/diseases7010001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 12/11/2022] Open
Abstract
SET and MYND Domain 1 (SMYD1) is a cardiac and skeletal muscle-specific, histone methyl transferase that is critical for both embryonic and adult heart development and function in both mice and men. We report here that skeletal muscle-specific, myogenin (myoG)-Cre-mediated conditional knockout (CKO) of Smyd1 results in perinatal death. As early as embryonic day 12.5, Smyd1 CKOs exhibit multiple skeletal muscle defects in proliferation, morphology, and gene expression. However, all myotonic descendants are not afflicted equally. Trunk muscles are virtually ablated with excessive accumulation of brown adipose tissue (BAT), forelimb muscles are disorganized and improperly differentiated, but other muscles, such as the masseter, are normal. While expression of major myogenic regulators went unscathed, adaptive and innate immune transcription factors critical for BAT development/physiology were downregulated. Whereas classical mitochondrial BAT accumulation went unscathed following loss of SMYD1, key transcription factors, including PRDM16, UCP-1, and CIDE-a that control skeletal muscle vs. adipose fate, were downregulated. Finally, in rare adults that survive perinatal lethality, SMYD1 controls specification of some, but not all, skeletal muscle fiber-types.
Collapse
|