1
|
Kang M, Kim T, Joo H, Kim D, Lee DW, Hwang JK. Standardized Boesenbergia pandurata Extract Prevents Dexamethasone-Induced Muscle Atrophy and Dysfunction in C57BL/6 Mice. J Med Food 2025; 28:182-190. [PMID: 39630507 DOI: 10.1089/jmf.2024.k.0182] [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] [Indexed: 12/18/2024] Open
Abstract
Muscle atrophy, characterized by diminished muscle mass and impaired function, poses a substantial global health concern. Boesenbergia pandurata (Roxb.) Schltr., commonly known as fingerroot, possesses a variety of advantageous activities, including anti-inflammatory, antioxidant, antibacterial, and anticancer effects. However, there are currently no preclinical studies available that explore the potential of B. pandurata extract (BPE) to mitigate muscle atrophy. In this study, we aimed to explore the protective effects of BPE, standardized to panduratin A content, against muscle atrophy and its underlying molecular mechanisms in a dexamethasone-induced muscle atrophy mouse model. Compared with the dexamethasone group, BPE significantly restored muscle mass, muscle volume, muscle fiber cross-sectional area, grip strength, and exercise endurance. Additionally, BPE suppressed inflammatory responses by downregulating the expressions of nuclear factor kappa B and inflammatory cytokines while also enhancing antioxidant effects by increasing the expressions of antioxidant enzymes. Moreover, BPE promoted protein synthesis and muscle differentiation by stimulating the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin signaling pathway. Furthermore, it suppressed myostatin expression and inhibited the expressions of E3 ubiquitin ligases by preventing the nuclear translocation of forkhead box O3a, thereby alleviating proteolysis. Overall, BPE effectively regulates unbalanced protein metabolism, suggesting its potential as a functional food ingredient for preventing muscle wasting diseases.
Collapse
Affiliation(s)
- Minseong Kang
- Graduate Program in Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
| | - Taeuk Kim
- Graduate Program in Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
| | - Heeyeon Joo
- Division of Health Food Research and Development, NEWTREE Co., Ltd., Seoul, Republic of Korea
| | - Doun Kim
- Division of Health Food Research and Development, NEWTREE Co., Ltd., Seoul, Republic of Korea
| | - Dong-Woo Lee
- Graduate Program in Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
| | - Jae-Kwan Hwang
- Graduate Program in Bioindustrial Engineering, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Park S, Kim MY, Jeong J, Yang S, Kim MS, Moon I. Quantitative analysis of the dexamethasone side effect on human-derived young and aged skeletal muscle by myotube and nuclei segmentation using deep learning. Bioinformatics 2024; 41:btae658. [PMID: 39752317 PMCID: PMC11723526 DOI: 10.1093/bioinformatics/btae658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/25/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
MOTIVATION Skeletal muscle cells (skMCs) combine together to create long, multi-nucleated structures called myotubes. By studying the size, length, and number of nuclei in these myotubes, we can gain a deeper understanding of skeletal muscle development. However, human experimenters may often derive unreliable results owing to the unusual shape of the myotube, which causes significant measurement variability. RESULTS We propose a new method for quantitative analysis of the dexamethasone side effect on human-derived young and aged skeletal muscle by simultaneous myotube and nuclei segmentation using deep learning combined with post-processing techniques. The deep learning model outputs myotube semantic segmentation, nuclei semantic segmentation, and nuclei center, and post-processing applies a watershed algorithm to accurately distinguish overlapped nuclei and identify myotube branches through skeletonization. To evaluate the performance of the model, the myotube diameter and the number of nuclei were calculated from the generated segmented images and compared with the results calculated by human experimenters. In particular, the proposed model produced outstanding outcomes when comparing human-derived primary young and aged skMCs treated with dexamethasone. The proposed standardized and consistent automated image segmentation system for myotubes is expected to help streamline the drug-development process for skeletal muscle diseases. AVAILABILITY AND IMPLEMENTATION The code and the data are available at https://github.com/tdn02007/QA-skMCs-Seg.
Collapse
Affiliation(s)
- Seonghwan Park
- Department of Robotics and Mechatronics Engineering, DGIST, Daegu, 42988, South Korea
| | - Min Young Kim
- Department of New Biology, DGIST, Daegu, 42988, South Korea
| | - Jaewon Jeong
- Department of Robotics and Mechatronics Engineering, DGIST, Daegu, 42988, South Korea
| | - Sohae Yang
- Department of New Biology, DGIST, Daegu, 42988, South Korea
| | - Minseok S Kim
- Department of New Biology, DGIST, Daegu, 42988, South Korea
- CTCELLS Inc., Seoul, 06307, South Korea
| | - Inkyu Moon
- Department of Robotics and Mechatronics Engineering, DGIST, Daegu, 42988, South Korea
| |
Collapse
|
3
|
Yeo C, Kim H, Jeon WJ, Lee J, Hong JY, Kim H, Lee YJ, Baek SH, Ha IH. Protective effect of Luffa cylindrica Roemer against dexamethasone-induced muscle atrophy in primary rat skeletal muscle cells. J Muscle Res Cell Motil 2024; 45:1-10. [PMID: 37845555 PMCID: PMC10844154 DOI: 10.1007/s10974-023-09661-5] [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: 08/01/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
Glucocorticoids (GCs) are commonly used in the treatment of chronic inflammatory conditions. However, the administration of high doses and long-term use of GCs can induce muscle atrophy (MA) in patients, leading to a decline in quality of life and increased mortality. MA leads to protein degradation in skeletal muscle, resulting in a reduction of muscle mass. This process is triggered by GCs like dexamethasone (DEX), which induce the expression of E3 ubiquitin ligases, namely Atrogin-1 and muscle RING-finger protein-1 (MuRF1). In this study, we examined the anti-MA potential of Luffa cylindrica Roemer (LCR) on DEX-treated primary skeletal myotubes. Primary skeletal myotubes stimulated with LCR alone resulted in a significant upregulation of myotube development, characterized by an increase in both the number and diameter of myotubes. Contrastingly, combined treatment with LCR and DEX reduced the expression of Atrogin-1, while treatment with DEX alone induced the expression of MuRF1. Furthermore, LCR treatment successfully restored the number and diameter of myotubes that had been diminished by DEX treatment. These findings suggest that LCR holds potential for treating MA, as an accelerating effect on muscle development and anti-MA effects on primary skeletal muscle cells were observed.
Collapse
Affiliation(s)
- Changhwan Yeo
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Hyunseong Kim
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Wan-Jin Jeon
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Junseon Lee
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Jin Young Hong
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Hyun Kim
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Yoon Jae Lee
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, 10326, Gyeonggi-do, Republic of Korea
| | - In-Hyuk Ha
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea.
| |
Collapse
|
4
|
Gartling G, Nakamura R, Sayce L, Kimball EE, Wilson A, Schneeberger S, Zimmerman Z, Garabedian MJ, Branski RC, Rousseau B. Acute Effects of Systemic Glucocorticoids on the Vocal Folds in a Pre-Clinical Model. Ann Otol Rhinol Laryngol 2024; 133:87-96. [PMID: 37497827 PMCID: PMC10818023 DOI: 10.1177/00034894231188571] [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] [Indexed: 07/28/2023]
Abstract
OBJECTIVES/HYPOTHESIS Systemic glucocorticoids (GC)s are employed to treat various voice disorders. However, GCs have varying pharmacodynamic properties with adverse effects ranging from changes in epithelial integrity, skeletal muscle catabolism, and altered body weight. We sought to characterize the acute temporal effects of systemic dexamethasone and methylprednisolone on vocal fold (VF) epithelial glucocorticoid receptor (GR) nuclear translocation, epithelial tight junction (ZO-1) expression, thyroarytenoid (TA) muscle fiber morphology, and body weight using an established pre-clinical model. We hypothesized dexamethasone and methylprednisolone will elicit changes in VF epithelial GR nuclear translocation, epithelial ZO-1 expression, TA muscle morphology, and body weight compared to placebo-treated controls. METHODS Forty-five New Zealand white rabbits received intramuscular injections of methylprednisolone (4.5 mg; n = 15), dexamethasone (450 µg; n = 15), or volume matched saline (n = 15) into the iliocostalis/longissimus muscle for 6 consecutive days. Vocal folds from 5 rabbits from each treatment group were harvested at 1-, 3-, or 7 days following the final injection and subjected to immunohistochemistry for ZO-1 and GR as well as TA muscle fiber cross-sectional area (CSA) measures. RESULTS Dexamethasone increased epithelial GR nuclear translocation and ZO-1 expression 1-day following injections compared to methylprednisolone (P = .024; P = .012). Dexamethasone and methylprednisolone increased TA CSA 1-day following injections (P = .011). Methylprednisolone decreased body weight 7 days following injections compared to controls (P = .004). CONCLUSIONS Systemic dexamethasone may more efficiently activate GR in the VF epithelium with a lower risk of body weight loss, suggesting a role for more refined approaches to GC selection for laryngeal pathology.
Collapse
Affiliation(s)
- Gary Gartling
- Communication Science and Disorders, University of Pittsburgh, Pittsburgh, PA, USA
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Ryosuke Nakamura
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Lea Sayce
- Communication Science and Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emily E. Kimball
- Hearing and Speech Sciences, Vanderbilt University, Nashville, TN, USA
- Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Azure Wilson
- Communication Science and Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steven Schneeberger
- Plastic and Reconstructive Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Zachary Zimmerman
- Communication Science and Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael J. Garabedian
- Microbiology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Urology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ryan C. Branski
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY, USA
- Otolaryngology-Head and Neck Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Bernard Rousseau
- Doisy College of Health Sciences, Saint Louis University, St. Louis, MO, USA
| |
Collapse
|
5
|
Ou BR, Hsu MH, Haung LY, Lin CJ, Kuo LL, Tsai YT, Chang YC, Lin WY, Huang TC, Wu YC, Yeh JY, Liang YC. Systematic Myostatin Expression Screening Platform for Identification and Evaluation of Myogenesis-Related Phytogenic in Pigs. Bioengineering (Basel) 2023; 10:1113. [PMID: 37892843 PMCID: PMC10604025 DOI: 10.3390/bioengineering10101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/17/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Skeletal muscle growth in livestock impacts meat quantity and quality. Concerns arise because certain feed additives, like beta-agonists, may affect food safety. Skeletal muscle is a specialized tissue consisting of nondividing and multinucleated muscle fibers. Myostatin (MSTN), a protein specific to skeletal muscle, is secreted and functions as a negative regulator of muscle mass by inhibiting the proliferation and differentiation of myoblasts. To enhance livestock muscle growth, phytogenic feed additives could be an alternative as they inhibit MSTN activity. The objective of this study was to establish a systematic screening platform using MSTN activity to evaluate phytogenics, providing scientific evidence of their assessment and potency. In this study, we established a screening platform to monitor myostatin promoter activity in rat L8 myoblasts. Extract of Glycyrrhiza uralensis (GUE), an oriental herbal medicine, was identified through this screening platform, and the active fractions of GUE were identified using a process-scale liquid column chromatography system. For in vivo study, GUE as a feed additive was investigated in growth-finishing pigs. The results showed that GUE significantly increased body weight, carcass weight, and lean content in pigs. Microbiota analysis indicated that GUE did not affect the composition of gut microbiota in pigs. In summary, this established rodent myoblast screening platform was used to identify a myogenesis-related phytogenic, GUE, and further demonstrated that the active fractions and compounds inhibited MSTN expression. These findings suggest a novel application for GUE in growth performance enhancement through modulation of MSTN expression. Moreover, this well-established screening platform holds significant potential for identifying and assessing a diverse range of phytogenics that contribute to the process of myogenesis.
Collapse
Affiliation(s)
- Bor-Rung Ou
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407, Taiwan; (B.-R.O.); (L.-Y.H.); (T.-C.H.)
| | - Ming-Hua Hsu
- Department of Chemistry, National Changhua University of Education, Changhua 500, Taiwan;
| | - Ling-Ya Haung
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407, Taiwan; (B.-R.O.); (L.-Y.H.); (T.-C.H.)
| | - Chuan-Ju Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; (C.-J.L.)
| | - Li-Li Kuo
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; (C.-J.L.)
| | - Yu-Ting Tsai
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407, Taiwan; (B.-R.O.); (L.-Y.H.); (T.-C.H.)
| | - Yu-Chia Chang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; (C.-J.L.)
| | - Wen-Yuh Lin
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407, Taiwan; (B.-R.O.); (L.-Y.H.); (T.-C.H.)
| | - Tsung-Chien Huang
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407, Taiwan; (B.-R.O.); (L.-Y.H.); (T.-C.H.)
| | - Yun-Chu Wu
- Department of Animal Science and Biotechnology, Tunghai University, Taichung 407, Taiwan; (B.-R.O.); (L.-Y.H.); (T.-C.H.)
| | - Jan-Ying Yeh
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung 413, Taiwan
| | - Yu-Chuan Liang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan; (C.-J.L.)
- College of Agriculture and Health, Tunghai University, Taichung 407, Taiwan
| |
Collapse
|
6
|
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: 44] [Impact Index Per Article: 22.0] [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
|
7
|
Yang M, Liu C, Jiang N, Liu Y, Luo S, Li C, Zhao H, Han Y, Chen W, Li L, Xiao L, Sun L. Myostatin: a potential therapeutic target for metabolic syndrome. Front Endocrinol (Lausanne) 2023; 14:1181913. [PMID: 37288303 PMCID: PMC10242177 DOI: 10.3389/fendo.2023.1181913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/04/2023] [Indexed: 06/09/2023] Open
Abstract
Metabolic syndrome is a complex metabolic disorder, its main clinical manifestations are obesity, hyperglycemia, hypertension and hyperlipidemia. Although metabolic syndrome has been the focus of research in recent decades, it has been proposed that the occurrence and development of metabolic syndrome is related to pathophysiological processes such as insulin resistance, adipose tissue dysfunction and chronic inflammation, but there is still a lack of favorable clinical prevention and treatment measures for metabolic syndrome. Multiple studies have shown that myostatin (MSTN), a member of the TGF-β family, is involved in the development and development of obesity, hyperlipidemia, diabetes, and hypertension (clinical manifestations of metabolic syndrome), and thus may be a potential therapeutic target for metabolic syndrome. In this review, we describe the transcriptional regulation and receptor binding pathway of MSTN, then introduce the role of MSTN in regulating mitochondrial function and autophagy, review the research progress of MSTN in metabolic syndrome. Finally summarize some MSTN inhibitors under clinical trial and proposed the use of MSTN inhibitor as a potential target for the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Ming Yang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chongbin Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Na Jiang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yan Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Shilu Luo
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chenrui Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Hao Zhao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Wei Chen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Li Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Li Xiao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| |
Collapse
|
8
|
Maidadi B, Ntchapda F, Miaffo D, Mahamad AT. Diabetes mellitus: Preventive and curative therapies with aqueous extract of Rytigynia senegalensis Blume (Rubiaceae) in Wistar rats. J Tradit Complement Med 2023. [DOI: 10.1016/j.jtcme.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
|
9
|
Choi RY, Kim BS, Ban EJ, Seo M, Lee JH, Kim IW. Mealworm Ethanol Extract Enhances Myogenic Differentiation and Alleviates Dexamethasone-Induced Muscle Atrophy in C2C12 Cells. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010058. [PMID: 36676007 PMCID: PMC9862036 DOI: 10.3390/life13010058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Aging, and other disease-related muscle disorders are serious health problems. Dexamethasone (DEX), a synthetic glucocorticoid, can trigger skeletal muscle atrophy. This study examined the effects of mealworm (Tenebrio molitor larva) ethanol extract (TME) on C2C12 myoblast differentiation and DEX-induced myotube atrophy. TME induced myotube formation compared to the differentiation medium (DM) group. TME also significantly increased the mRNA expression of muscle creatine kinase (CKm) and myogenic regulatory factors (MRFs), such as myogenin (MyoG), myogenic factor (Myf)5, and MRF4 (Myf6). TME dramatically increased the muscle-specific protein, MyoG, compared to the control, whereas the expression of myogenic differentiation 1 (MyoD) remained unchanged. It also activated the mammalian target of rapamycin (mTOR) signaling pathway. In the DEX-induced muscle atrophy C2C12 model, TME reduced the gene expression of atrogin-1, muscle RING finger protein-1 (MuRF-1), and myostatin, which are involved in protein degradation in skeletal muscles. Furthermore, TME elevated the phosphorylation of forkhead box O3 (FoxO3α) and protein kinase B (Akt). These findings suggest that TME can enhance myotube hypertrophy by regulating the mTOR signaling pathway, and can rescue DEX-induced muscle atrophy by alleviating atrophic muscle markers mediated by Akt activation. Thus, TME can be a potential therapeutic agent for treating muscle weakness and atrophy.
Collapse
|
10
|
Tamta AK, Shivanaiah B, Ningaraju S, Prabhashankar AB, Sundaresan NR. Cultured Neonatal Murine Primary Myotubes as a Model to Study Muscle Atrophy. Curr Protoc 2022; 2:e616. [PMID: 36440976 DOI: 10.1002/cpz1.616] [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] [Indexed: 06/16/2023]
Abstract
Besides genetic disorders, skeletal muscle atrophy mainly occurs as a consequence of underlying conditions such as prolonged inactivity, aging, and metabolic diseases, ultimately contributing to the risk of disability. Disturbances in cellular and molecular mechanisms involved in proteolysis and protein synthesis underpin muscle fiber shrinkage and decreased muscle fiber diameter. Stress-induced primary myotube culture is an established model for studying muscle atrophy. An in vitro model is an essential criterion in establishing preliminary data in a cell-autonomous manner that can later be validated using in vivo models. Here, we describe protocols for the isolation, culture, and differentiation of primary murine myotubes and the induction of myotube atrophy using dexamethasone, a synthetic corticosteroid. We further elaborate the procedure to validate degenerative parameters, such as assessing muscle fiber diameter, expression of muscle atrophy genes, and protein synthesis status under dexamethasone treatment. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Isolation and culture of primary myoblasts from rat or mouse pups Support Protocol 1: Preparation of coated tissue culture ware Support Protocol 2: Subculture of myoblasts Basic Protocol 2: Induction and assessment of myotube atrophy.
Collapse
Affiliation(s)
- Ankit Kumar Tamta
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Bhoomika Shivanaiah
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Sunayana Ningaraju
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Arathi Bangalore Prabhashankar
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| | - Nagalingam Ravi Sundaresan
- Cardiovascular and Muscle Research Laboratory, Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
11
|
Effects of Lactobacillus curvatus HY7602-Fermented Antlers in Dexamethasone-Induced Muscle Atrophy. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8090454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This study assessed the improvements yielded by Lactobacillus curvatus HY7602-fermented antlers (FA) in dexamethasone-induced muscle atrophy and the effects of bioactive compounds increased by fermentation. Dexamethasone-treated C2C12 myoblast cells were treated with FA and non-fermented antlers (NFA). FA showed inhibitory effects on muscle protein degradation in the C2C12 cells. Hsb:ICR mice were orally administered saline (control(CON) and dexamethasone only (DEX)), oxymetholone (DEX+OXY), NFA (DEX+NFA), and FA (DEX+FA) via gavage. Before the end of the experiment, dexamethasone was intraperitoneally (IP) injected into the mice, except in the control group, to induce muscle atrophy. Compared with the DEX group, the DEX+FA group exhibited a significant prevention in the reduction of hindlimb strength, calf thickness, calf muscle weight, and the cross-sectional area of muscle fibers (p < 0.05). The FA-induced improvements in muscle atrophy were associated with a decreased gene expression of protein degradation and growth inhibition, and an increased gene expression of protein synthesis and growth factors. Sialic acid, a bioactive compound associated with muscles, was increased by 51.41% after fermentation and suppressed the expression of protein degradation genes in the C2C12 cells. L. curvatus HY7602-fermented antlers with increased sialic acid after fermentation may therefore be useful for preventing and improving muscle atrophy.
Collapse
|
12
|
Abati E, Manini A, Comi GP, Corti S. Inhibition of myostatin and related signaling pathways for the treatment of muscle atrophy in motor neuron diseases. Cell Mol Life Sci 2022; 79:374. [PMID: 35727341 PMCID: PMC9213329 DOI: 10.1007/s00018-022-04408-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/26/2022]
Abstract
Myostatin is a negative regulator of skeletal muscle growth secreted by skeletal myocytes. In the past years, myostatin inhibition sparked interest among the scientific community for its potential to enhance muscle growth and to reduce, or even prevent, muscle atrophy. These characteristics make it a promising target for the treatment of muscle atrophy in motor neuron diseases, namely, amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), which are rare neurological diseases, whereby the degeneration of motor neurons leads to progressive muscle loss and paralysis. These diseases carry a huge burden of morbidity and mortality but, despite this unfavorable scenario, several therapeutic advancements have been made in the past years. Indeed, a number of different curative therapies for SMA have been approved, leading to a revolution in the life expectancy and outcomes of SMA patients. Similarly, tofersen, an antisense oligonucleotide, is now undergoing clinical trial phase for use in ALS patients carrying the SOD1 mutation. However, these therapies are not able to completely halt or reverse progression of muscle damage. Recently, a trial evaluating apitegromab, a myostatin inhibitor, in SMA patients was started, following positive results from preclinical studies. In this context, myostatin inhibition could represent a useful strategy to tackle motor symptoms in these patients. The aim of this review is to describe the myostatin pathway and its role in motor neuron diseases, and to summarize and critically discuss preclinical and clinical studies of myostatin inhibitors in SMA and ALS. Then, we will highlight promises and pitfalls related to the use of myostatin inhibitors in the human setting, to aid the scientific community in the development of future clinical trials.
Collapse
Affiliation(s)
- Elena Abati
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Neurology Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Arianna Manini
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Giacomo Pietro Comi
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Neurology Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy.
- Neurology Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
13
|
Monotropein Improves Dexamethasone-Induced Muscle Atrophy via the AKT/mTOR/FOXO3a Signaling Pathways. Nutrients 2022; 14:nu14091859. [PMID: 35565825 PMCID: PMC9103778 DOI: 10.3390/nu14091859] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
The present study aimed to investigate the effects of monotropein (MON) on improving dexamethasone (DEX)-induced muscle atrophy in mice and C2C12 mouse skeletal muscle cells. The body weights, grip strengths, and muscle weights of mice were assessed. The histological change in the gastrocnemius tissues was also observed through H&E staining. The expression of myosin heavy chain (MyHC), muscle ring finger 1 (MuRF1), and muscle atrophy F-box (Atrogin1) and the phosphorylation of AKT, mTOR, and FOXO3a in the muscle tissues of mice and C2C12 myotubes were analyzed using Western blotting. MON improved muscle atrophy in mice and C2C12 myotubes by regulating catabolic states via the AKT/mTOR/FOXO3a signaling pathways, and enhanced muscle function by the increases of muscle mass and strength in mice. This suggests that MON could be used for the prevention and treatment of muscle atrophy in patients.
Collapse
|
14
|
Yang HW, Oh S, Chung DM, Seo M, Park SJ, Jeon YJ, Byun K, Ryu B. Ishophloroglucin A, Isolated from Ishige okamurae, Alleviates Dexamethasone-Induced Muscle Atrophy through Muscle Protein Metabolism In Vivo. Mar Drugs 2022; 20:280. [PMID: 35621931 PMCID: PMC9147101 DOI: 10.3390/md20050280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 02/05/2023] Open
Abstract
The in vitro capacity of Ishige okamurae extract (IO) to improve impaired muscle function has been previously examined. However, the mechanism underlying IO-mediated muscle protein metabolism and the role of its component, Ishophloroglucin A (IPA), in mice with dexamethasone (Dexa)-induced muscle atrophy remains unknown. In the present study, we evaluated the effect of IO and IPA supplementation on Dexa-induced muscle atrophy by assessing muscle protein metabolism in gastrocnemius and soleus muscles of mice. IO and IPA supplementation improved the Dexa-induced decrease in muscle weight and width, leading to enhanced grip strength. In addition, IO and IPA supplementation regulated impaired protein synthesis (PI3K and Akt) or degradation (muscle-specific ubiquitin ligase muscle RING finger and atrogin-1) by modulating mRNA levels in gastrocnemius and soleus muscles. Additionally, IO and IPA upregulated mRNA levels associated with muscle growth activation (transient receptor potential vanilloid type 4 and adenosine A1 receptor) or inhibition (myostatin and sirtuin 1) in gastrocnemius and soleus muscle tissues of Dexa-induced mice. Collectively, these results suggest that IO and IO-derived IPA can regulate muscle growth through muscle protein metabolism in Dexa-induced muscle atrophy.
Collapse
Affiliation(s)
- Hye-Won Yang
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, 102 Jejudaehak-ro, Jeju 63243, Korea; (H.-W.Y.); (Y.-J.J.)
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea;
| | - Dong-Min Chung
- Shinwoo Co., Ltd., Jinju 52839, Korea; (D.-M.C.); (M.S.); (S.J.P.)
| | - Minyoung Seo
- Shinwoo Co., Ltd., Jinju 52839, Korea; (D.-M.C.); (M.S.); (S.J.P.)
| | - Shin Jae Park
- Shinwoo Co., Ltd., Jinju 52839, Korea; (D.-M.C.); (M.S.); (S.J.P.)
| | - You-Jin Jeon
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, 102 Jejudaehak-ro, Jeju 63243, Korea; (H.-W.Y.); (Y.-J.J.)
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
| | - Kyunghee Byun
- Functional Cellular Networks Laboratory, Department of Medicine, Graduate School, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Korea;
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Korea
| | - BoMi Ryu
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, 102 Jejudaehak-ro, Jeju 63243, Korea; (H.-W.Y.); (Y.-J.J.)
| |
Collapse
|
15
|
Felice F, Cesare MM, Fredianelli L, De Leo M, Conti V, Braca A, Di Stefano R. Effect of Tomato Peel Extract Grown under Drought Stress Condition in a Sarcopenia Model. Molecules 2022; 27:molecules27082563. [PMID: 35458760 PMCID: PMC9031685 DOI: 10.3390/molecules27082563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 11/16/2022] Open
Abstract
Tomatoes and their derivates represent an important source of natural biologically active components. The present study aims to investigate the protective effect of tomato peel extracts, grown in normal (RED-Ctr) or in drought stress (RED-Ds) conditions, on an experimental model of sarcopenia. The phenolic profile and total polyphenols content (TPC) of RED-Ctr and RED-Ds were determined by Ultra High-Performance Liquid Chromatography (UHPLC) analyses coupled to electrospray ionization high-resolution mass spectrometry (ESI-HR-MS). Human skeletal muscle myoblasts (HSMM) were differentiated in myotubes, and sarcopenia was induced by dexamethasone (DEXA) treatment. Differentiation and sarcopenia were evaluated by both real-time PCR and immunofluorescent techniques. Data show that myosin heavy chain 2 (MYH2), troponin T (TNNT1), and miogenin (MYOG) were expressed in differentiated myotubes. 5 μg Gallic Acid Equivalent (GAE/mL) of TPC from RED-Ds extract significantly reduced muscle atrophy induced by DEXA. Moreover, Forkhead BoxO1 (FOXO1) expression, involved in cell atrophy, was significantly decreased by RED-Ds extract. The protective effect of tomato peel extracts depended on their qualitative polyphenolic composition, resulting effectively in the in vitro model of sarcopenia.
Collapse
Affiliation(s)
- Francesca Felice
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy;
- Correspondence:
| | - Maria Michela Cesare
- Department of Life Sciences, University of Siena, Via P.A. Mattioli 4, 53100 Siena, Italy; (M.M.C.); (V.C.)
| | - Luca Fredianelli
- Institute for Chemical-Physical Processes of the Italian Research Council (CNR-IPCF), Via Moruzzi 1, 56100 Pisa, Italy;
| | - Marinella De Leo
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.L.); (A.B.)
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56100 Pisa, Italy
- CISUP, Centre for Instrumentation Sharing, University of Pisa, 56126 Pisa, Italy
| | - Veronica Conti
- Department of Life Sciences, University of Siena, Via P.A. Mattioli 4, 53100 Siena, Italy; (M.M.C.); (V.C.)
| | - Alessandra Braca
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.L.); (A.B.)
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56100 Pisa, Italy
- CISUP, Centre for Instrumentation Sharing, University of Pisa, 56126 Pisa, Italy
| | - Rossella Di Stefano
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy;
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.L.); (A.B.)
| |
Collapse
|
16
|
Rodgers BD, Ward CW. Myostatin/Activin Receptor Ligands in Muscle and the Development Status of Attenuating Drugs. Endocr Rev 2022; 43:329-365. [PMID: 34520530 PMCID: PMC8905337 DOI: 10.1210/endrev/bnab030] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Indexed: 02/07/2023]
Abstract
Muscle wasting disease indications are among the most debilitating and often deadly noncommunicable disease states. As a comorbidity, muscle wasting is associated with different neuromuscular diseases and myopathies, cancer, heart failure, chronic pulmonary and renal diseases, peripheral neuropathies, inflammatory disorders, and, of course, musculoskeletal injuries. Current treatment strategies are relatively ineffective and can at best only limit the rate of muscle degeneration. This includes nutritional supplementation and appetite stimulants as well as immunosuppressants capable of exacerbating muscle loss. Arguably, the most promising treatments in development attempt to disrupt myostatin and activin receptor signaling because these circulating factors are potent inhibitors of muscle growth and regulators of muscle progenitor cell differentiation. Indeed, several studies demonstrated the clinical potential of "inhibiting the inhibitors," increasing muscle cell protein synthesis, decreasing degradation, enhancing mitochondrial biogenesis, and preserving muscle function. Such changes can prevent muscle wasting in various disease animal models yet many drugs targeting this pathway failed during clinical trials, some from serious treatment-related adverse events and off-target interactions. More often, however, failures resulted from the inability to improve muscle function despite preserving muscle mass. Drugs still in development include antibodies and gene therapeutics, all with different targets and thus, safety, efficacy, and proposed use profiles. Each is unique in design and, if successful, could revolutionize the treatment of both acute and chronic muscle wasting. They could also be used in combination with other developing therapeutics for related muscle pathologies or even metabolic diseases.
Collapse
Affiliation(s)
| | - Christopher W Ward
- Department of Orthopedics and Center for Biomedical Engineering and Technology (BioMET), University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Han S, Cui C, Zhao X, Zhang Y, Zhang Y, Zhao J, Shen X, He H, Wang J, Ma M, Li D, Zhu Q, Yin H. Filamin C regulates skeletal muscle atrophy by stabilizing dishevelled-2 to inhibit autophagy and mitophagy. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 27:147-164. [PMID: 34976434 PMCID: PMC8683659 DOI: 10.1016/j.omtn.2021.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/28/2021] [Indexed: 11/16/2022]
Abstract
FilaminC (Flnc) is a member of the actin binding protein family, which is preferentially expressed in the cardiac and skeletal muscle tissues. Although it is known to interact with proteins associated with myofibrillar myopathy, its unique role in skeletal muscle remains largely unknown. In this study, we identify the biological functions of Flnc in vitro and in vivo using chicken primary myoblast cells and animal models, respectively. From the results, we observe that the growth rate and mass of the skeletal muscle of fast-growing chickens (broilers) were significantly higher than those in slow-growing chickens (layers). Furthermore, we find that the expression of Flnc in the skeletal muscle of broilers was higher than that in the layers. Our results indicated that Flnc was highly expressed in the skeletal muscle, especially in the skeletal muscle of broilers than in layers. This suggests that Flnc plays a positive regulatory role in myoblast development. Flnc knockdown resulted in muscle atrophy, whereas the overexpression of Flnc promotes muscle hypertrophy in vivo in an animal model. We also found that Flnc interacted with dishevelled-2 (Dvl2), activated the wnt/β-catenin signaling pathway, and controlled skeletal muscle development. Flnc also antagonized the LC3-mediated autophagy system by decreasing Dvl2 ubiquitination. Moreover, Flnc knockdown activated and significantly increased mitophagy. In summary, these results indicate that the absence of Flnc induces autophagy or mitophagy and regulates muscle atrophy.
Collapse
Affiliation(s)
- Shunshun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Can Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xiyu Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yao Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Yun Zhang
- College of Management, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jing Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Xiaoxu Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Haorong He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Jianping Wang
- Key Laboratory for Animal Disease Resistance Nutrition of China, Institute of Animal Nutrition, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Menggen Ma
- College of Resources, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Corresponding author Qing Zhu, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| | - Huadong Yin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
- Corresponding author Huadong Yin, Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China.
| |
Collapse
|
18
|
Flis DJ, Bialobrodzka EG, Rodziewicz-Flis EA, Jost Z, Borkowska A, Ziolkowski W, Kaczor JJ. The Effect of Long-Lasting Swimming on Rats Skeletal Muscles Energy Metabolism after Nine Days of Dexamethasone Treatment. Int J Mol Sci 2022; 23:748. [PMID: 35054933 PMCID: PMC8775511 DOI: 10.3390/ijms23020748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 12/04/2022] Open
Abstract
This study investigates the effect of Dexamethasone (Dex) treatment on blood and skeletal muscle metabolites level and skeletal muscle activity of enzymes related to energy metabolism after long-duration swimming. To evaluate whether Dex treatment, swimming, and combining these factors act on analyzed data, rats were randomly divided into four groups: saline treatment non-exercise and exercise and Dex treatment non-exercised and exercised. Animals in both exercised groups underwent long-lasting swimming. The concentration of lipids metabolites, glucose, and lactate were measured in skeletal muscles and blood according to standard colorimetric and fluorimetric methods. Also, activities of enzymes related to aerobic and anaerobic metabolism were measured in skeletal muscles. The results indicated that Dex treatment induced body mass loss and increased lipid metabolites in the rats' blood but did not alter these changes in skeletal muscles. Interestingly, prolonged swimming applied after 9 days of Dex treatment significantly intensified changes induced by Dex; however, there was no difference in skeletal muscle enzymatic activities. This study shows for the first time the cumulative effect of exercise and Dex on selected elements of lipid metabolism, which seems to be essential for the patient's health due to the common use of glucocorticoids like Dex.
Collapse
Affiliation(s)
- Damian Jozef Flis
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, Dębinki 7 Street, 80-211 Gdansk, Poland
| | | | - Ewa Aleksandra Rodziewicz-Flis
- Department of Basic Physiotherapy, Gdansk University of Physical Education and Sport, K. Gorkiego 1 Street, 80-336 Gdansk, Poland;
| | - Zbigniew Jost
- Department of Biochemistry, Gdansk University of Physical Education and Sport, K. Gorkiego 1 Street, 80-336 Gdansk, Poland;
| | - Andzelika Borkowska
- Department of Bioenergetics and Physiology of Exercise, Faculty of Health Sciences, Medical University of Gdansk, Dębinki 1 Street, 80-211 Gdansk, Poland;
| | - Wieslaw Ziolkowski
- Department of Rehabilitation Medicine, Faculty of Health Sciences, Medical University of Gdansk, Al. Zwycięstwa 30, 80-219 Gdansk, Poland;
| | - Jan Jacek Kaczor
- Department of Animal and Human Physiology, University of Gdansk, J. Bazynskiego 8 Street, 80-308 Gdansk, Poland;
| |
Collapse
|
19
|
Griffen C, Duncan M, Hattersley J, Weickert MO, Dallaway A, Renshaw D. Effects of resistance exercise and whey protein supplementation on skeletal muscle strength, mass, physical function, and hormonal and inflammatory biomarkers in healthy active older men: a randomised, double-blind, placebo-controlled trial. Exp Gerontol 2021; 158:111651. [PMID: 34896568 DOI: 10.1016/j.exger.2021.111651] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE To determine the individual and combined effects of 12 weeks of resistance exercise (RE) and whey protein supplementation on skeletal muscle strength (primary outcome), mass and physical function, and hormonal and inflammatory biomarkers in older adults. METHODS Thirty-six healthy older men [(mean±SE) age: 67±1 y; BMI: 25.5±0.4 kg/m2] were randomised to either control (CON; n=9), whey protein (PRO; n=9), RE+control (EX+CON; n=9), or RE+whey protein (EX+PRO; n=9) in a double-blinded fashion. Whole-body RE (2 sets of 8 repetitions and 1 set to volitional failure at 80% 1RM) was performed twice weekly. Supplements (PRO, 25 g whey protein isolate; CON, 23.75 g maltodextrin) were consumed twice daily. RESULTS EX+CON and EX+PRO increased leg extension (+19±3 kg and +20±3 kg, respectively) and leg press 1RM (+27±3 kg and +39±2 kg, respectively) greater than the CON and PRO groups (P<0.001, Cohen's d=1.50-1.90). RE (EX+CON and EX+PRO groups pooled) also increased fat-free mass (FFM) (+0.9±0.3 kg) and 6-min walk test distance (+21±5 m) and decreased fat mass (-0.4±0.4 kg), and interleukin-6 (-1.0±0.4 pg/mL) and tumor necrosis factor-alpha concentration (-0.7±0.3 pg/mL) greater than non-exercise (CON and PRO groups pooled; P<0.05, Cohen's f=0.37-0.45). Whey protein supplementation (PRO and EX+PRO groups pooled) increased 4-m gait speed greater than control (CON and EX+CON groups pooled) (+0.08±0.03 m/s; P=0.007, f=0.51). CONCLUSION RE increased muscle strength, FFM and physical function, and decreased markers of systemic inflammation in healthy active older men. Whey protein supplementation alone increased gait speed. No synergistic effects were observed. This study was registered at clinicaltrials.gov as NCT03299972.
Collapse
Affiliation(s)
- Corbin Griffen
- Centre for Sport, Exercise and Life Sciences, Research Institute of Health and Wellbeing, Coventry University, Coventry, CV1 2DS, United Kingdom; Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom.
| | - Michael Duncan
- Centre for Sport, Exercise and Life Sciences, Research Institute of Health and Wellbeing, Coventry University, Coventry, CV1 2DS, United Kingdom; School of Life Sciences, Faculty of Health and Life Sciences, Coventry University, Coventry, CV1 2DS, United Kingdom
| | - John Hattersley
- Centre for Sport, Exercise and Life Sciences, Research Institute of Health and Wellbeing, Coventry University, Coventry, CV1 2DS, United Kingdom; Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; School of Engineering, University of Warwick, Coventry, CV4 7HL, United Kingdom
| | - Martin O Weickert
- Centre for Sport, Exercise and Life Sciences, Research Institute of Health and Wellbeing, Coventry University, Coventry, CV1 2DS, United Kingdom; Department of Endocrinology and Diabetes, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom; Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom
| | - Alexander Dallaway
- Centre for Sport, Exercise and Life Sciences, Research Institute of Health and Wellbeing, Coventry University, Coventry, CV1 2DS, United Kingdom; Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, United Kingdom
| | - Derek Renshaw
- Centre for Sport, Exercise and Life Sciences, Research Institute of Health and Wellbeing, Coventry University, Coventry, CV1 2DS, United Kingdom
| |
Collapse
|
20
|
Park C, Ji SY, Lee H, Choi SH, Kwon CY, Kim SY, Lee ET, Choo ST, Kim GY, Choi YH, Kim MR. Mori Ramulus Suppresses Hydrogen Peroxide-Induced Oxidative Damage in Murine Myoblast C2C12 Cells through Activation of AMPK. Int J Mol Sci 2021; 22:ijms222111729. [PMID: 34769159 PMCID: PMC8583786 DOI: 10.3390/ijms222111729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 01/25/2023] Open
Abstract
Mori Ramulus, the dried twigs of Morus alba L., has been attracting attention for its potent antioxidant activity, but its role in muscle cells has not yet been elucidated. The purpose of this study was to evaluate the protective effect of aqueous extracts of Mori Ramulus (AEMR) against oxidative stress caused by hydrogen peroxide (H2O2) in C2C12 mouse myoblasts, and in dexamethasone (DEX)-induced muscle atrophied models. Our results showed that AEMR rescued H2O2-induced cell viability loss and the collapse of the mitochondria membrane potential. AEMR was also able to activate AMP-activated protein kinase (AMPK) in H2O2-treated C2C12 cells, whereas compound C, a pharmacological inhibitor of AMPK, blocked the protective effects of AEMR. In addition, H2O2-triggered DNA damage was markedly attenuated in the presence of AEMR, which was associated with the inhibition of reactive oxygen species (ROS) generation. Further studies showed that AEMR inhibited cytochrome c release from mitochondria into the cytoplasm, and Bcl-2 suppression and Bax activation induced by H2O2. Furthermore, AEMR diminished H2O2-induced activation of caspase-3, which was associated with the ability of AEMR to block the degradation of poly (ADP-ribose) polymerase, thereby attenuating H2O2-induced apoptosis. However, compound C greatly abolished the protective effect of AEMR against H2O2-induced C2C12 cell apoptosis, including the restoration of mitochondrial dysfunction. Taken together, these results demonstrate that AEMR could protect C2C12 myoblasts from oxidative damage by maintaining mitochondrial function while eliminating ROS, at least with activation of the AMPK signaling pathway. In addition, oral administration of AEMR alleviated gastrocnemius and soleus muscle loss in DEX-induced muscle atrophied rats. Our findings support that AEMR might be a promising therapeutic candidate for treating oxidative stress-mediated myoblast injury and muscle atrophy.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-Eui University, Busan 47340, Korea;
| | - Seon Yeong Ji
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Korea; (S.Y.J.); (H.L.)
- Anti-Aging Research Center, Dong-Eui University, Busan 47340, Korea
| | - Hyesook Lee
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Korea; (S.Y.J.); (H.L.)
- Anti-Aging Research Center, Dong-Eui University, Busan 47340, Korea
| | - Sung Hyun Choi
- Department of System Management, Korea Lift College, Geochang 50141, Korea;
| | - Chan-Young Kwon
- Department of Oriental Neuropsychiatry, College of Korean Medicine, Dong-Eui University, Busan 47340, Korea;
| | - So Young Kim
- Department of Pharmacology, College of Korean Medicine, Daegu Haany University, Daegu 42158, Korea;
| | - Eun Tag Lee
- Agricultural Corporation, Ebiche Co., Ltd., Yeongcheon 38819, Korea; (E.T.L.); (S.T.C.)
| | - Sung Tae Choo
- Agricultural Corporation, Ebiche Co., Ltd., Yeongcheon 38819, Korea; (E.T.L.); (S.T.C.)
| | - Gi-Young Kim
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea;
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Korea; (S.Y.J.); (H.L.)
- Anti-Aging Research Center, Dong-Eui University, Busan 47340, Korea
- Correspondence: (Y.H.C.); (M.R.K.); Tel.: +82-51-890-3319 (Y.H.C.); +82-53-770-2241 (M.R.K.)
| | - Mi Ryeo Kim
- Department of Pharmacology, College of Korean Medicine, Daegu Haany University, Daegu 42158, Korea;
- Correspondence: (Y.H.C.); (M.R.K.); Tel.: +82-51-890-3319 (Y.H.C.); +82-53-770-2241 (M.R.K.)
| |
Collapse
|
21
|
Chen F, Hao L, Zhu S, Yang X, Shi W, Zheng K, Wang T, Chen H. Potential Adverse Effects of Dexamethasone Therapy on COVID-19 Patients: Review and Recommendations. Infect Dis Ther 2021; 10:1907-1931. [PMID: 34296386 PMCID: PMC8298044 DOI: 10.1007/s40121-021-00500-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
In the context of the coronavirus disease 2019 (COVID-19) pandemic, the global healthcare community has raced to find effective therapeutic agents against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To date, dexamethasone is the first and an important therapeutic to significantly reduce the risk of death in COVID-19 patients with severe disease. Due to powerful anti-inflammatory and immunosuppressive effects, dexamethasone could attenuate SARS-CoV-2-induced uncontrolled cytokine storm, severe acute respiratory distress syndrome and lung injury. Nevertheless, dexamethasone treatment is a double-edged sword, as numerous studies have revealed that it has significant adverse impacts later in life. In this article, we reviewed the literature regarding the adverse effects of dexamethasone administration on different organ systems as well as related disease pathogenesis in an attempt to clarify the potential harms that may arise in COVID-19 patients receiving dexamethasone treatment. Overall, taking the threat of COVID19 pandemic into account, we think it is necessary to apply dexamethasone as a pharmaceutical therapy in critical patients. However, its adverse side effects cannot be ignored. Our review will help medical professionals in the prognosis and follow-up of patients treated with dexamethasone. In addition, given that a considerable amount of uncertainty, confusion and even controversy still exist, further studies and more clinical trials are urgently needed to improve our understanding of the parameters and the effects of dexamethasone on patients with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Fei Chen
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China.
| | - Lanting Hao
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Shiheng Zhu
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Xinyuan Yang
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Wenhao Shi
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Kai Zheng
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Tenger Wang
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| | - Huiran Chen
- Department of Physiology, Jining Medical University, 133 Hehua Rd, Jining, 272067, China
| |
Collapse
|
22
|
Kim S, Kim K, Park J, Jun W. Curcuma longa L. Water Extract Improves Dexamethasone-Induced Sarcopenia by Modulating the Muscle-Related Gene and Oxidative Stress in Mice. Antioxidants (Basel) 2021; 10:1000. [PMID: 34201533 PMCID: PMC8300838 DOI: 10.3390/antiox10071000] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/25/2022] Open
Abstract
Dexamethasone (DEX) promotes proteolysis, which causes muscle atrophy. Muscle atrophy is connected to sarcopenia. We evaluated the effect of Curcuma longa L. water extract (CLW) on DEX-induced muscle atrophy. ICR mice were divided into three groups (eight mice per group) to investigate the capability of CLW in inhibiting muscle atrophy. The control group (Ex-CON) was administered distilled water (DW) by gavage and subjected to exercise; the muscle atrophy group (Ex-DEX) was administered DW by gavage, an injection of DEX (1 mg/kg body weight/day) intraperitoneally (IP), and subjected to exercise; and the treatment group (Ex-CLW) was administered CLW (1 g/kg body weight/day) by gavage, DEX IP injection, and subjected to exercise. Following the injection of DEX, the expression levels of myostatin, MuRF-1, and Atrogin-1 were increased. However, these expression levels were decreased in the Ex-CLW group, thereby leading to the conclusion that CLW inhibits muscle atrophy. ROS (that was overproduced by DEX) decreased antioxidant enzyme activity and increased malondialdehyde (MDA) levels, which led to muscle atrophy. When CLW was ingested, the antioxidant enzyme activities increased while the MDA levels decreased. These findings suggest that CLW could serve as a natural product for the prevention of muscle atrophy by modulating muscle atrophy-related genes and increasing antioxidant potential.
Collapse
Affiliation(s)
- Shintae Kim
- Division of Food and Nutrition, Chonnam National University, Gwangju 61187, Korea;
| | - Kyungmi Kim
- Department of Biofood Analysis, Korea Bio Polytechnic, Ganggyung 32946, Korea;
| | - Jeongjin Park
- Division of Food and Nutrition, Chonnam National University, Gwangju 61187, Korea;
- Research Institute for Human Ecology, Chonnam National University, Gwangju 61187, Korea
| | - Woojin Jun
- Division of Food and Nutrition, Chonnam National University, Gwangju 61187, Korea;
- Research Institute for Human Ecology, Chonnam National University, Gwangju 61187, Korea
| |
Collapse
|
23
|
Zheng Y, Liu T, Li Q, Li J. Integrated analysis of long non-coding RNAs (lncRNAs) and mRNA expression profiles identifies lncRNA PRKG1-AS1 playing important roles in skeletal muscle aging. Aging (Albany NY) 2021; 13:15044-15060. [PMID: 34051073 PMCID: PMC8221296 DOI: 10.18632/aging.203067] [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: 09/24/2019] [Accepted: 04/28/2021] [Indexed: 12/27/2022]
Abstract
This study aimed to identify long non-coding RNAs (lncRNAs) involving in the skeletal muscle aging process. Skeletal muscle samples from old and young subjects were collected for lncRNA-sequencing. Differentially expressed genes (DEGs) and DElncRNAs between young and old groups were identified and a co-expression network was built. Further, a dexamethasone-induced muscle atrophy cell model was established to characterize the function of a critical lncRNA. A total of 424 DEGs, including 271 upregulated genes and 153 downregulated genes as well as 152 DElncRNAs including 76 up-regulated and 76 down-regulated lncRNAs were obtained. Functional analysis demonstrated that the DEGs were significantly related to immune response. Coexpression network demonstrated lncRNA AC004797.1, PRKG1-AS1 and GRPC5D-AS1 were crucial lncRNAs. Their expressions were further validated by qRT-PCR in human skeletal muscle and the muscle atrophy cell model. Further in vitro analysis suggested that knock-down of PRKG1-AS1 could significantly increase cell viability and decrease cell apoptosis. qRT-PCR and western blot analyses demonstrated that knock-down of PRKG1-AS1 could increase the expression of MyoD, MyoG and Mef2c. This study demonstrated that lncRNAs of GPRC5D-AS1, AC004797.1 and PRKG1-AS1 might involve the aging-associated disease processes.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, Jilin, P.R. China
| | - Ting Liu
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, Jilin, P.R. China
| | - Qun Li
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun 130021, Jilin, P.R. China
| | - Jie Li
- Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, Jilin, P.R. China
| |
Collapse
|
24
|
Lee SJ. Targeting the myostatin signaling pathway to treat muscle loss and metabolic dysfunction. J Clin Invest 2021; 131:148372. [PMID: 33938454 DOI: 10.1172/jci148372] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Since the discovery of myostatin (MSTN; also known as GDF-8) as a critical regulator of skeletal muscle mass in 1997, there has been an extensive effort directed at understanding the cellular and physiological mechanisms underlying MSTN activity, with the long-term goal of developing strategies and agents capable of blocking MSTN signaling to treat patients with muscle loss. Considerable progress has been made in elucidating key components of this regulatory system, and in parallel with this effort has been the development of numerous biologics that have been tested in clinical trials for a wide range of indications, including muscular dystrophy, sporadic inclusion body myositis, spinal muscular atrophy, cachexia, muscle loss due to aging or following falls, obesity, and type 2 diabetes. Here, I review what is known about the MSTN regulatory system and the current state of efforts to target this pathway for clinical applications.
Collapse
Affiliation(s)
- Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA.,University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, Connecticut, USA
| |
Collapse
|
25
|
Exercise Reduces the Resumption of Tumor Growth and Proteolytic Pathways in the Skeletal Muscle of Mice Following Chemotherapy. Cancers (Basel) 2020; 12:cancers12113466. [PMID: 33233839 PMCID: PMC7699885 DOI: 10.3390/cancers12113466] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Doxorubicin is a chemotherapeutic agent that contributes to muscle wasting. Based on the evidence that many cancer variants are associated with cachexia and that cancer patients are usually treated with chemotherapeutic agents, it is important to determine strategies to mitigate muscle atrophy. Muscle loss is a poor prognosis during cancer treatment, and exercise has emerged as a potential strategy utilized in this context. Once an ongoing regimen of chemotherapeutic treatment is not always possible, our results demonstrated that continuity of endurance exercise is a potential strategy that can be adopted when chemotherapy needs to be interrupted, minimizing the resumption of tumor growth and avoiding muscle loss. Abstract The pathogenesis of muscle atrophy plays a central role in cancer cachexia, and chemotherapy contributes to this condition. Therefore, the present study aimed to evaluate the effects of endurance exercise on time-dependent muscle atrophy caused by doxorubicin. For this, C57 BL/6 mice were subcutaneously inoculated with Lewis lung carcinoma cells (LLC group). One week after the tumor establishment, a group of these animals initiated the doxorubicin chemotherapy alone (LLC + DOX group) or combined with endurance exercise (LLC + DOX + EXER group). One group of animals was euthanized after the chemotherapy cycle, whereas the remaining animals were euthanized one week after the last administration of doxorubicin. The practice of exercise combined with chemotherapy showed beneficial effects such as a decrease in tumor growth rate after chemotherapy interruption and amelioration of premature death due to doxorubicin toxicity. Moreover, the protein degradation levels in mice undergoing exercise returned to basal levels after chemotherapy; in contrast, the mice treated with doxorubicin alone experienced an increase in the mRNA expression levels of the proteolytic pathways in gastrocnemius muscle (Trim63, Fbxo32, Myostatin, FoxO). Collectively, our results suggest that endurance exercise could be utilized during and after chemotherapy for mitigating muscle atrophy promoted by doxorubicin and avoid the resumption of tumor growth.
Collapse
|
26
|
Gouvêa AL, Martinez CG, Kurtenbach E. Determining Maximal Muscle Strength in Mice: Validity and Reliability of an Adapted Swimming Incremental Overload Test. J Strength Cond Res 2020; 34:2360-2368. [DOI: 10.1519/jsc.0000000000002777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
27
|
Vitamin E Blocks Connexin Hemichannels and Prevents Deleterious Effects of Glucocorticoid Treatment on Skeletal Muscles. Int J Mol Sci 2020; 21:ijms21114094. [PMID: 32521774 PMCID: PMC7312599 DOI: 10.3390/ijms21114094] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 01/06/2023] Open
Abstract
Glucocorticoids are frequently used as anti-inflammatory and immunosuppressive agents. However, high doses and/or prolonged use induce undesired secondary effects such as muscular atrophy. Recently, de novo expression of connexin43 and connexin45 hemichannels (Cx43 HCs and Cx45 HCs, respectively) has been proposed to play a critical role in the mechanism underlying myofiber atrophy induced by dexamethasone (Dex: a synthetic glucocorticoid), but their involvement in specific muscle changes promoted by Dex remains poorly understood. Moreover, treatments that could prevent the undesired effects of glucocorticoids on skeletal muscles remain unknown. In the present work, a 7-day Dex treatment in adult mice was found to induce weight loss and skeletal muscle changes including expression of functional Cx43/Cx45 HCs, elevated atrogin immunoreactivity, atrophy, oxidative stress and mitochondrial dysfunction. All these undesired effects were absent in muscles of mice simultaneously treated with Dex and vitamin E (VitE). Moreover, VitE was found to rapidly inhibit the activity of Cx HCs in freshly isolated myofibers of Dex treated mice. Exposure to alkaline pH induced free radical generation only in HeLa cells expressing Cx43 or Cx45 where Ca2+ was present in the extracellular milieu, response that was prevented by VitE. Besides, VitE and two other anti-oxidant compounds, Tempol and Resveratrol, were found to inhibit Cx43 HCs in HeLa cells transfectants. Thus, we propose that in addition to their intrinsic anti-oxidant potency, some antioxidants could be used to reduce expression and/or opening of Cx HCs and consequently reduce the undesired effect of glucocorticoids on skeletal muscles.
Collapse
|
28
|
A Chalcone from Ashitaba ( Angelica keiskei) Stimulates Myoblast Differentiation and Inhibits Dexamethasone-Induced Muscle Atrophy. Nutrients 2019; 11:nu11102419. [PMID: 31658768 PMCID: PMC6835314 DOI: 10.3390/nu11102419] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 09/30/2019] [Accepted: 10/05/2019] [Indexed: 02/07/2023] Open
Abstract
Ashitaba, Angelica keiskei Koidzumi (AK), as a traditional medicine in Korea, Japan, and China, has been known as an elixir of life having therapeutic potential. However, there is no scientific evidence to support that Ashitaba can enhance or maintain muscle strength. To find a new therapeutic agent from the medicinal plant, we evaluated the anti-myopathy effect of chalcones from ethanol extract of AK (EAK) in cellular and animal models of muscle atrophy. To examine anti-myopathy activity, EAK was treated into dexamethasone injected rats and muscle thickness and histopathological images were analyzed. Oral administration of EAK (250 or 500 mg/kg) alleviated muscle atrophic damages and down-regulated the mRNA levels of muscle-specific ubiquitin-E3 ligases. Among ten compounds isolated from EAK, 4-hydroxyderricin was the most effective principle in stimulating myogenesis of C2C12 myoblasts via activation of p38 mitogen-activated protein kinase (MAPK). In three cellular muscle atrophy models with C2C12 myoblasts damaged by dexamethasone or cancer cell-conditioned medium, 4-hydroxyderricin protected the myosin heavy chain (MHC) degradation through suppressing expressions of MAFbx, MuRF-1 and myostatin. These results suggest that the ethanol extract and its active principle, 4-hydroxyderricin from AK, can overcome the muscle atrophy through double mechanisms of decreasing muscle protein degradation and activating myoblast differentiation.
Collapse
|
29
|
Wragg NM, Mosqueira D, Blokpeol-Ferreras L, Capel A, Player DJ, Martin NRW, Liu Y, Lewis MP. Development of a 3D Tissue-Engineered Skeletal Muscle and Bone Co-culture System. Biotechnol J 2019; 15:e1900106. [PMID: 31468704 DOI: 10.1002/biot.201900106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/05/2019] [Indexed: 12/26/2022]
Abstract
In vitro 3D tissue-engineered (TE) structures have been shown to better represent in vivo tissue morphology and biochemical pathways than monolayer culture, and are less ethically questionable than animal models. However, to create systems with even greater relevance, multiple integrated tissue systems should be recreated in vitro. In the present study, the effects and conditions most suitable for the co-culture of TE skeletal muscle and bone are investigated. High-glucose Dulbecco's modified Eagle medium (HG-DMEM) supplemented with 20% fetal bovine serum followed by HG-DMEM with 2% horse serum is found to enable proliferation of both C2C12 muscle precursor cells and TE85 human osteosarcoma cells, fusion of C2C12s into myotubes, as well as an upregulation of RUNX2/CBFa1 in TE85s. Myotube formation is also evident within indirect contact monolayer cultures. Finally, in 3D co-cultures, TE85 collagen/hydroxyapatite constructs have significantly greater expression of RUNX2/CBFa1 and osteocalcin/BGLAP in the presence of collagen-based C2C12 skeletal muscle constructs; however, fusion within these constructs appears reduced. This work demonstrates the first report of the simultaneous co-culture and differentiation of 3D TE skeletal muscle and bone, and represents a significant step toward a full in vitro 3D musculoskeletal junction model.
Collapse
Affiliation(s)
- Nicholas M Wragg
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Diogo Mosqueira
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Lia Blokpeol-Ferreras
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Andrew Capel
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Darren J Player
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,Institute of Orthopaedics and Musculoskeletal Sciences, RNOH University College London, Stanmore, UK
| | - Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Yang Liu
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
30
|
Wooten H, McGlone JJ, Wachtel M, Thompson G, Rakhshandeh AR, Rakhshandeh A. A glucocorticoid receptor agonist improves post-weaning growth performance in segregated early-weaned pigs. Animal 2019; 13:1972-1981. [PMID: 30626452 DOI: 10.1017/s1751731118003634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
While beneficial for sow reproductive efficiency and biosecurity, segregated early weaning (SEW) leads to a systemic immune response that adversely affects the digestive physiology and post-weaning growth of pigs. Two experiments were conducted to evaluate the effects of a glucocorticoid receptor agonist (GA) on growth performance, measures of immune function and intestinal integrity of SEW pigs. In both experiments, pigs were fed corn-soybean meal-based starter diets. In the first experiment, 48 pigs (initial BW 4.8 ± 0.7 kg) were weaned at 21 ± 1 days and randomly assigned to three GA treatment groups: 0, 0.2 and 0.6 mg GA/kg of BW injected intramuscularly. Treatments were administered one day before weaning. Pigs in the 0 mg GA group received sterile saline in place of GA. Body weight was measured daily from one day before to 7 days post-weaning, and then weekly until 28 days post-weaning. Piglets treated with 0.2 mg GA had a higher BW than piglets in other treatment groups during the 28-day course of the study (P <0.02). To further explore the mechanisms behind this result, a second experiment was performed in which a total of 18 gilts (BW 5.6 ± 0.85 kg) were randomly assigned into three treatment groups: suckling plus saline (UWS), weaned treated with GA (WGA; 0.2 mg GA/kg BW) and weaned plus saline (CON). Treatments were administered one day before and 3 days post-weaning. The WGA and CON groups were weaned at 23 ± 2 days, while the UWS group remained with sow for the duration of the study. Body weight was measured daily and blood plasma was collected at 0, 1, 4 and 5 days post-weaning. All gilts were euthanized 5 days after weaning and jejunum samples were collected for mucosal scrapings, histomorphological analysis and gene expression analysis. Plasma levels of interleukin-1β (IL-1β) and haptoglobin were lower in WGA pigs compared with CON (P <0.02), while plasma total antioxidant capacity was higher in WGA pigs compared with both CON and UWS groups (P <0.01). Relative to CON, GA downregulated IL-18 gene expression in the jejunum, as assessed by both tissue homogenate and mucosal scrapings, but it upregulated claudin-IV gene expression only in the tissue homogenate (P <0.01). These results suggest that GA treatment improves the growth performance of SEW pigs in part by mitigating the negative effects of systemic inflammation. However, the effect of GA on barrier integrity requires further investigation.
Collapse
Affiliation(s)
- H Wooten
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX 79409-2141, USA
| | - J J McGlone
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX 79409-2141, USA
| | - M Wachtel
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - A R Rakhshandeh
- Department of Biology, South Plains College, Levelland, TX 79336, USA
| | - A Rakhshandeh
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, TX 79409-2141, USA
| |
Collapse
|
31
|
Hong Y, Lee JH, Jeong KW, Choi CS, Jun HS. Amelioration of muscle wasting by glucagon-like peptide-1 receptor agonist in muscle atrophy. J Cachexia Sarcopenia Muscle 2019; 10:903-918. [PMID: 31020810 PMCID: PMC6711418 DOI: 10.1002/jcsm.12434] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Skeletal muscle atrophy is defined as a reduction of muscle mass caused by excessive protein degradation. However, the development of therapeutic interventions is still in an early stage. Although glucagon-like peptide-1 receptor (GLP-1R) agonists, such as exendin-4 (Ex-4) and dulaglutide, are widely used for the treatment of diabetes, their effects on muscle pathology are unknown. In this study, we investigated the therapeutic potential of GLP-1R agonist for muscle wasting and the mechanisms involved. METHODS Mouse C2C12 myotubes were used to evaluate the in vitro effects of Ex-4 in the presence or absence of dexamethasone (Dex) on the regulation of the expression of muscle atrophic factors and the underlying mechanisms using various pharmacological inhibitors. In addition, we investigated the in vivo therapeutic effect of Ex-4 in a Dex-induced mouse muscle atrophy model (20 mg/kg/day i.p.) followed by injection of Ex-4 (100 ng/day i.p.) for 12 days and chronic kidney disease (CKD)-induced muscle atrophy model. Furthermore, we evaluated the effect of a long-acting GLP-1R agonist by treatment of dulaglutide (1 mg/kg/week s.c.) for 3 weeks, in DBA/2J-mdx mice, a Duchenne muscular dystrophy model. RESULTS Ex-4 suppressed the expression of myostatin (MSTN) and muscle atrophic factors such as F-box only protein 32 (atrogin-1) and muscle RING-finger protein-1 (MuRF-1) in Dex-treated C2C12 myotubes. The suppression effect was via protein kinase A and protein kinase B signalling pathways through GLP-1R. In addition, Ex-4 treatment inhibited glucocorticoid receptor (GR) translocation by up-regulating the proteins of GR inhibitory complexes. In a Dex-induced muscle atrophy model, Ex-4 ameliorated muscle atrophy by suppressing muscle atrophic factors and enhancing myogenic factors (MyoG and MyoD), leading to increased muscle mass and function. In the CKD muscle atrophy model, Ex-4 also increased muscle mass, myofiber size, and muscle function. In addition, treatment with a long-acting GLP-1R agonist, dulaglutide, recovered muscle mass and function in DBA/2J-mdx mice. CONCLUSIONS GLP-1R agonists ameliorate muscle wasting by suppressing MSTN and muscle atrophic factors and enhancing myogenic factors through GLP-1R-mediated signalling pathways. These novel findings suggest that activating GLP-1R signalling may be useful for the treatment of atrophy-related muscular diseases.
Collapse
Affiliation(s)
- Yeonhee Hong
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Yeonsu-ku, Incheon, Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Jong Han Lee
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Yeonsu-ku, Incheon, Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea
| | - Kwang Won Jeong
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Yeonsu-ku, Incheon, Korea
| | - Cheol Soo Choi
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea.,Gachon Medical Research Institute, Gil Hospital, Incheon, Korea
| | - Hee-Sook Jun
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Yeonsu-ku, Incheon, Korea.,Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Korea.,Gachon Medical Research Institute, Gil Hospital, Incheon, Korea
| |
Collapse
|
32
|
Adhikary S, Choudhary D, Tripathi AK, Karvande A, Ahmad N, Kothari P, Trivedi R. FGF-2 targets sclerostin in bone and myostatin in skeletal muscle to mitigate the deleterious effects of glucocorticoid on musculoskeletal degradation. Life Sci 2019; 229:261-276. [PMID: 31082400 DOI: 10.1016/j.lfs.2019.05.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 10/26/2022]
Abstract
AIM Myokines are associated with regulation of bone and muscle mass. However, limited information is available regarding the impact of myokines on glucocorticoid (GC) mediated adverse effects on the musculoskeletal system. This study investigates the role of myokine fibroblast growth factor-2 (FGF-2) in regulating GC-induced deleterious effects on bone and skeletal muscle. METHODS Primary osteoblast cells and C2C12 myoblast cell line were treated with FGF-2 and then exposed to dexamethasone (GC). FGF-2 mediated attenuation of the inhibitory effect of GC on osteoblast and myoblast differentiation and muscle atrophy was assessed through quantitative PCR and western blot analysis. Further, FGF-2 was administered subcutaneously to dexamethasone treated mice to collect bone and skeletal muscle tissue for in vivo analysis of bone microarchitecture, mechanical strength, histomorphometry and for histological alterations in treated tissue samples. KEY FINDINGS FGF-2 abrogated the dexamethasone induced inhibitory effect on osteoblast differentiation by modulating BMP-2 pathway and inhibiting Wnt antagonist sclerostin. Further, dexamethasone induced atrophy in C2C12 cells was mitigated by FGF-2 as evident from down regulation of atrogenes expression. FGF-2 prevented GC-induced impairment of mineral density, biomechanical strength, trabecular bone volume, cortical thickness and bone formation rate in mice. Additionally, skeletal muscle tissue from GC treated mice displayed weak myostatin immunostaining and reduced expression of atrogenes following FGF-2 treatment. SIGNIFICANCE FGF-2 mitigated GC induced effects through inhibition of sclerostin and myostatin expression in bone and muscle respectively. Taken together, this study exhibited the role of exogenous FGF-2 in sustaining osteoblastogenesis and inhibiting muscle atrophy in presence of glucocorticoid.
Collapse
Affiliation(s)
- Sulekha Adhikary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Dharmendra Choudhary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ashish Kumar Tripathi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anirudha Karvande
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Naseer Ahmad
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Priyanka Kothari
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
33
|
Grade CVC, Mantovani CS, Alvares LE. Myostatin gene promoter: structure, conservation and importance as a target for muscle modulation. J Anim Sci Biotechnol 2019; 10:32. [PMID: 31044074 PMCID: PMC6477727 DOI: 10.1186/s40104-019-0338-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Myostatin (MSTN) is one of the key factors regulating myogenesis. Because of its role as a negative regulator of muscle mass deposition, much interest has been given to its protein and, in recent years, several studies have analysed MSTN gene regulation. This review discusses the MSTN gene promoter, focusing on its structure in several animal species, both vertebrate and invertebrate. We report the important binding sites considering their degree of phylogenetic conservation and roles they play in the promoter activity. Finally, we discuss recent studies focusing on MSTN gene regulation via promoter manipulation and the potential applications they have both in medicine and agriculture.
Collapse
Affiliation(s)
- Carla Vermeulen Carvalho Grade
- 1Universidade Federal da Integração Latino-Americana, UNILA, Instituto Latino-Americano de Ciências da Vida e da Natureza, Avenida Tarquínio Joslin dos Santos, 1000, Foz do Iguaçu, PR CEP 85870-901 Brazil
| | - Carolina Stefano Mantovani
- 2Departamento de Bioquímica e Biologia Tecidual, Universidade Estadual de Campinas - UNICAMP, Rua Monteiro Lobato, 255, Campinas, SP CEP 13083-862 Brazil
| | - Lúcia Elvira Alvares
- 2Departamento de Bioquímica e Biologia Tecidual, Universidade Estadual de Campinas - UNICAMP, Rua Monteiro Lobato, 255, Campinas, SP CEP 13083-862 Brazil
| |
Collapse
|
34
|
Llano-Diez M, Fury W, Okamoto H, Bai Y, Gromada J, Larsson L. RNA-sequencing reveals altered skeletal muscle contraction, E3 ligases, autophagy, apoptosis, and chaperone expression in patients with critical illness myopathy. Skelet Muscle 2019; 9:9. [PMID: 30992050 PMCID: PMC6466682 DOI: 10.1186/s13395-019-0194-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 03/31/2019] [Indexed: 12/17/2022] Open
Abstract
Background Critical illness myopathy (CIM) is associated with severe skeletal muscle wasting and impaired function in intensive care unit (ICU) patients. The mechanisms underlying CIM remain incompletely understood. To elucidate the biological activities occurring at the transcriptional level in the skeletal muscle of ICU patients with CIM, the gene expression profiles, potential upstream regulators, and enrichment pathways were characterized using RNA sequencing (RNA-seq). We also compared the skeletal muscle gene signatures in ICU patients with CIM and genes perturbed by mechanical loading in one leg of the ICU patients, with an aim of reducing the loss of muscle function. Methods RNA-seq was used to assess gene expression changes in tibialis anterior skeletal muscle samples from seven critically ill, immobilized, and mechanically ventilated ICU patients with CIM and matched control subjects. We also examined skeletal muscle gene expression for both legs of six ICU patients with CIM, where one leg was mechanically loaded for 10 h/day for an average of 9 days. Results In total, 6257 of 17,221 detected genes were differentially expressed (84% upregulated; p < 0.05 and fold change ≥ 1.5) in skeletal muscle from ICU patients with CIM when compared to control subjects. The differentially expressed genes were highly associated with gene changes identified in patients with myopathy, sepsis, long-term inactivity, polymyositis, tumor, and repeat exercise resistance. Upstream regulator analysis revealed that the CIM signature could be a result of the activation of MYOD1, p38 MAPK, or treatment with dexamethasone. Passive mechanical loading only reversed expression of 0.74% of the affected genes (46 of 6257 genes). Conclusions RNA-seq analysis revealed that the marked muscle atrophy and weakness observed in ICU patients with CIM were associated with the altered expression of genes involved in muscle contraction, newly identified E3 ligases, autophagy and calpain systems, apoptosis, and chaperone expression. In addition, MYOD1, p38 MAPK, and dexamethasone were identified as potential upstream regulators of skeletal muscle gene expression in ICU patients with CIM. Mechanical loading only marginally affected the skeletal muscle transcriptome profiling of ICU patients diagnosed with CIM. Electronic supplementary material The online version of this article (10.1186/s13395-019-0194-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Monica Llano-Diez
- Department of Physiology and Pharmacology, Karolinska Institutet, Bioclinicum, J8:30, SE-171 77, Stockholm, Sweden
| | - Wen Fury
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Haruka Okamoto
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Yu Bai
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Jesper Gromada
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Lars Larsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Bioclinicum, J8:30, SE-171 77, Stockholm, Sweden. .,Department of Clinical Neuroscience, Karolinska Institutet and Karolinska Hospital, Stockholm, Sweden.
| |
Collapse
|
35
|
Pyropia yezoensis Protein Supplementation Prevents Dexamethasone-Induced Muscle Atrophy in C57BL/6 Mice. Mar Drugs 2018; 16:md16090328. [PMID: 30208614 PMCID: PMC6163250 DOI: 10.3390/md16090328] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/05/2018] [Accepted: 09/09/2018] [Indexed: 12/29/2022] Open
Abstract
We investigated the protective effects of Pyropia yezoensis crude protein (PYCP) against dexamethasone (DEX)-induced myotube atrophy and its underlying mechanisms. DEX (3 mg/kg body weight, intraperitoneal injection) and PYCP (150 and 300 mg/kg body weight, oral) were administrated to mice for 18 days, and the effects of PYCP on DEX-induced muscle atrophy were evaluated. Body weight, calf thickness, and gastrocnemius and tibialis anterior muscle weight were significantly decreased by DEX administration (p < 0.05), while PYCP supplementation effectively prevented the DEX-induced decrease in body weight, calf thickness, and muscle weight. PYCP supplementation also attenuated the DEX-induced increase in serum glucose, creatine kinase, and lactate dehydrogenase levels. Additionally, PYCP supplementation reversed DEX-induced muscle atrophy via the regulation of the insulin-like growth factor-I/protein kinase B/rapamycin-sensitive mTOR complex I/forkhead box O signaling pathway. The mechanistic investigation revealed that PYCP inhibited the ubiquitin-proteasome and autophagy-lysosome pathways in DEX-administrated C57BL/6 mice. These findings demonstrated that PYCP increased protein synthesis and decreased protein breakdown to prevent muscle atrophy. Therefore, PYCP supplementation appears to be useful for preventing muscle atrophy.
Collapse
|
36
|
Larson AA, Syverud BC, Florida SE, Rodriguez BL, Pantelic MN, Larkin LM. Effects of Dexamethasone Dose and Timing on Tissue-Engineered Skeletal Muscle Units. Cells Tissues Organs 2018; 205:197-207. [PMID: 30121672 DOI: 10.1159/000490884] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 06/12/2018] [Indexed: 11/19/2022] Open
Abstract
Our lab showed that administration of dexamethasone (DEX) stimulated myogenesis and resulted in advanced structure in our engineered skeletal muscle units (SMU). While administration of 25 nM DEX resulted in the most advanced structure, 10 nM dosing resulted in the greatest force production. We hypothesized that administration of 25 nM DEX during the entire fabrication process was toxic to the cells and that administration of DEX at precise time points during myogenesis would result in SMU with a more advanced structure and function. Thus, we fabricated SMU with 25 nM DEX administered at early proliferation (days 0-4), late proliferation (days 3-5), and early differentiation (days 5-7) stages of myogenesis and compared them to SMU treated with 10 nM DEX (days 0-16). Cell proliferation was measured with a BrdU assay (day 4) and myogenesis was examined by immunostaining for MyoD (day 4), myogenin (day 7), and α-actinin (day 11). Following SMU formation, isometric tetanic force production was measured. An analysis of cell proliferation indicated that 25 nM DEX administered at early proliferation (days 0-4) provided 21.5% greater myogenic proliferation than 10 nM DEX (days 0-4). In addition, 25 nM DEX administered at early differentiation (days 5-7) showed the highest density of myogenin-positive cells, demonstrating the greatest improvement in differentiation of myoblasts. However, the most advanced sarcomeric structure and the highest force production were exhibited with sustained administration of 10 nM DEX (days 0-16). In conclusion, alteration of the timing of 25 nM DEX administration did not enhance the structure or function of our SMU. SMU were optimally fabricated with sustained administration of 10 nM DEX.
Collapse
Affiliation(s)
- Alexie A Larson
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Brian C Syverud
- Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Shelby E Florida
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Molly N Pantelic
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lisa M Larkin
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.,Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
37
|
Mechanisms of stress-related muscle atrophy in fish: An ex vivo approach. Mech Dev 2018; 154:162-169. [PMID: 29981836 DOI: 10.1016/j.mod.2018.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 06/06/2018] [Accepted: 07/02/2018] [Indexed: 11/21/2022]
|
38
|
Shin K, Ko YG, Jeong J, Kwon H. Fbxw7β is an inducing mediator of dexamethasone-induced skeletal muscle atrophy in vivo with the axis of Fbxw7β-myogenin-atrogenes. Mol Biol Rep 2018; 45:625-631. [PMID: 29671242 DOI: 10.1007/s11033-018-4185-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/02/2018] [Indexed: 12/11/2022]
Abstract
Muscle atrophy is induced by several pathways, e.g., it can be attributed to inherited cachectic symptoms, genetic disorders, sarcopenia, or chronic side effects of treatments. However, the underlying regulatory mechanisms that contribute to muscle atrophy have not been fully elucidated. In this study, we evaluated the role of Fbxw7β, an ubiquitin E3 ligase, in a dexamethasone-induced muscle atrophy model. In this model, endogenous Fbxw7β was up-regulated; furthermore, the Fbxw7β-myogenin-atrogene axis was upregulated, supporting our previous results linking Fbxw7β to muscle atrophy in vitro. Also, muscle atrophy was associated with the Fbxw7β-myogenin-atrogene axis and the down-regulation of Dach2, a repressor of myogenin. Taken together, these results suggest that the ubiquitin E3 ligase Fbxw7β and the Fbxw7β-myogenin-atrogene axis have important roles in a dexamethasone-induced muscle atrophy model in vivo and in vitro. Additionally, the Fbxw7β-Dach2-myogenin-atrogene axis is a potential mechanism underlying muscle atrophy in cases of abnormal Fbxw7β expression-induced muscle atrophy or myogenic degenerative disease.
Collapse
Affiliation(s)
- Kyungshin Shin
- Radiation Molecular Diagnosis Research Team, Korea Institute of Radiological and Medical Science, Seoul, 01812, Republic of Korea.,Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Young-Gyu Ko
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Jaemin Jeong
- Department of Surgery, Hanyang Unviersity College of Medicine, 222 Wangsimni-ro, Seoungdong-gu, Seoul, 04763, Republic of Korea.
| | - Heechung Kwon
- Radiation Molecular Diagnosis Research Team, Korea Institute of Radiological and Medical Science, Seoul, 01812, Republic of Korea. .,Division of Radiation Cancer Center, KIRAMS, 75 Nowon-Gil, Nowon-Gu, Seoul, 01812, Republic of Korea.
| |
Collapse
|
39
|
Lim JM, Lee YJ, Cho HR, Park DC, Jung GW, Ku SK, Choi JS. Extracellular polysaccharides purified from Aureobasidium pullulans SM‑2001 (Polycan) inhibit dexamethasone‑induced muscle atrophy in mice. Int J Mol Med 2018; 41:1245-1264. [PMID: 29138805 PMCID: PMC5819910 DOI: 10.3892/ijmm.2017.3251] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 10/31/2017] [Indexed: 12/21/2022] Open
Abstract
The present study assessed the beneficial skeletal muscle‑preserving effects of extracellular polysaccharides from Aureobasidium pullulans SM‑2001 (Polycan) (EAP) on dexamethasone (DEXA)‑induced catabolic muscle atrophy in mice. To investigate whether EAP prevented catabolic DEXA‑induced muscle atrophy, and to examine its mechanisms of action, EAP (100, 200 and 400 mg/kg) was administered orally, once a day for 24 days. EAP treatment was initiated 2 weeks prior to DEXA treatment (1 mg/kg, once a day for 10 days) in mice. Body weight alterations, serum biochemistry, calf thickness, calf muscle strength, gastrocnemius muscle thickness and weight, gastrocnemius muscle antioxidant defense parameters, gastrocnemius muscle mRNA expression, histology and histomorphometry were subsequently assessed. After 24 days, DEXA control mice exhibited muscle atrophy according to all criteria indices. However, these muscle atrophy symptoms were significantly inhibited by oral treatment with all three doses of EAP. Regarding possible mechanisms of action, EAP exhibited favorable ameliorating effects on DEXA‑induced catabolic muscle atrophy via antioxidant and anti‑inflammatory effects; these effects were mediated by modulation of the expression of genes involved in muscle protein synthesis (AKT serine/threonine kinase 1, phosphatidylinositol 3‑kinase, adenosine A1 receptor and transient receptor potential cation channel subfamily V member 4) and degradation (atrogin‑1, muscle RING‑finger protein‑1, myostatin and sirtuin 1). Therefore, these results indicated that EAP may be helpful in improving muscle atrophies of various etiologies. EAP at 400 mg/kg exhibited favorable muscle protective effects against DEXA‑induced catabolic muscle atrophy, comparable with the effects of oxymetholone (50 mg/kg), which has been used to treat various muscle disorders.
Collapse
Affiliation(s)
- Jong-Min Lim
- Glucan Corporation, #305 Marine Bio-Industry Development Center, Busan 46048
| | | | - Hyung-Rae Cho
- Glucan Corporation, #305 Marine Bio-Industry Development Center, Busan 46048
| | - Dong-Chan Park
- Glucan Corporation, #305 Marine Bio-Industry Development Center, Busan 46048
| | - Go-Woon Jung
- Glucan Corporation, #305 Marine Bio-Industry Development Center, Busan 46048
| | - Sae Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan-si, Gyeongsangbuk-do 38610
| | - Jae-Suk Choi
- Major in Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Busan 46958, Republic of Korea
| |
Collapse
|
40
|
Enhancing tumor response to targeted chemotherapy through up-regulation of folate receptor α expression induced by dexamethasone and valproic acid. J Control Release 2018; 269:36-44. [DOI: 10.1016/j.jconrel.2017.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/01/2017] [Accepted: 11/08/2017] [Indexed: 01/03/2023]
|
41
|
|
42
|
Dasarathy S. Myostatin and beyond in cirrhosis: all roads lead to sarcopenia. J Cachexia Sarcopenia Muscle 2017; 8:864-869. [PMID: 29168629 PMCID: PMC5700432 DOI: 10.1002/jcsm.12262] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Srinivasan Dasarathy
- Professor of Medicine, Cleveland Clinic Lerner College of Medicine; Director, Liver Metabolism Research; Staff, Departments of Gastroenterology, Hepatology and Pathobiology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
43
|
Son YH, Jang EJ, Kim YW, Lee JH. Sulforaphane prevents dexamethasone-induced muscle atrophy via regulation of the Akt/Foxo1 axis in C2C12 myotubes. Biomed Pharmacother 2017; 95:1486-1492. [DOI: 10.1016/j.biopha.2017.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/18/2017] [Accepted: 09/03/2017] [Indexed: 01/06/2023] Open
|
44
|
Duchesne E, Dufresne SS, Dumont NA. Impact of Inflammation and Anti-inflammatory Modalities on Skeletal Muscle Healing: From Fundamental Research to the Clinic. Phys Ther 2017; 97:807-817. [PMID: 28789470 DOI: 10.1093/ptj/pzx056] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 05/12/2017] [Indexed: 02/07/2023]
Abstract
Anti-inflammatory modalities are commonly used for the treatment of various musculoskeletal injuries. Although inflammation was originally believed to interfere with skeletal muscle regeneration, several recent studies have highlighted the beneficial effects of inflammatory cells on muscle healing. This discrepancy is attributable to an evolving understanding of the complex inflammatory process. To better appreciate the paradoxical roles of inflammation, clinicians must have a better comprehension of the fundamental mechanisms regulating the inflammatory response. In this perspective article, cellular, animal, and human studies were analyzed to summarize recent knowledge regarding the impact of inflammation on muscle regeneration in acute or chronic conditions. The effect of anti-inflammatory drugs on the treatment of various muscle injuries was also considered. Overall, this work aims to summarize the current state of the literature on the inflammatory process associated with muscle healing in order to give clinicians the necessary tools to have a more efficient and evidence-based approach to the treatment of muscle injuries and disorders.
Collapse
Affiliation(s)
- Elise Duchesne
- Département des Sciences de la Santé, Université du Québec à Chicoutimi, Saguenay, Quebec, Canada; and Groupe de Recherche Interdisciplinaire sur les Maladies Neuromusculaires, Centre Intégré Universitaire de Santé et de Services Sociaux du Saguenay-Lac-St-Jean, Saguenay, Quebec, Canada
| | - Sébastien S Dufresne
- Département des Sciences de la Santé, Université du Québec à Chicoutimi, Saguenay, Quebec, Canada; and CHU de Québec Research Center, Quebec City, Quebec, Canada; and Faculty of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Nicolas A Dumont
- Musculoskeletal Diseases and Rehabilitation Department, Ste-Justine Hospital Research Center, Montreal, Quebec, Canada; and Department of Rehabilitation, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
45
|
Syverud BC, VanDusen KW, Larkin LM. Growth Factors for Skeletal Muscle Tissue Engineering. Cells Tissues Organs 2016; 202:169-179. [PMID: 27825154 DOI: 10.1159/000444671] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2016] [Indexed: 12/18/2022] Open
Abstract
Tissue-engineered skeletal muscle holds promise as a source of graft tissue for repair of volumetric muscle loss and as a model system for pharmaceutical testing. To reach this potential, engineered tissues must advance past the neonatal phenotype that characterizes the current state of the art. In this review, we describe native skeletal muscle development and identify important growth factors controlling this process. By comparing in vivo myogenesis to in vitro satellite cell cultures and tissue engineering approaches, several key similarities and differences that may potentially advance tissue-engineered skeletal muscle were identified. In particular, hepatocyte and fibroblast growth factors used to accelerate satellite cell activation and proliferation, followed by addition of insulin-like growth factor as a potent inducer of differentiation, are proven methods for increased myogenesis in engineered muscle. Additionally, we review our recent novel application of dexamethasone (DEX), a glucocorticoid that stimulates myoblast differentiation, in skeletal muscle tissue engineering. Using our established skeletal muscle unit (SMU) fabrication protocol, timing- and dose-dependent effects of DEX were measured. The supplemented SMUs demonstrated advanced sarcomeric structure and significantly increased myotube diameter and myotube fusion compared to untreated controls. Most significantly, these SMUs exhibited a fivefold rise in force production. Thus, we concluded that DEX may serve to improve myogenesis, advance muscle structure, and increase force production in engineered skeletal muscle.
Collapse
|
46
|
Rodrigues RWP, Abrahin O, Sousa ECD, Marçal AC. EFEITOS DO EXERCÍCIO RESISTIDO DE ALTA INTENSIDADE EM RATOS QUE RECEBERAM DEXAMETASONA. REV BRAS MED ESPORTE 2016. [DOI: 10.1590/1517-869220162203148503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
RESUMO Introdução: A dexametasona administrada cronicamente promove alterações deletérias no metabolismo dos carboidratos. Objetivo: Avaliar os efeitos do exercício resistido de alta intensidade sobre a sensibilidade à insulina, tolerância à glicose e força muscular de ratos submetidos ao uso crônico de dexametasona. Métodos: Foram utilizados 40 ratos machos divididos randomicamente em quatro grupos: 1) Controle Sedentário (CS); 2) Controle Treinado (CT); 3) Dexametasona Sedentário (DS) e 4) Dexametasona Treinado (DT). O exercício resistido foi realizado em aparelho de agachamento composto por três séries, 10 repetições, com intensidade de 75% de 1 RM durante quatro semanas. Concomitantemente, os grupos DS e DT recebiam diariamente dexametasona intraperitoneal (0,2 g/kg) e os grupos CS e CT recebiam somente solução salina (0,9%). Ao final do protocolo foram realizados testes de tolerância à glicose, sensibilidade à insulina e teste de força máxima. Resultados: Nos grupos treinados (CT e DT) houve aumento da força muscular de 14,78% e 36,87% respectivamente, sem ganho significativo nos grupos sedentários. No teste de tolerância à glicose, os grupos treinados (CT e DT) apresentaram amplitudes atenuadas da glicose plasmática quando comparados aos grupos sedentários (CS e DS). No teste de sensibilidade à insulina, o grupo DT apresentou menor área sob a curva em relação ao grupo DS. Conclusão: O exercício resistido de alta intensidade melhora a sensibilidade à insulina, tolerância à glicose e a força muscular em ratos que receberam a dexametasona.
Collapse
Affiliation(s)
| | - Odilon Abrahin
- Universidade Federal de Sergipe, Brazil; Universidade Federal de Sergipe, Brazil; Universidade do Estado do Pará, Brazil
| | | | | |
Collapse
|
47
|
Wang R, Jiao H, Zhao J, Wang X, Lin H. Glucocorticoids Enhance Muscle Proteolysis through a Myostatin-Dependent Pathway at the Early Stage. PLoS One 2016; 11:e0156225. [PMID: 27227776 PMCID: PMC4882021 DOI: 10.1371/journal.pone.0156225] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 05/11/2016] [Indexed: 01/12/2023] Open
Abstract
Myostatin, a member of the TGF-β superfamily of secreted proteins, is expressed primarily in skeletal muscle. It negatively regulates muscle mass and is associated with glucocorticoid-induced muscle atrophy. However, it remains unclear whether myostatin is involved in glucocorticoid-induced muscle protein turnover. The aim of the present study was to investigate the role of myostatin in protein metabolism during dexamethasone (DEX) treatment. Protein synthesis rates and the expression of the genes for myostatin, ubiquitin-proteasome atrogin-1, MuRF1, FoxO1/3a and mTOR/p70S6K were determined. The results show that DEX decreased (P<0.05) protein synthesis rates while increasing the abundance of myostatin. DEX increased (P<0.05) the level of phospho-FoxO1/3a (Thr 24/32) and the expression of MuRF1. In contrast, DEX treatment had no detectable effect on atrogin-1 protein levels (P>0.05). The phosphorylation levels of mTOR and p70S6K were decreased by DEX treatment (P<0.05). Follistatin treatment inhibited the DEX-induced increase in myostatin (P<0.05) and the activation of phosphor-FoxO1/3a (Thr 24/32) (P< 0.05) and MuRF1 (P<0.05). Follistatin treatment had no influence on the protein synthesis rate or on the phosphorylation levels of mTOR (Ser 2448) and p70S6K (Thr 389) (P> 0.05). In conclusion, the present study suggests that the myostatin signalling pathway is associated with glucocorticoid-induced muscle protein catabolism at the beginning of exposure. Myostatin is not a main pathway associated with the suppression of muscle protein synthesis by glucocorticoids.
Collapse
Affiliation(s)
- Ruxia Wang
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, P. R. China
| | - Hongchao Jiao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, P. R. China
| | - Jingpeng Zhao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, P. R. China
| | - Xiaojuan Wang
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, P. R. China
| | - Hai Lin
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, P. R. China
- * E-mail:
| |
Collapse
|
48
|
Syverud BC, VanDusen KW, Larkin LM. Effects of Dexamethasone on Satellite Cells and Tissue Engineered Skeletal Muscle Units. Tissue Eng Part A 2016; 22:480-9. [PMID: 26790477 DOI: 10.1089/ten.tea.2015.0545] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Tissue engineered skeletal muscle has potential for application as a graft source for repairing soft tissue injuries, a model for testing pharmaceuticals, and a biomechanical actuator system for soft robots. However, engineered muscle to date has not produced forces comparable to native muscle, limiting its potential for repair and for use as an in vitro model for pharmaceutical testing. In this study, we examined the trophic effects of dexamethasone (DEX), a glucocorticoid that stimulates myoblast differentiation and fusion into myotubes, on our tissue engineered three-dimensional skeletal muscle units (SMUs). Using our established SMU fabrication protocol, muscle isolates were cultured with three experimental DEX concentrations (5, 10, and 25 nM) and compared to untreated controls. Following seeding onto a laminin-coated Sylgard substrate, the administration of DEX was initiated on day 0 or day 6 in growth medium or on day 9 after the switch to differentiation medium and was sustained until the completion of SMU fabrication. During this process, total cell proliferation was measured with a BrdU assay, and myogenesis and structural advancement of muscle cells were observed through immunostaining for MyoD, myogenin, desmin, and α-actinin. After SMU formation, isometric tetanic force production was measured to quantify function. The histological and functional assessment of the SMU showed that the administration of 10 nM DEX beginning on either day 0 or day 6 yielded optimal SMUs. These optimized SMUs exhibited formation of advanced sarcomeric structure and significant increases in myotube diameter and myotube fusion index, compared with untreated controls. Additionally, the optimized SMUs matured functionally, as indicated by a fivefold rise in force production. In conclusion, we have demonstrated that the addition of DEX to our process of engineering skeletal muscle tissue improves myogenesis, advances muscle structure, and increases force production in the resulting SMUs.
Collapse
Affiliation(s)
- Brian C Syverud
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | - Keith W VanDusen
- 2 Department of Molecular and Integrated Physiology, University of Michigan , Ann Arbor, Michigan
| | - Lisa M Larkin
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan.,2 Department of Molecular and Integrated Physiology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
49
|
Bodine SC, Furlow JD. Glucocorticoids and Skeletal Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [PMID: 26215994 DOI: 10.1007/978-1-4939-2895-8_7] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glucocorticoids are known to regulate protein metabolism in skeletal muscle, producing a catabolic effect that is opposite that of insulin. In many catabolic diseases, such as sepsis, starvation, and cancer cachexia, endogenous glucocorticoids are elevated contributing to the loss of muscle mass and function. Further, exogenous glucocorticoids are often given acutely and chronically to treat inflammatory conditions such as asthma, chronic obstructive pulmonary disease, and rheumatoid arthritis, resulting in muscle atrophy. This chapter will detail the nature of glucocorticoid-induced muscle atrophy and discuss the mechanisms thought to be responsible for the catabolic effects of glucocorticoids on muscle.
Collapse
Affiliation(s)
- Sue C Bodine
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA,
| | | |
Collapse
|
50
|
Anaya-Segura MA, García-Martínez FA, Montes-Almanza LA, Díaz BG, Avila-Ramírez G, Alvarez-Maya I, Coral-Vazquez RM, Mondragón-Terán P, Escobar-Cedillo RE, García-Calderón N, Vazquez-Cardenas NA, García S, López-Hernandez LB. Non-Invasive Biomarkers for Duchenne Muscular Dystrophy and Carrier Detection. Molecules 2015; 20:11154-72. [PMID: 26091074 PMCID: PMC6272409 DOI: 10.3390/molecules200611154] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 06/02/2015] [Accepted: 06/08/2015] [Indexed: 01/16/2023] Open
Abstract
Non-invasive biological indicators of the absence/presence or progress of the disease that could be used to support diagnosis and to evaluate the effectiveness of treatment are of utmost importance in Duchenne Muscular Dystrophy (DMD). This neuromuscular disorder affects male children, causing weakness and disability, whereas female relatives are at risk of being carriers of the disease. A biomarker with both high sensitivity and specificity for accurate prediction is preferred. Until now creatine kinase (CK) levels have been used for DMD diagnosis but these fail to assess disease progression. Herein we examined the potential applicability of serum levels of matrix metalloproteinase 9 (MMP-9) and matrix metalloproteinase 2 (MMP-2), tissue inhibitor of metalloproteinases 1 (TIMP-1), myostatin (GDF-8) and follistatin (FSTN) as non-invasive biomarkers to distinguish between DMD steroid naïve patients and healthy controls of similar age and also for carrier detection. Our data suggest that serum levels of MMP-9, GDF-8 and FSTN are useful to discriminate DMD from controls (p < 0.05), to correlate with some neuromuscular assessments for DMD, and also to differentiate between Becker muscular dystrophy (BMD) and Limb-girdle muscular dystrophy (LGMD) patients. In DMD individuals under steroid treatment, GDF-8 levels increased as FSTN levels decreased, resembling the proportions of these proteins in healthy controls and also the baseline ratio of patients without steroids. GDF-8 and FSTN serum levels were also useful for carrier detection (p < 0.05). Longitudinal studies with larger cohorts are necessary to confirm that these molecules correlate with disease progression. The biomarkers presented herein could potentially outperform CK levels for carrier detection and also harbor potential for monitoring disease progression.
Collapse
Affiliation(s)
- Monica Alejandra Anaya-Segura
- Research Center in Technology and Design Assistance of Jalisco State (CIATEJ, AC), National Council of Science and Technology (CONACYT), Guadalajara 44270, Mexico.
| | | | - Luis Angel Montes-Almanza
- National Medical Centre \"20 de Noviembre\", Institute for Social Security of State Workers, Mexico City 03100, Mexico.
| | | | | | - Ikuri Alvarez-Maya
- Research Center in Technology and Design Assistance of Jalisco State (CIATEJ, AC), National Council of Science and Technology (CONACYT), Guadalajara 44270, Mexico.
| | - Ramón Mauricio Coral-Vazquez
- Studies Section of Postgraduate and Research, School of Medicine, National Polytechnic Institute, Mexico City 11340, Mexico.
| | - Paul Mondragón-Terán
- National Medical Centre \"20 de Noviembre\", Institute for Social Security of State Workers, Mexico City 03100, Mexico.
| | | | - Noemí García-Calderón
- Asociación de Distrofia Muscular de Occidente A.C., Guadalajara 44380, Mexico.
- Mexican Institute of Social Security-CMNO, Guadalajara 44340, Mexico.
| | | | - Silvia García
- National Medical Centre \"20 de Noviembre\", Institute for Social Security of State Workers, Mexico City 03100, Mexico.
| | - Luz Berenice López-Hernandez
- National Medical Centre \"20 de Noviembre\", Institute for Social Security of State Workers, Mexico City 03100, Mexico.
| |
Collapse
|