1
|
Alonso-Puyo J, Izagirre-Fernandez O, Crende O, Seco-Calvo J, Fernandez-Atutxa A, Fernandez-Lazaro D, Garcia-Gallastegi P, Sanz B. The Non-Linear Profile of Aging: U-Shaped Expression of Myostatin, Follistatin and Intermediate Signals in a Longitudinal In Vitro Murine Cell Sarcopenia Model. Proteomes 2024; 12:34. [PMID: 39585121 PMCID: PMC11587466 DOI: 10.3390/proteomes12040034] [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: 10/16/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024] Open
Abstract
Sarcopenia is linked to the decline in muscle mass, strength and function during aging. It affects the quality and life expectancy and can lead to dependence. The biological process underlying sarcopenia is unclear, but the proteins myostatin and follistatin are involved in the balance between muscle breakdown and synthesis. While myostatin promotes muscle breakdown, follistatin promotes muscle growth, but several works have shown an inconsistent association of these proteins with aging-related parameters in serum of older people. We aimed to know the evolution of these putative sarcopenia biomarkers along muscle aging in an in vitro model. We created and phenotyped a longitudinal murine model (C2C12 cells). Then, we analyzed the protein and genetic expression of myostatin and follistatin as well as the signaling pathway regulators mTOR and RPS6KB1. Myostatin and RPS6KB1 showed a similar tendency in both protein and genetic expression with aging (basal-up-down). Follistatin, on the other hand, shows the opposite tendency (basal-down-up). Regarding mTOR, the tendencies differ when analyzing proteins (basal-up-down) or genes (basal-down-down). Our work demonstrates a U-shape tendency for myostatin and follistatin and for the signaling pathway regulators. These results could be of the utmost importance when designing further research on seeking molecular biomarkers and/or targets for sarcopenia.
Collapse
Affiliation(s)
- Janire Alonso-Puyo
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., 48940 Leioa, Spain; (J.A.-P.); (J.S.-C.); (P.G.-G.)
| | - Oihane Izagirre-Fernandez
- Cell Biology and Histology Department, Basque Country University School of Medicine, Nursery University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., 48940 Leioa, Spain; (O.I.-F.); (O.C.)
| | - Olatz Crende
- Cell Biology and Histology Department, Basque Country University School of Medicine, Nursery University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., 48940 Leioa, Spain; (O.I.-F.); (O.C.)
| | - Jesús Seco-Calvo
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., 48940 Leioa, Spain; (J.A.-P.); (J.S.-C.); (P.G.-G.)
- Institute of Biomedicine (IBIOMED), Universidad de León, Vegazana Universitary Campus, 27071 León, Spain
| | - Ainhoa Fernandez-Atutxa
- Nursery I Department, Basque Country University School of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., 48940 Leioa, Spain;
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Diego Fernandez-Lazaro
- Department of Cellular Biology, Genetics, Histology and Pharmacology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain;
- Neurobiology Research Group, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
| | - Patricia Garcia-Gallastegi
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., 48940 Leioa, Spain; (J.A.-P.); (J.S.-C.); (P.G.-G.)
| | - Begoña Sanz
- Physiology Department, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Barrio Sarriena, sn., 48940 Leioa, Spain; (J.A.-P.); (J.S.-C.); (P.G.-G.)
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| |
Collapse
|
2
|
Salimi K, Alvandi M, Saberi Pirouz M, Rakhshan K, Howatson G. Regulating eEF2 and eEF2K in skeletal muscle by exercise. Arch Physiol Biochem 2024; 130:503-514. [PMID: 36633938 DOI: 10.1080/13813455.2023.2164898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/15/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023]
Abstract
Skeletal muscle is a flexible and adaptable tissue that strongly responds to exercise training. The skeletal muscle responds to exercise by increasing muscle protein synthesis (MPS) when energy is available. One of protein synthesis's major rate-limiting and critical regulatory steps is the translation elongation pathway. The process of translation elongation in skeletal muscle is highly regulated. It requires elongation factors that are intensely affected by various physiological stimuli such as exercise and the total available energy of cells. Studies have shown that exercise involves the elongation pathway by numerous signalling pathways. Since the elongation pathway, has been far less studied than the other translation steps, its comprehensive prospect and quantitative understanding remain in the dark. This study highlights the current understanding of the effect of exercise training on the translation elongation pathway focussing on the molecular factors affecting the pathway, including Ca2+, AMPK, PKA, mTORC1/P70S6K, MAPKs, and myostatin. We further discussed the mode and volume of exercise training intervention on the translation elongation pathway.What is the topic of this review? This review summarises the impacts of exercise training on the translation elongation pathway in skeletal muscle focussing on eEF2 and eEF2K.What advances does it highlight? This review highlights mechanisms and factors that profoundly influence the translation elongation pathway and argues that exercise might modulate the response. This review also combines the experimental observations focussing on the regulation of translation elongation during and after exercise. The findings widen our horizon to the notion of mechanisms involved in muscle protein synthesis (MPS) through translation elongation response to exercise training.
Collapse
Affiliation(s)
- Kia Salimi
- Department of Exercise Physiology, Faculty of Sport and Exercise Sciences, University of Tehran, Tehran, Iran
| | - Masoomeh Alvandi
- Department of Biological Science in Sport and Health, University of Shahid Beheshti, Tehran, Iran
| | - Mahdi Saberi Pirouz
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Kamran Rakhshan
- Department of Medical Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Electrophysiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Glyn Howatson
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
- Water Research Group, North West University, Potchefstroom, South Africa
| |
Collapse
|
3
|
Xiong H, Zhang Y, Zhao Z, Sha Q. Whole-genome SNP allele frequency differences between Tibetan and Large white pigs reveal genes associated with skeletal muscle growth. BMC Genomics 2024; 25:588. [PMID: 38862895 PMCID: PMC11167949 DOI: 10.1186/s12864-024-10508-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND The skeletal muscle growth rate and body size of Tibetan pigs (TIB) are lower than Large white pigs (LW). However, the underlying genetic basis attributing to these differences remains uncertain. To address this knowledge gap, the present study employed whole-genome sequencing of TIB (slow growth) and LW (fast growth) individuals, and integrated with existing NCBI sequencing datasets of TIB and LW individuals, enabling the identification of a comprehensive set of genetic variations for each breed. The specific and predominant SNPs in the TIB and LW populations were detected by using a cutoff value of 0.50 for SNP allele frequency and absolute allele frequency differences (△AF) between the TIB and LW populations. RESULTS A total of 21,767,938 SNPs were retrieved from 44 TIB and 29 LW genomes. The analysis detected 2,893,106 (13.29%) and 813,310 (3.74%) specific and predominant SNPs in the TIB and LW populations, and annotated to 24,560 genes. Further GO analysis revealed 291 genes involved in biological processes related to striated and/or skeletal muscle differentiation, proliferation, hypertrophy, regulation of striated muscle cell differentiation and proliferation, and myoblast differentiation and fusion. These 291 genes included crucial regulators of muscle cell determination, proliferation, differentiation, and hypertrophy, such as members of the Myogenic regulatory factors (MRF) (MYOD, MYF5, MYOG, MYF6) and Myocyte enhancer factor 2 (MEF2) (MEF2A, MEF2C, MEF2D) families, as well as muscle growth inhibitors (MSTN, ACVR1, and SMAD1); KEGG pathway analysis revealed 106 and 20 genes were found in muscle growth related positive and negative regulatory signaling pathways. Notably, genes critical for protein synthesis, such as MTOR, IGF1, IGF1R, IRS1, INSR, and RPS6KA6, were implicated in these pathways. CONCLUSION This study employed an effective methodology to rigorously identify the potential genes associated with skeletal muscle development. A substantial number of SNPs and genes that potentially play roles in the divergence observed in skeletal muscle growth between the TIB and LW breeds were identified. These findings offer valuable insights into the genetic underpinnings of skeletal muscle development and present opportunities for enhancing meat production through pig breeding.
Collapse
Affiliation(s)
- Heli Xiong
- Animal Nutrition and Swine Institute, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, 650224, China.
| | - Yan Zhang
- Animal Nutrition and Swine Institute, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, 650224, China
| | - Zhiyong Zhao
- Animal Nutrition and Swine Institute, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, 650224, China
| | - Qian Sha
- Animal Nutrition and Swine Institute, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, 650224, China
| |
Collapse
|
4
|
Morel J, Pignard AS, Castells J, Allibert V, Hatimi L, Buhot B, Velarde M, Durieux AC, Freyssenet D. Myostatin gene invalidation does not prevent skeletal muscle mass loss during experimental sepsis in mice. J Physiol 2024; 602:2839-2854. [PMID: 38748517 DOI: 10.1113/jp284973] [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: 06/27/2023] [Accepted: 04/26/2024] [Indexed: 06/15/2024] Open
Abstract
Loss of muscle mass and function induced by sepsis contributes to physical inactivity and disability in intensive care unit patients. Limiting skeletal muscle deconditioning may thus be helpful in reducing the long-term effect of muscle wasting in patients. We tested the hypothesis that invalidation of the myostatin gene, which encodes a powerful negative regulator of skeletal muscle mass, could prevent or attenuate skeletal muscle wasting and improve survival of septic mice. Sepsis was induced by caecal ligature and puncture (CLP) in 13-week-old C57BL/6J wild-type and myostatin knock-out male mice. Survival rates were similar in wild-type and myostatin knock-out mice seven days after CLP. Loss in muscle mass was also similar in wild-type and myostatin knock-out mice 4 and 7 days after CLP. The loss in muscle mass was molecularly supported by an increase in the transcript level of E3-ubiquitin ligases and autophagy-lysosome markers. This transcriptional response was blunted in myostatin knock-out mice. No change was observed in the protein level of markers of the anabolic insulin/IGF1-Akt-mTOR pathway. Muscle strength was similarly decreased in wild-type and myostatin knock-out mice 4 and 7 days after CLP. This was associated with a modified expression of genes involved in ion homeostasis and excitation-contraction coupling, suggesting that a long-term functional recovery following experimental sepsis may be impaired by a dysregulated expression of molecular determinants of ion homeostasis and excitation-contraction coupling. In conclusion, myostatin gene invalidation does not provide any benefit in preventing skeletal muscle mass loss and strength in response to experimental sepsis. KEY POINTS: Survival rates are similar in wild-type and myostatin knock-out mice seven days after the induction of sepsis. Loss in muscle mass and muscle strength are similar in wild-type and myostatin knock-out mice 4 and 7 days after the induction of an experimental sepsis. Despite evidence of a transcriptional regulation, the protein level of markers of the anabolic insulin/IGF1-Akt-mTOR pathway remained unchanged. RT-qPCR analysis of autophagy-lysosome pathway markers indicates that activity of the pathway may be altered by experimental sepsis in wild-type and myostatin knock-out mice. Experimental sepsis induces greater variations in the mRNA levels of wild-type mice than those of myostatin knock-out mice, without providing any significant catabolic resistance or functional benefits.
Collapse
Affiliation(s)
- Jérome Morel
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
- Département d'anesthésie et réanimation, Centre Hospitalier Universitaire de Saint Etienne, Saint Etienne, France
| | - Anne Sophie Pignard
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
- Département d'anesthésie et réanimation, Centre Hospitalier Universitaire de Saint Etienne, Saint Etienne, France
| | - Josiane Castells
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Valentine Allibert
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Lahcène Hatimi
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Benjamin Buhot
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Mathias Velarde
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Anne Cécile Durieux
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité, Université Jean Monnet Saint Etienne, Lyon 1, Université Savoie Mont-Blanc, Saint Etienne, France
| |
Collapse
|
5
|
Heitman K, Alexander MS, Faul C. Skeletal Muscle Injury in Chronic Kidney Disease-From Histologic Changes to Molecular Mechanisms and to Novel Therapies. Int J Mol Sci 2024; 25:5117. [PMID: 38791164 PMCID: PMC11121428 DOI: 10.3390/ijms25105117] [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: 04/09/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic kidney disease (CKD) is associated with significant reductions in lean body mass and in the mass of various tissues, including skeletal muscle, which causes fatigue and contributes to high mortality rates. In CKD, the cellular protein turnover is imbalanced, with protein degradation outweighing protein synthesis, leading to a loss of protein and cell mass, which impairs tissue function. As CKD itself, skeletal muscle wasting, or sarcopenia, can have various origins and causes, and both CKD and sarcopenia share common risk factors, such as diabetes, obesity, and age. While these pathologies together with reduced physical performance and malnutrition contribute to muscle loss, they cannot explain all features of CKD-associated sarcopenia. Metabolic acidosis, systemic inflammation, insulin resistance and the accumulation of uremic toxins have been identified as additional factors that occur in CKD and that can contribute to sarcopenia. Here, we discuss the elevation of systemic phosphate levels, also called hyperphosphatemia, and the imbalance in the endocrine regulators of phosphate metabolism as another CKD-associated pathology that can directly and indirectly harm skeletal muscle tissue. To identify causes, affected cell types, and the mechanisms of sarcopenia and thereby novel targets for therapeutic interventions, it is important to first characterize the precise pathologic changes on molecular, cellular, and histologic levels, and to do so in CKD patients as well as in animal models of CKD, which we describe here in detail. We also discuss the currently known pathomechanisms and therapeutic approaches of CKD-associated sarcopenia, as well as the effects of hyperphosphatemia and the novel drug targets it could provide to protect skeletal muscle in CKD.
Collapse
Affiliation(s)
- Kylie Heitman
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Matthew S. Alexander
- Division of Neurology, Department of Pediatrics, The University of Alabama at Birmingham and Children’s of Alabama, Birmingham, AL 35294, USA
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christian Faul
- Division of Nephrology and Section of Mineral Metabolism, Department of Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
6
|
Wu M, Liu C, Sun D. Glucocorticoid-Induced Myopathy: Typology, Pathogenesis, Diagnosis, and Treatment. Horm Metab Res 2024; 56:341-349. [PMID: 38224966 DOI: 10.1055/a-2246-2900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Glucocorticoid-induced myopathy is a non-inflammatory toxic myopathy typified by proximal muscle weakness, muscle atrophy, fatigue, and easy fatigability. These vague symptoms coupled with underlying disorders may mask the signs of glucocorticoid-induced myopathy, leading to an underestimation of the disease's impact. This review briefly summarizes the classification, pathogenesis, and treatment options for glucocorticoid-induced muscle wasting. Additionally, we discuss current diagnostic measures in clinical research and routine care used for diagnosing and monitoring glucocorticoid-induced myopathy, which includes gait speed tests, muscle strength tests, hematologic tests, bioelectrical impedance analysis (BIA), dual-energy X-ray absorptiometry (DXA), computed tomography (CT), magnetic resonance imaging (MRI), electromyography, quantitative muscle ultrasound, histological examination, and genetic analysis. Continuous monitoring of patients receiving glucocorticoid therapy plays an important role in enabling early detection of glucocorticoid-induced myopathy, allowing physicians to modify treatment plans before significant clinical weakness arises.
Collapse
Affiliation(s)
- Mengmeng Wu
- Department of Nephrology, Xuzhou Medical University Affiliated Hospital, Xuzhou, China
- Graduate School, Xuzhou Medical University, Xuzhou, China
| | - Caixia Liu
- Department of Nephrology, Xuzhou Medical University Affiliated Hospital, Xuzhou, China
| | - Dong Sun
- Department of Nephrology, Xuzhou Medical University Affiliated Hospital, Xuzhou, China
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
7
|
Aslam MA, Ma EB, Huh JY. Pathophysiology of sarcopenia: Genetic factors and their interplay with environmental factors. Metabolism 2023; 149:155711. [PMID: 37871831 DOI: 10.1016/j.metabol.2023.155711] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023]
Abstract
Sarcopenia is a geriatric disorder characterized by a progressive decline in muscle mass and function. This disorder has been associated with a range of adverse health outcomes, including fractures, functional deterioration, and increased mortality. The pathophysiology of sarcopenia is highly complex and multifactorial, involving both genetic and environmental factors as key contributors. This review consolidates current knowledge on the genetic factors influencing the pathogenesis of sarcopenia, particularly focusing on the altered gene expression of structural and metabolic proteins, growth factors, hormones, and inflammatory cytokines. While the influence of environmental factors such as physical inactivity, chronic diseases, smoking, alcohol consumption, and sleep disturbances on sarcopenia is relatively well understood, there is a dearth of studies examining their mechanistic roles. Therefore, this review emphasizes the interplay between genetic and environmental factors, elucidating their cumulative role in exacerbating the progression of sarcopenia beyond their individual effects. The unique contribution of this review lies in synthesizing the latest evidence on the genetic factors and their interaction with environmental factors, aiming to inform the development of novel therapeutic or preventive interventions for sarcopenia.
Collapse
Affiliation(s)
- Muhammad Arif Aslam
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Eun Bi Ma
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
8
|
Batsukh S, Oh S, Rheu K, Lee BJ, Choi CH, Son KH, Byun K. Rice Germ Attenuates Chronic Unpredictable Mild Stress-Induced Muscle Atrophy. Nutrients 2023; 15:2719. [PMID: 37375622 DOI: 10.3390/nu15122719] [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: 05/26/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic stress leads to hypothalamic-pituitary-adrenal axis dysfunction, increasing cortisol levels. Glucocorticoids (GCs) promote muscle degradation and inhibit muscle synthesis, eventually causing muscle atrophy. In this study, we aimed to evaluate whether rice germ supplemented with 30% γ-aminobutyric acid (RG) attenuates muscle atrophy in an animal model of chronic unpredictable mild stress (CUMS). We observed that CUMS raised the adrenal gland weight and serum adrenocorticotropic hormone (ACTH) and cortisol levels, and these effects were reversed by RG. CUMS also enhanced the expression of the GC receptor (GR) and GC-GR binding in the gastrocnemius muscle, which were attenuated by RG. The expression levels of muscle degradation-related signaling pathways, such as the Klf15, Redd-1, FoxO3a, Atrogin-1, and MuRF1 pathways, were enhanced by CUMS and attenuated by RG. Muscle synthesis-related signaling pathways, such as the IGF-1/AKT/mTOR/s6k/4E-BP1 pathway, were reduced by CUMS and enhanced by RG. Moreover, CUMS raised oxidative stress by enhancing the levels of iNOS and acetylated p53, which are involved in cell cycle arrest, whereas RG attenuated both iNOS and acetylated p53 levels. Cell proliferation in the gastrocnemius muscle was reduced by CUMS and enhanced by RG. The muscle weight, muscle fiber cross-sectional area, and grip strength were reduced by CUMS and enhanced by RG. Therefore, RG attenuated ACTH levels and cortisol-related muscle atrophy in CUMS animals.
Collapse
Affiliation(s)
- Sosorburam Batsukh
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Seyeon Oh
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| | - Kyoungmin Rheu
- Marine Bioprocess Co., Ltd., Smart Marine BioCenter, Busan 46048, Republic of Korea
| | - Bae-Jin Lee
- Marine Bioprocess Co., Ltd., Smart Marine BioCenter, Busan 46048, Republic of Korea
| | - Chang Hu Choi
- Department of Thoracic and Cardiovascular Surgery, Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gil Medical Center, Gachon University, Incheon 21565, Republic of Korea
| | - Kyunghee Byun
- Department of Anatomy & Cell Biology, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
- Functional Cellular Networks Laboratory, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health & Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
9
|
Guo R, Wang H, Meng C, Gui H, Li Y, Chen F, Zhang C, Zhang H, Ding Q, Zhang J, Zhang J, Qian Y, Zhong J, Cao S. Efficient and Specific Generation of MSTN-Edited Hu Sheep Using C-CRISPR. Genes (Basel) 2023; 14:1216. [PMID: 37372396 DOI: 10.3390/genes14061216] [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: 03/24/2023] [Revised: 05/11/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Hu sheep, an indigenous breed in China known for its high fecundity, are being studied to improve their growth and carcass traits. MSTN is a negative regulator of muscle development, and its inactivation results in muscularity. The C-CRISPR system, utilizing multiple neighboring sgRNAs targeting a key exon, has been successfully used to generate genes for complete knockout (KO) monkeys and mice in one step. In this study, the C-CRISPR system was used to generate MSTN-edited Hu sheep; 70 embryos injected with Cas9 mRNA and four sgRNAs targeting exon 3 of sheep MSTN were transferred to 13 recipients. Out of 10 lambs born from five recipients after full-term pregnancies, nine had complete MSTN KO with various mutations. No off-target effects were found. These MSTN-KO Hu sheep showed a double-muscled (DM) phenotype, characterized by a higher body weight at 3 and 4 months old, prominent muscular protrusion, clearly visible intermuscular groves, and muscle hypertrophy. The molecular analysis indicated enhanced AKT and suppressed ERK1/2 signaling in the gluteus muscle of the edited Hu sheep. In conclusion, MSTN complete KO Hu sheep with a DM phenotype were efficiently and specifically generated using C-CRISPR, and the C-CRISPR method is a promising tool for farm animal breeding.
Collapse
Affiliation(s)
- Rihong Guo
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Huili Wang
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Chunhua Meng
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Hongbing Gui
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
| | - Yinxia Li
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Fang Chen
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Chenjian Zhang
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
| | - Han Zhang
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
| | - Qiang Ding
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jianli Zhang
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jun Zhang
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Yong Qian
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jifeng Zhong
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Shaoxian Cao
- Jiangsu Provincial Engineering Research Center for Precision Animal Breeding, Nanjing 210014, China
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
- Key Laboratory of Crop and Animal Integrated Farming, Ministry of Agriculture and Rural Affairs, Nanjing 210014, China
| |
Collapse
|
10
|
Martin A, Gallot YS, Freyssenet D. Molecular mechanisms of cancer cachexia-related loss of skeletal muscle mass: data analysis from preclinical and clinical studies. J Cachexia Sarcopenia Muscle 2023; 14:1150-1167. [PMID: 36864755 PMCID: PMC10235899 DOI: 10.1002/jcsm.13073] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 06/15/2022] [Accepted: 08/14/2022] [Indexed: 03/04/2023] Open
Abstract
Cancer cachexia is a systemic hypoanabolic and catabolic syndrome that diminishes the quality of life of cancer patients, decreases the efficiency of therapeutic strategies and ultimately contributes to decrease their lifespan. The depletion of skeletal muscle compartment, which represents the primary site of protein loss during cancer cachexia, is of very poor prognostic in cancer patients. In this review, we provide an extensive and comparative analysis of the molecular mechanisms involved in the regulation of skeletal muscle mass in human cachectic cancer patients and in animal models of cancer cachexia. We summarize data from preclinical and clinical studies investigating how the protein turnover is regulated in cachectic skeletal muscle and question to what extent the transcriptional and translational capacities, as well as the proteolytic capacity (ubiquitin-proteasome system, autophagy-lysosome system and calpains) of skeletal muscle are involved in the cachectic syndrome in human and animals. We also wonder how regulatory mechanisms such as insulin/IGF1-AKT-mTOR pathway, endoplasmic reticulum stress and unfolded protein response, oxidative stress, inflammation (cytokines and downstream IL1ß/TNFα-NF-κB and IL6-JAK-STAT3 pathways), TGF-ß signalling pathways (myostatin/activin A-SMAD2/3 and BMP-SMAD1/5/8 pathways), as well as glucocorticoid signalling, modulate skeletal muscle proteostasis in cachectic cancer patients and animals. Finally, a brief description of the effects of various therapeutic strategies in preclinical models is also provided. Differences in the molecular and biochemical responses of skeletal muscle to cancer cachexia between human and animals (protein turnover rates, regulation of ubiquitin-proteasome system and myostatin/activin A-SMAD2/3 signalling pathways) are highlighted and discussed. Identifying the various and intertwined mechanisms that are deregulated during cancer cachexia and understanding why they are decontrolled will provide therapeutic targets for the treatment of skeletal muscle wasting in cancer patients.
Collapse
Affiliation(s)
- Agnès Martin
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| | - Yann S. Gallot
- LBEPS, Univ Evry, IRBA, Université Paris SaclayEvryFrance
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| |
Collapse
|
11
|
Tao R, Stöhr O, Wang C, Qiu W, Copps KD, White MF. Hepatic follistatin increases basal metabolic rate and attenuates diet-induced obesity during hepatic insulin resistance. Mol Metab 2023; 71:101703. [PMID: 36906067 PMCID: PMC10033741 DOI: 10.1016/j.molmet.2023.101703] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
OBJECTIVE Body weight change and obesity follow the variance of excess energy input balanced against tightly controlled EE (energy expenditure). Since insulin resistance can reduce energy storage, we investigated whether genetic disruption of hepatic insulin signaling reduced adipose mass with increased EE. METHODS Insulin signaling was disrupted by genetic inactivation of Irs1 (Insulin receptor substrate 1) and Irs2 in hepatocytes of LDKO mice (Irs1L/L·Irs2L/L·CreAlb), creating a state of complete hepatic insulin resistance. We inactivated FoxO1 or the FoxO1-regulated hepatokine Fst (Follistatin) in the liver of LDKO mice by intercrossing LDKO mice with FoxO1L/L or FstL/L mice. We used DEXA (dual-energy X-ray absorptiometry) to assess total lean mass, fat mass and fat percentage, and metabolic cages to measure EE (energy expenditure) and estimate basal metabolic rate (BMR). High-fat diet was used to induce obesity. RESULTS Hepatic disruption of Irs1 and Irs2 (LDKO mice) attenuated HFD (high-fat diet)-induced obesity and increased whole-body EE in a FoxO1-dependent manner. Hepatic disruption of the FoxO1-regulated hepatokine Fst normalized EE in LDKO mice and restored adipose mass during HFD consumption; moreover, hepatic Fst disruption alone increased fat mass accumulation, whereas hepatic overexpression of Fst reduced HFD-induced obesity. Excess circulating Fst in overexpressing mice neutralized Mstn (Myostatin), activating mTORC1-promoted pathways of nutrient uptake and EE in skeletal muscle. Similar to Fst overexpression, direct activation of muscle mTORC1 also reduced adipose mass. CONCLUSIONS Thus, complete hepatic insulin resistance in LDKO mice fed a HFD revealed Fst-mediated communication between the liver and muscle, which might go unnoticed during ordinary hepatic insulin resistance as a mechanism to increase muscle EE and constrain obesity.
Collapse
Affiliation(s)
- Rongya Tao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Oliver Stöhr
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Caixia Wang
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Wei Qiu
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Kyle D Copps
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Morris F White
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
12
|
Chen M, Lian D, Li Y, Zhao Y, Xu X, Liu Z, Zhang J, Zhang X, Wu S, Qi S, Deng S, Yu K, Lian Z. Global Long Noncoding RNA Expression Profiling of MSTN and FGF5 Double-Knockout Sheep Reveals the Key Gatekeepers of Skeletal Muscle Development. DNA Cell Biol 2023; 42:163-175. [PMID: 36917699 DOI: 10.1089/dna.2022.0574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Improving livestock and poultry growth rates and increasing meat production are urgently needed worldwide. Previously, we produced a myostatin (MSTN) and fibroblast growth factor 5 (FGF5) double-knockout (MF-/-) sheep by CRISPR Cas9 system to improve meat production, and also wool production. Both MF-/- sheep and the F1 generation (MF+/-) sheep showed an obvious "double-muscle" phenotype. In this study, we identified the expression profiles of long noncoding RNAs (lncRNAs) in wild-type and MF+/- sheep, then screened out the key candidate lncRNAs that can regulate myogenic differentiation and skeletal muscle development. These key candidate lncRNAs can serve as critical gatekeepers for muscle contraction, calcium ion transport and skeletal muscle cell differentiation, apoptosis, autophagy, and skeletal muscle inflammation, further revealing that lncRNAs play crucial roles in regulating muscle phenotype in MF+/- sheep. In conclusion, our newly identified lncRNAs may emerge as novel molecules for muscle development or muscle disease and provide a new reference for MSTN-mediated regulation of skeletal muscle development.
Collapse
Affiliation(s)
- Mingming Chen
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Di Lian
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yan Li
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing, China
| | - Yue Zhao
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xueling Xu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhimei Liu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jinlong Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural Sciences, Tianjin, China
| | - Xiaosheng Zhang
- Institute of Animal Husbandry and Veterinary Medicine, Tianjin Academy of Agricultural Sciences, Tianjin, China
| | - Sujun Wu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shiyu Qi
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shoulong Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Kun Yu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhengxing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
13
|
Hanson AM, Young MH, Harrison BC, Zhou X, Han HQ, Stodieck LS, Ferguson VL. Inhibiting myostatin signaling partially mitigates structural and functional adaptations to hindlimb suspension in mice. NPJ Microgravity 2023; 9:2. [PMID: 36646717 PMCID: PMC9842652 DOI: 10.1038/s41526-022-00233-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/12/2022] [Indexed: 01/18/2023] Open
Abstract
Novel treatments for muscle wasting are of significant value to patients with disease states that result in muscle weakness, injury recovery after immobilization and bed rest, and for astronauts participating in long-duration spaceflight. We utilized an anti-myostatin peptibody to evaluate how myostatin signaling contributes to muscle loss in hindlimb suspension. Male C57BL/6 mice were left non-suspended (NS) or were hindlimb suspended (HS) for 14 days and treated with a placebo vehicle (P) or anti-myostatin peptibody (D). Hindlimb suspension (HS-P) resulted in rapid and significantly decreased body mass (-5.6% by day 13) with hindlimb skeletal muscle mass losses between -11.2% and -22.5% and treatment with myostatin inhibitor (HS-D) partially attenuated these losses. Myostatin inhibition increased hindlimb strength with no effect on soleus tetanic strength. Soleus mass and fiber CSA were reduced with suspension and did not increase with myostatin inhibition. In contrast, the gastrocnemius showed histological evidence of wasting with suspension that was partially mitigated with myostatin inhibition. While expression of genes related to protein degradation (Atrogin-1 and Murf-1) in the tibialis anterior increased with suspension, these atrogenes were not significantly reduced by myostatin inhibition despite a modest activation of the Akt/mTOR pathway. Taken together, these findings suggest that myostatin is important in hindlimb suspension but also motivates the study of other factors that contribute to disuse muscle wasting. Myostatin inhibition benefitted skeletal muscle size and function, which suggests therapeutic potential for both spaceflight and terrestrial applications.
Collapse
Affiliation(s)
- Andrea M. Hanson
- grid.266190.a0000000096214564Aerospace Engineering Sciences, BioServe Space Technologies, University of Colorado, Boulder, CO USA
| | - Mary H. Young
- grid.266190.a0000000096214564Aerospace Engineering Sciences, BioServe Space Technologies, University of Colorado, Boulder, CO USA
| | - Brooke C. Harrison
- grid.266190.a0000000096214564Department of Molecular Cellular and Developmental Biology, University of Colorado, Boulder, CO USA
| | - Xiaolan Zhou
- grid.417886.40000 0001 0657 5612Amgen Inc., Thousand Oaks, CA USA ,Present Address: AliveGen USA Inc., Thousand Oaks, CA USA
| | - H. Q. Han
- grid.417886.40000 0001 0657 5612Amgen Inc., Thousand Oaks, CA USA ,Present Address: AliveGen USA Inc., Thousand Oaks, CA USA
| | - Louis S. Stodieck
- grid.266190.a0000000096214564Aerospace Engineering Sciences, BioServe Space Technologies, University of Colorado, Boulder, CO USA
| | - Virginia L. Ferguson
- grid.266190.a0000000096214564Aerospace Engineering Sciences, BioServe Space Technologies, University of Colorado, Boulder, CO USA ,grid.266190.a0000000096214564Department of Mechanical Engineering, University of Colorado, Boulder, CO USA ,grid.266190.a0000000096214564BioFrontiers Institute, University of Colorado, Boulder, CO USA
| |
Collapse
|
14
|
Zumbaugh MD, Johnson SE, Shi TH, Gerrard DE. Molecular and biochemical regulation of skeletal muscle metabolism. J Anim Sci 2022; 100:6652332. [PMID: 35908794 PMCID: PMC9339271 DOI: 10.1093/jas/skac035] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/02/2022] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle hypertrophy is a culmination of catabolic and anabolic processes that are interwoven into major metabolic pathways, and as such modulation of skeletal muscle metabolism may have implications on animal growth efficiency. Muscle is composed of a heterogeneous population of muscle fibers that can be classified by metabolism (oxidative or glycolytic) and contractile speed (slow or fast). Although slow fibers (type I) rely heavily on oxidative metabolism, presumably to fuel long or continuous bouts of work, fast fibers (type IIa, IIx, and IIb) vary in their metabolic capability and can range from having a high oxidative capacity to a high glycolytic capacity. The plasticity of muscle permits continuous adaptations to changing intrinsic and extrinsic stimuli that can shift the classification of muscle fibers, which has implications on fiber size, nutrient utilization, and protein turnover rate. The purpose of this paper is to summarize the major metabolic pathways in skeletal muscle and the associated regulatory pathways.
Collapse
Affiliation(s)
- Morgan D Zumbaugh
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Sally E Johnson
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Tim H Shi
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - David E Gerrard
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
15
|
Swanson DL, Zhang Y, Jimenez AG. Skeletal muscle and metabolic flexibility in response to changing energy demands in wild birds. Front Physiol 2022; 13:961392. [PMID: 35936893 PMCID: PMC9353400 DOI: 10.3389/fphys.2022.961392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 06/29/2022] [Indexed: 12/20/2022] Open
Abstract
Phenotypically plastic responses of animals to adjust to environmental variation are pervasive. Reversible plasticity (i.e., phenotypic flexibility), where adult phenotypes can be reversibly altered according to prevailing environmental conditions, allow for better matching of phenotypes to the environment and can generate fitness benefits but may also be associated with costs that trade-off with capacity for flexibility. Here, we review the literature on avian metabolic and muscle plasticity in response to season, temperature, migration and experimental manipulation of flight costs, and employ an integrative approach to explore the phenotypic flexibility of metabolic rates and skeletal muscle in wild birds. Basal (minimum maintenance metabolic rate) and summit (maximum cold-induced metabolic rate) metabolic rates are flexible traits in birds, typically increasing with increasing energy demands. Because skeletal muscles are important for energy use at the organismal level, especially to maximum rates of energy use during exercise or shivering thermogenesis, we consider flexibility of skeletal muscle at the tissue and ultrastructural levels in response to variations in the thermal environment and in workloads due to flight exercise. We also examine two major muscle remodeling regulatory pathways: myostatin and insulin-like growth factor -1 (IGF-1). Changes in myostatin and IGF-1 pathways are sometimes, but not always, regulated in a manner consistent with metabolic rate and muscle mass flexibility in response to changing energy demands in wild birds, but few studies have examined such variation so additional study is needed to fully understand roles for these pathways in regulating metabolic flexibility in birds. Muscle ultrastrutural variation in terms of muscle fiber diameter and associated myonuclear domain (MND) in birds is plastic and highly responsive to thermal variation and increases in workload, however, only a few studies have examined ultrastructural flexibility in avian muscle. Additionally, the relationship between myostatin, IGF-1, and satellite cell (SC) proliferation as it relates to avian muscle flexibility has not been addressed in birds and represents a promising avenue for future study.
Collapse
Affiliation(s)
- David L. Swanson
- Department of Biology, University of South Dakota, Vermillion, SD, United States
| | - Yufeng Zhang
- College of Health Science, University of Memphis, Memphis, TN, United States
| | - Ana Gabriela Jimenez
- Department of Biology, Colgate University, Hamilton, NY, United States
- *Correspondence: Ana Gabriela Jimenez,
| |
Collapse
|
16
|
Ahmad SS, Ahmad K, Shaikh S, You HJ, Lee EY, Ali S, Lee EJ, Choi I. Molecular Mechanisms and Current Treatment Options for Cancer Cachexia. Cancers (Basel) 2022; 14:cancers14092107. [PMID: 35565236 PMCID: PMC9105812 DOI: 10.3390/cancers14092107] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The primary characteristics of cancer cachexia are weakness, weight loss, atrophy, fat reduction, and systemic inflammation. Cachexia is strongly associated with cancers involving the lungs, pancreas, esophagus, stomach, and liver, which account for half of all cancer deaths. TGF-β, MSTN, activin, IGF-1/PI3K/AKT, and JAK-STAT signaling pathways are known to underlie muscle atrophy and cachexia. Anamorelin (appetite stimulation), megestrol acetate, eicosapentaenoic acid, phytocannabinoids, targeting MSTN/activin, and molecules targeting proinflammatory cytokines, such as TNF-α and IL-6, are being tested as treatment options for cancer cachexia. Abstract Cancer cachexia is a condition marked by functional, metabolic, and immunological dysfunctions associated with skeletal muscle (SM) atrophy, adipose tissue loss, fat reduction, systemic inflammation, and anorexia. Generally, the condition is caused by a variety of mediators produced by cancer cells and cells in tumor microenvironments. Myostatin and activin signaling, IGF-1/PI3K/AKT signaling, and JAK-STAT signaling are known to play roles in cachexia, and thus, these pathways are considered potential therapeutic targets. This review discusses the current state of knowledge of the molecular mechanisms underlying cachexia and the available therapeutic options and was undertaken to increase understanding of the various factors/pathways/mediators involved and to identify potential treatment options.
Collapse
Affiliation(s)
- Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
| | - Hye Jin You
- Tumor Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Gyeonggi-do, Korea; (H.J.Y.); (E.-Y.L.)
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, 323, Ilsan-ro, Ilsandong-gu, Goyaan 10408, Gyeonggi-do, Korea
| | - Eun-Young Lee
- Tumor Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang 10408, Gyeonggi-do, Korea; (H.J.Y.); (E.-Y.L.)
| | - Shahid Ali
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
- Correspondence: (E.J.L.); (I.C.)
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea; (S.S.A.); (K.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Korea;
- Correspondence: (E.J.L.); (I.C.)
| |
Collapse
|
17
|
Hillege MMG, Shi A, Galli RA, Wu G, Bertolino P, Hoogaars WMH, Jaspers RT. Lack of Tgfbr1 and Acvr1b synergistically stimulates myofibre hypertrophy and accelerates muscle regeneration. eLife 2022; 11:77610. [PMID: 35323108 PMCID: PMC9005187 DOI: 10.7554/elife.77610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/05/2022] [Indexed: 12/02/2022] Open
Abstract
In skeletal muscle, transforming growth factor-β (TGF-β) family growth factors, TGF-β1 and myostatin, are involved in atrophy and muscle wasting disorders. Simultaneous interference with their signalling pathways may improve muscle function; however, little is known about their individual and combined receptor signalling. Here, we show that inhibition of TGF-β signalling by simultaneous muscle-specific knockout of TGF-β type I receptors Tgfbr1 and Acvr1b in mice, induces substantial hypertrophy, while such effect does not occur by single receptor knockout. Hypertrophy is induced by increased phosphorylation of Akt and p70S6K and reduced E3 ligases expression, while myonuclear number remains unaltered. Combined knockout of both TGF-β type I receptors increases the number of satellite cells, macrophages and improves regeneration post cardiotoxin-induced injury by stimulating myogenic differentiation. Extra cellular matrix gene expression is exclusively elevated in muscle with combined receptor knockout. Tgfbr1 and Acvr1b are synergistically involved in regulation of myofibre size, regeneration, and collagen deposition.
Collapse
Affiliation(s)
- Michèle M G Hillege
- Department of Human Movement, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Andi Shi
- Department of Human Movement, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ricardo A Galli
- Department of Human Movement, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Philippe Bertolino
- Centre de Recherche en Cancérologie de Lyon, Université de Lyon, UMR INSERM U1052, CNRS 5286, Lyon, France
| | - Willem M H Hoogaars
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Richard T Jaspers
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
18
|
Attwaters M, Hughes SM. Cellular and molecular pathways controlling muscle size in response to exercise. FEBS J 2022; 289:1428-1456. [PMID: 33755332 DOI: 10.1111/febs.15820] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/27/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022]
Abstract
From the discovery of ATP and motor proteins to synaptic neurotransmitters and growth factor control of cell differentiation, skeletal muscle has provided an extreme model system in which to understand aspects of tissue function. Muscle is one of the few tissues that can undergo both increase and decrease in size during everyday life. Muscle size depends on its contractile activity, but the precise cellular and molecular pathway(s) by which the activity stimulus influences muscle size and strength remain unclear. Four correlates of muscle contraction could, in theory, regulate muscle growth: nerve-derived signals, cytoplasmic calcium dynamics, the rate of ATP consumption and physical force. Here, we summarise the evidence for and against each stimulus and what is known or remains unclear concerning their molecular signal transduction pathways and cellular effects. Skeletal muscle can grow in three ways, by generation of new syncytial fibres, addition of nuclei from muscle stem cells to existing fibres or increase in cytoplasmic volume/nucleus. Evidence suggests the latter two processes contribute to exercise-induced growth. Fibre growth requires increase in sarcolemmal surface area and cytoplasmic volume at different rates. It has long been known that high-force exercise is a particularly effective growth stimulus, but how this stimulus is sensed and drives coordinated growth that is appropriately scaled across organelles remains a mystery.
Collapse
Affiliation(s)
- Michael Attwaters
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, UK
| |
Collapse
|
19
|
Chen MM, Zhao YP, Zhao Y, Deng SL, Yu K. Regulation of Myostatin on the Growth and Development of Skeletal Muscle. Front Cell Dev Biol 2022; 9:785712. [PMID: 35004684 PMCID: PMC8740192 DOI: 10.3389/fcell.2021.785712] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/08/2021] [Indexed: 01/01/2023] Open
Abstract
Myostatin (MSTN), a member of the transforming growth factor-β superfamily, can negatively regulate the growth and development of skeletal muscle by autocrine or paracrine signaling. Mutation of the myostatin gene under artificial or natural conditions can lead to a significant increase in muscle quality and produce a double-muscle phenotype. Here, we review the similarities and differences between myostatin and other members of the transforming growth factor-β superfamily and the mechanisms of myostatin self-regulation. In addition, we focus extensively on the regulation of myostatin functions involved in myogenic differentiation, myofiber type conversion, and skeletal muscle protein synthesis and degradation. Also, we summarize the induction of reactive oxygen species generation and oxidative stress by myostatin in skeletal muscle. This review of recent insights into the function of myostatin will provide reference information for future studies of myostatin-regulated skeletal muscle formation and may have relevance to agricultural fields of study.
Collapse
Affiliation(s)
- Ming-Ming Chen
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yi-Ping Zhao
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Yue Zhao
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Shou-Long Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Kun Yu
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
20
|
Bilski J, Pierzchalski P, Szczepanik M, Bonior J, Zoladz JA. Multifactorial Mechanism of Sarcopenia and Sarcopenic Obesity. Role of Physical Exercise, Microbiota and Myokines. Cells 2022; 11:cells11010160. [PMID: 35011721 PMCID: PMC8750433 DOI: 10.3390/cells11010160] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/27/2021] [Accepted: 12/31/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity and ageing place a tremendous strain on the global healthcare system. Age-related sarcopenia is characterized by decreased muscular strength, decreased muscle quantity, quality, and decreased functional performance. Sarcopenic obesity (SO) is a condition that combines sarcopenia and obesity and has a substantial influence on the older adults’ health. Because of the complicated pathophysiology, there are disagreements and challenges in identifying and diagnosing SO. Recently, it has become clear that dysbiosis may play a role in the onset and progression of sarcopenia and SO. Skeletal muscle secretes myokines during contraction, which play an important role in controlling muscle growth, function, and metabolic balance. Myokine dysfunction can cause and aggravate obesity, sarcopenia, and SO. The only ways to prevent and slow the progression of sarcopenia, particularly sarcopenic obesity, are physical activity and correct nutritional support. While exercise cannot completely prevent sarcopenia and age-related loss in muscular function, it can certainly delay development and slow down the rate of sarcopenia. The purpose of this review was to discuss potential pathways to muscle deterioration in obese individuals. We also want to present the current understanding of the role of various factors, including microbiota and myokines, in the process of sarcopenia and SO.
Collapse
Affiliation(s)
- Jan Bilski
- Department of Biomechanics and Kinesiology, Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-008 Krakow, Poland
- Correspondence: ; Tel.: +48-12-421-93-51
| | - Piotr Pierzchalski
- Department of Medical Physiology, Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-126 Krakow, Poland; (P.P.); (J.B.)
| | - Marian Szczepanik
- Department of Medical Biology, Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-034 Krakow, Poland;
| | - Joanna Bonior
- Department of Medical Physiology, Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-126 Krakow, Poland; (P.P.); (J.B.)
| | - Jerzy A. Zoladz
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, 31-066 Krakow, Poland;
| |
Collapse
|
21
|
Esposito P, Picciotto D, Battaglia Y, Costigliolo F, Viazzi F, Verzola D. Myostatin: Basic biology to clinical application. Adv Clin Chem 2022; 106:181-234. [PMID: 35152972 DOI: 10.1016/bs.acc.2021.09.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Myostatin is a member of the transforming growth factor (TGF)-β superfamily. It is expressed by animal and human skeletal muscle cells where it limits muscle growth and promotes protein breakdown. Its effects are influenced by complex mechanisms including transcriptional and epigenetic regulation and modulation by extracellular binding proteins. Due to its actions in promoting muscle atrophy and cachexia, myostatin has been investigated as a promising therapeutic target to counteract muscle mass loss in experimental models and patients affected by different muscle-wasting conditions. Moreover, growing evidence indicates that myostatin, beyond to regulate skeletal muscle growth, may have a role in many physiologic and pathologic processes, such as obesity, insulin resistance, cardiovascular and chronic kidney disease. In this chapter, we review myostatin biology, including intracellular and extracellular regulatory pathways, and the role of myostatin in modulating physiologic processes, such as muscle growth and aging. Moreover, we discuss the most relevant experimental and clinical evidence supporting the extra-muscle effects of myostatin. Finally, we consider the main strategies developed and tested to inhibit myostatin in clinical trials and discuss the limits and future perspectives of the research on myostatin.
Collapse
Affiliation(s)
- Pasquale Esposito
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Daniela Picciotto
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Yuri Battaglia
- Nephrology and Dialysis Unit, St. Anna University Hospital, Ferrara, Italy
| | - Francesca Costigliolo
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Viazzi
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniela Verzola
- Clinica Nefrologica, Dialisi, Trapianto, Department of Internal Medicine, University of Genoa and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
22
|
Nassar R, Vernus B, Carnac G, Fouret G, Goustard B, Casas F, Tintignac L, Cassar-Malek I, Picard B, Seiliez I, Brioche T, Koechlin-Ramonatxo C, Bertrand-Gaday C, Hamade A, Najjar F, Chabi B, Bonnieu A. Myostatin gene inactivation increases post-mortem calpain-dependent muscle proteolysis in mice. Meat Sci 2021; 185:108726. [PMID: 34973590 DOI: 10.1016/j.meatsci.2021.108726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 12/13/2022]
Abstract
Myostatin deficiency leads to extensive skeletal muscle hypertrophy, but its consequence on post-mortem muscle proteolysis is unknown. Here, we compared muscle myofibrillar protein degradation, and autophagy, ubiquitin-proteasome and Ca2+-dependent proteolysis relative to the energetic and redox status in wild-type (WT) and myostatin knock-out mice (KO) during early post-mortem storage. KO muscles showed higher degradation of myofibrillar proteins in the first 24 h after death, associated with preserved antioxidant status, compared with WT muscles. Analysis of key autophagy and ubiquitin-proteasome system markers indicated that these two pathways were not upregulated in post-mortem muscle (both genotypes), but basal autophagic flux and ATP content were lower in KO muscles. Proteasome and caspase activities were not different between WT and KO mice. Conversely, calpain activity was higher in KO muscles, concomitantly with higher troponin T and desmin degradation. Altogether, these results suggest that calpains but not the autophagy, proteasome and caspase systems, explain the difference in post-mortem muscle protein proteolysis between both genotypes.
Collapse
Affiliation(s)
- Rim Nassar
- DMEM, University of Montpellier, INRAE, Montpellier, France; Laboratoire d'Innovation thérapeutique, Lebanese University, Beyrouth, Liban
| | - Barbara Vernus
- DMEM, University of Montpellier, INRAE, Montpellier, France
| | - Gilles Carnac
- PHYMEDEXP, University of Montpellier, CNRS, INSERM, CHRU, Montpellier, France
| | - Gilles Fouret
- DMEM, University of Montpellier, INRAE, Montpellier, France
| | | | - François Casas
- DMEM, University of Montpellier, INRAE, Montpellier, France
| | - Lionel Tintignac
- Département de Biomédecine, Basel University, Basel, Switzerland
| | - Isabelle Cassar-Malek
- University Clermont Auvergne, INRAE, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France
| | - Brigitte Picard
- University Clermont Auvergne, INRAE, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France
| | - Iban Seiliez
- Université de Pau et des Pays de l'Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, F-64310 Saint-Pée-sur-Nivelle, France
| | - Thomas Brioche
- DMEM, University of Montpellier, INRAE, Montpellier, France
| | | | | | - Aline Hamade
- Laboratoire d'Innovation thérapeutique, Lebanese University, Beyrouth, Liban
| | - Fadia Najjar
- Laboratoire d'Innovation thérapeutique, Lebanese University, Beyrouth, Liban
| | - Béatrice Chabi
- DMEM, University of Montpellier, INRAE, Montpellier, France
| | - Anne Bonnieu
- DMEM, University of Montpellier, INRAE, Montpellier, France.
| |
Collapse
|
23
|
Masuzawa R, Takahashi K, Takano K, Nishino I, Sakai T, Endo T. DA-Raf and the MEK inhibitor trametinib reverse skeletal myocyte differentiation inhibition or muscle atrophy caused by myostatin and GDF11 through the non-Smad Ras-ERK pathway. J Biochem 2021; 171:109-122. [PMID: 34676394 DOI: 10.1093/jb/mvab116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/18/2021] [Indexed: 12/25/2022] Open
Abstract
Myostatin (Mstn) and GDF11 are critical factors that are involved in muscle atrophy in the young and sarcopenia in the elderly, respectively. These TGF-β superfamily proteins activate not only Smad signaling but also non-Smad signaling including the Ras-mediated ERK pathway (Raf-MEK-ERK phosphorylation cascade). Although Mstn and GDF11 have been shown to induce muscle atrophy or sarcopenia by Smad2/3-mediated Akt inhibition, participation of the non-Smad Ras-ERK pathway in atrophy and sarcopenia has not been well determined. We show here that both Mstn and GDF11 prevented skeletal myocyte differentiation but that the MEK inhibitor U0126 or trametinib restored differentiation in Mstn- or GDF11-treated myocytes. These MEK inhibitors induced the expression of DA-Raf1 (DA-Raf), which is a dominant-negative antagonist of the Ras-ERK pathway. Exogenous expression of DA-Raf in Mstn- or GDF11-treated myocytes restored differentiation. Furthermore, administration of trametinib to aged mice resulted in an increase in myofiber size, or recovery from muscle atrophy. The trametinib administration downregulated ERK activity in these muscles. These results imply that the Mstn/GDF11-induced Ras-ERK pathway plays critical roles in the inhibition of myocyte differentiation and muscle regeneration, which leads to muscle atrophy. Trametinib and similar approved drugs might be applicable to the treatment of muscle atrophy in sarcopenia or cachexia.
Collapse
Affiliation(s)
- Ryuichi Masuzawa
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan
| | - Kazuya Takahashi
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan
| | - Kazunori Takano
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8502, Japan
| | - Toshiyuki Sakai
- Drug Discovery Center and Department of Drug Discovery Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan
| |
Collapse
|
24
|
Sun Z, Xu D, Zhao L, Li X, Li S, Huang X, Li C, Sun L, Liu B, Jiang Z, Zhang L. A new therapeutic effect of fenofibrate in Duchenne muscular dystrophy: The promotion of myostatin degradation. Br J Pharmacol 2021; 179:1237-1250. [PMID: 34553378 DOI: 10.1111/bph.15678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/14/2021] [Accepted: 08/19/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Duchenne muscular dystrophy (DMD) is a degenerative muscle disease with no effective drug treatment. This study investigated the positive effects of fenofibrate on dystrophic muscles. EXPERIMENTAL APPROACH Myostatin expression in serum and muscle tissue of DMD patients and mdx mice were tested. Primary myoblasts isolated from mdx mice were challenged with an inflammatory stimulus and treated with fenofibrate. In animal experiments, 6-week-old male mdx mice were treated with fenofibrate (100 mg/kg) administered orally once per day for 6 weeks. Tests of muscle function plus histology and biochemical analyses of serum were conducted to evaluate the effects of fenofibrate. The expressions of myostatin, MuRF1, and atrogin-1 in skeletal muscle were evaluated by Western blotting and real-time PCR. Total and oxidative myosin heavy chain (MHC) were assessed via immunofluorescence. KEY RESULTS Increased expression of myostatin protein was found in dystrophic muscle of DMD patients and mdx mice. Fenofibrate enhanced myofibre differentiation by downregulating the expression of myostatin protein but not mRNA in primary myoblasts of mdx mice. Fenofibrate significantly improved muscle function while ameliorating muscle damage in mdx mice. These benefits are accompanied by an anti-inflammatory effect. Fenofibrate treatment returned myofibre function by inhibiting the expressions of myostatin, MuRF1, and atrogin-1 protein in the gastrocnemius muscle and diaphragm, while leaving the mRNA level of myostatin unaffected. CONCLUSIONS AND IMPLICATIONS Fenofibrate substantially slows muscle dystrophy by promoting the degradation of myostatin protein, which may indicate a new therapeutic focus for DMD patients.
Collapse
Affiliation(s)
- Zeren Sun
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Dengqiu Xu
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Lei Zhao
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Xihua Li
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Sijia Li
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Xiaofei Huang
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Chunjie Li
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Bing Liu
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China.,Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
25
|
Haberecht-Müller S, Krüger E, Fielitz J. Out of Control: The Role of the Ubiquitin Proteasome System in Skeletal Muscle during Inflammation. Biomolecules 2021; 11:biom11091327. [PMID: 34572540 PMCID: PMC8468834 DOI: 10.3390/biom11091327] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 02/07/2023] Open
Abstract
The majority of critically ill intensive care unit (ICU) patients with severe sepsis develop ICU-acquired weakness (ICUAW) characterized by loss of muscle mass, reduction in myofiber size and decreased muscle strength leading to persisting physical impairment. This phenotype results from a dysregulated protein homeostasis with increased protein degradation and decreased protein synthesis, eventually causing a decrease in muscle structural proteins. The ubiquitin proteasome system (UPS) is the predominant protein-degrading system in muscle that is activated during diverse muscle atrophy conditions, e.g., inflammation. The specificity of UPS-mediated protein degradation is assured by E3 ubiquitin ligases, such as atrogin-1 and MuRF1, which target structural and contractile proteins, proteins involved in energy metabolism and transcription factors for UPS-dependent degradation. Although the regulation of activity and function of E3 ubiquitin ligases in inflammation-induced muscle atrophy is well perceived, the contribution of the proteasome to muscle atrophy during inflammation is still elusive. During inflammation, a shift from standard- to immunoproteasome was described; however, to which extent this contributes to muscle wasting and whether this changes targeting of specific muscular proteins is not well described. This review summarizes the function of the main proinflammatory cytokines and acute phase response proteins and their signaling pathways in inflammation-induced muscle atrophy with a focus on UPS-mediated protein degradation in muscle during sepsis. The regulation and target-specificity of the main E3 ubiquitin ligases in muscle atrophy and their mode of action on myofibrillar proteins will be reported. The function of the standard- and immunoproteasome in inflammation-induced muscle atrophy will be described and the effects of proteasome-inhibitors as treatment strategies will be discussed.
Collapse
Affiliation(s)
- Stefanie Haberecht-Müller
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, 17475 Greifswald, Germany;
- Correspondence: (E.K.); (J.F.)
| | - Jens Fielitz
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, 17475 Greifswald, Germany
- Correspondence: (E.K.); (J.F.)
| |
Collapse
|
26
|
Singh A, Yadav A, Phogat J, Dabur R. Dynamics of autophagy and ubiquitin proteasome system coordination and interplay in skeletal muscle atrophy. Curr Mol Pharmacol 2021; 15:475-486. [PMID: 34365963 DOI: 10.2174/1874467214666210806163851] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/26/2021] [Accepted: 04/05/2021] [Indexed: 11/22/2022]
Abstract
Skeletal muscles are considered the largest reservoirs of the protein pool in the body and are critical for the maintenances of body homeostasis. Skeletal muscle atrophy is supported by various physiopathological conditions that lead to loss of muscle mass and contractile capacity of the skeletal muscle. Lysosomal mediated autophagy and ubiquitin-proteasomal system (UPS) concede the major intracellular systems of muscle protein degradation that result in the loss of mass and strength. Both systems recognize ubiquitination as a signal of degradation through different mechanisms, a sign of dynamic interplay between systems. Hence, growing shreds of evidence suggest the interdependency of autophagy and UPS in the progression of skeletal muscle atrophy under various pathological conditions. Therefore, understanding the molecular dynamics as well associated factors responsible for their interdependency is a necessity for the new therapeutic insights to counteract the muscle loss. Based on current literature, the present review summarizes the factors interplay in between the autophagy and UPS in favor of enhanced proteolysis of skeletal muscle and how they affect the anabolic signaling pathways under various conditions of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Ajay Singh
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Aarti Yadav
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Jatin Phogat
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| | - Rajesh Dabur
- Clinical Biochemistry Laboratory, Department of Biochemistry, Maharshi Dayanand University, Rohtak-124001, Haryana. India
| |
Collapse
|
27
|
Random errors in protein synthesis activate an age-dependent program of muscle atrophy in mice. Commun Biol 2021; 4:703. [PMID: 34103648 PMCID: PMC8187632 DOI: 10.1038/s42003-021-02204-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 05/12/2021] [Indexed: 12/13/2022] Open
Abstract
Random errors in protein synthesis are prevalent and ubiquitous, yet their effect on organismal health has remained enigmatic for over five decades. Here, we studied whether mice carrying the ribosomal ambiguity (ram) mutation Rps2-A226Y, recently shown to increase the inborn error rate of mammalian translation, if at all viable, present any specific, possibly aging-related, phenotype. We introduced Rps2-A226Y using a Cre/loxP strategy. Resulting transgenic mice were mosaic and showed a muscle-related phenotype with reduced grip strength. Analysis of gene expression in skeletal muscle using RNA-Seq revealed transcriptomic changes occurring in an age-dependent manner, involving an interplay of PGC1α, FOXO3, mTOR, and glucocorticoids as key signaling pathways, and finally resulting in activation of a muscle atrophy program. Our results highlight the relevance of translation accuracy, and show how disturbances thereof may contribute to age-related pathologies. By introducing a ribosomal ambiguity mutation into mice, Moore et al. establish an in-vivo model to investigate how age-related diseases are related to decreasing accuracy in protein synthesis. Their findings potentially offer new insights into the pathological changes observed in age-related diseases, such as muscle atrophy
Collapse
|
28
|
Yoon JH, Kwon KS. Receptor-Mediated Muscle Homeostasis as a Target for Sarcopenia Therapeutics. Endocrinol Metab (Seoul) 2021; 36:478-490. [PMID: 34218646 PMCID: PMC8258343 DOI: 10.3803/enm.2021.1081] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/13/2021] [Accepted: 05/15/2021] [Indexed: 12/19/2022] Open
Abstract
Sarcopenia is a disease characterized by age-related decline of skeletal muscle mass and function. The molecular mechanisms of the pathophysiology of sarcopenia form a complex network due to the involvement of multiple interconnected signaling pathways. Therefore, signaling receptors are major targets in pharmacological strategies in general. To provide a rationale for pharmacological interventions for sarcopenia, we herein describe several druggable signaling receptors based on their role in skeletal muscle homeostasis and changes in their activity with aging. A brief overview is presented of the efficacy of corresponding drug candidates under clinical trials. Strategies targeting the androgen receptor, vitamin D receptor, Insulin-like growth factor-1 receptor, and ghrelin receptor primarily focus on promoting anabolic action using natural ligands or mimetics. Strategies involving activin receptors and angiotensin receptors focus on inhibiting catabolic action. This review may help to select specific targets or combinations of targets in the future.
Collapse
Affiliation(s)
- Jong Hyeon Yoon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Korea
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Korea
- Aventi Inc., Daejeon, Korea
| |
Collapse
|
29
|
Myostatin Inhibition-Induced Increase in Muscle Mass and Strength Was Amplified by Resistance Exercise Training, and Dietary Essential Amino Acids Improved Muscle Quality in Mice. Nutrients 2021; 13:nu13051508. [PMID: 33947024 PMCID: PMC8146053 DOI: 10.3390/nu13051508] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/16/2022] Open
Abstract
It has been frequently reported that myostatin inhibition increases muscle mass, but decreases muscle quality (i.e., strength/muscle mass). Resistance exercise training (RT) and essential amino acids (EAAs) are potent anabolic stimuli that synergistically increase muscle mass through changes in muscle protein turnover. In addition, EAAs are known to stimulate mitochondrial biogenesis. We have investigated if RT amplifies the anabolic potential of myostatin inhibition while EAAs enhance muscle quality through stimulations of mitochondrial biogenesis and/or muscle protein turnover. Mice were assigned into ACV (myostatin inhibitor), ACV+EAA, ACV+RT, ACV+EAA +RT, or control (CON) over 4 weeks. RT, but not EAA, increased muscle mass above ACV. Despite differences in muscle mass gain, myofibrillar protein synthesis was stimulated similarly in all vs. CON, suggesting a role for changes in protein breakdown in muscle mass gains. There were increases in MyoD expression but decreases in Atrogin-1/MAFbx expression in ACV+EAA, ACV+RT, and ACV+EAA+RT vs. CON. EAA increased muscle quality (e.g., grip strength and maximal carrying load) without corresponding changes in markers of mitochondrial biogenesis and neuromuscular junction stability. In conclusion, RT amplifies muscle mass and strength through changes in muscle protein turnover in conjunction with changes in implicated signaling, while EAAs enhance muscle quality through unknown mechanisms.
Collapse
|
30
|
Kim DH, Choi YM, Suh Y, Shin S, Lee J, Hwang S, Lee SS, Lee K. Research Note: Increased myostatin expression and decreased expression of myogenic regulatory factors in embryonic ages in a quail line with muscle hypoplasia. Poult Sci 2021; 100:100978. [PMID: 33588344 PMCID: PMC7896188 DOI: 10.1016/j.psj.2021.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 11/30/2020] [Accepted: 01/07/2021] [Indexed: 11/16/2022] Open
Abstract
Genetic selection of quail for a low body weight for more than 80 generations established a low-weight (LW) Japanese quail line that has been previously characterized to have a muscle hypoplasia phenotype. The aim of this study is to investigate the relationship of temporal expression levels of myostatin (Mstn) and myogenic regulatory factors (MRFs) with hypoplastic muscle growth in the LW line. During embryonic day (E) 13 to 15, gain of embryo weight was 2-fold lower (P < 0.001) in the LW line than that in the random bred control (CON). Gains in body weight and pectoralis muscle weight from hatch to posthatch day (P) 28 were also significantly lower (P < 0.01) in the LW line but increased by 4-fold (P < 0.05) during P42 to P75. PCR analysis showed that expression levels of Mstn were greater in the LW at embryonic stage (E12 to E14, P < 0.05), but there was no difference after hatch. In addition, expression levels of Pax7 and myogenin (MyoG) at E12 were 23-fold (P < 0.05) and 3.4-fold (P < 0.05) lesser in the LW line, respectively. At E14, expression of Pax3, Pax7, and MyoG gene was 3.5-fold (P < 0.05), 6.5-fold (P = 0.065), and 4.4-fold (P < 0.01) less than that in the CON. Taken together, high expression levels of Mstn and low expression of MRFs during embryonic stages can be associated with development of muscle hypoplasia and delayed muscle growth in the LW quail line. These data provide evidence that genetic selection for a low body weight resulting in an avian model with muscle hypoplasia has altered the expression profiles of myogenic factors.
Collapse
Affiliation(s)
- Dong-Hwan Kim
- Department of Animal Sciences, The Ohio State University, Columbus OH 43210, USA
| | - Young Min Choi
- Department of Animal Sciences, Kyungpook National University, Sangju 37224, South Korea
| | - Yeunsu Suh
- Department of Animal Sciences, The Ohio State University, Columbus OH 43210, USA
| | - Sangsu Shin
- Department of Animal Biotechnology, Kyungpook National University, Sangju 37224, South Korea
| | - Joonbum Lee
- Department of Animal Sciences, The Ohio State University, Columbus OH 43210, USA; Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH 43210, USA
| | - Seongsoo Hwang
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Wanju-gun, Jeonbuk 55365, Republic of Korea
| | - Sang Suk Lee
- Department of Animal Science and Technology, Sunchon National University, Jeonnam 57922, South Korea
| | - Kichoon Lee
- Department of Animal Sciences, The Ohio State University, Columbus OH 43210, USA; Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
31
|
Peris-Moreno D, Cussonneau L, Combaret L, Polge C, Taillandier D. Ubiquitin Ligases at the Heart of Skeletal Muscle Atrophy Control. Molecules 2021; 26:molecules26020407. [PMID: 33466753 PMCID: PMC7829870 DOI: 10.3390/molecules26020407] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle loss is a detrimental side-effect of numerous chronic diseases that dramatically increases mortality and morbidity. The alteration of protein homeostasis is generally due to increased protein breakdown while, protein synthesis may also be down-regulated. The ubiquitin proteasome system (UPS) is a master regulator of skeletal muscle that impacts muscle contractile properties and metabolism through multiple levers like signaling pathways, contractile apparatus degradation, etc. Among the different actors of the UPS, the E3 ubiquitin ligases specifically target key proteins for either degradation or activity modulation, thus controlling both pro-anabolic or pro-catabolic factors. The atrogenes MuRF1/TRIM63 and MAFbx/Atrogin-1 encode for key E3 ligases that target contractile proteins and key actors of protein synthesis respectively. However, several other E3 ligases are involved upstream in the atrophy program, from signal transduction control to modulation of energy balance. Controlling E3 ligases activity is thus a tempting approach for preserving muscle mass. While indirect modulation of E3 ligases may prove beneficial in some situations of muscle atrophy, some drugs directly inhibiting their activity have started to appear. This review summarizes the main signaling pathways involved in muscle atrophy and the E3 ligases implicated, but also the molecules potentially usable for future therapies.
Collapse
|
32
|
Kim MJ, O'Connor MB. Drosophila Activin signaling promotes muscle growth through InR/TORC1-dependent and -independent processes. Development 2021; 148:dev190868. [PMID: 33234715 PMCID: PMC7823159 DOI: 10.1242/dev.190868] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 11/16/2020] [Indexed: 12/25/2022]
Abstract
The Myostatin/Activin branch of the TGF-β superfamily acts as a negative regulator of vertebrate skeletal muscle size, in part, through downregulation of insulin/insulin-like growth factor 1 (IGF-1) signaling. Surprisingly, recent studies in Drosophila indicate that motoneuron-derived Activin signaling acts as a positive regulator of muscle size. Here we demonstrate that Drosophila Activin signaling promotes the growth of muscle cells along all three axes: width, thickness and length. Activin signaling positively regulates the insulin receptor (InR)/TORC1 pathway and the level of Myosin heavy chain (Mhc), an essential sarcomeric protein, via increased Pdk1 and Akt1 expression. Enhancing InR/TORC1 signaling in the muscle of Activin pathway mutants restores Mhc levels close to those of the wild type, but only increases muscle width. In contrast, hyperactivation of the Activin pathway in muscles increases overall larval body and muscle fiber length, even when Mhc levels are lowered by suppression of TORC1. Together, these results indicate that the Drosophila Activin pathway regulates larval muscle geometry and body size via promoting InR/TORC1-dependent Mhc production and the differential assembly of sarcomeric components into either pre-existing or new sarcomeric units depending on the balance of InR/TORC1 and Activin signals.
Collapse
Affiliation(s)
- Myung-Jun Kim
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael B O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
33
|
Molecular Mechanisms of Glucocorticoid-Induced Insulin Resistance. Int J Mol Sci 2021; 22:ijms22020623. [PMID: 33435513 PMCID: PMC7827500 DOI: 10.3390/ijms22020623] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/29/2020] [Accepted: 01/02/2021] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoids (GCs) are steroids secreted by the adrenal cortex under the hypothalamic-pituitary-adrenal axis control, one of the major neuro-endocrine systems of the organism. These hormones are involved in tissue repair, immune stability, and metabolic processes, such as the regulation of carbohydrate, lipid, and protein metabolism. Globally, GCs are presented as ‘flight and fight’ hormones and, in that purpose, they are catabolic hormones required to mobilize storage to provide energy for the organism. If acute GC secretion allows fast metabolic adaptations to respond to danger, stress, or metabolic imbalance, long-term GC exposure arising from treatment or Cushing’s syndrome, progressively leads to insulin resistance and, in fine, cardiometabolic disorders. In this review, we briefly summarize the pharmacological actions of GC and metabolic dysregulations observed in patients exposed to an excess of GCs. Next, we describe in detail the molecular mechanisms underlying GC-induced insulin resistance in adipose tissue, liver, muscle, and to a lesser extent in gut, bone, and brain, mainly identified by numerous studies performed in animal models. Finally, we present the paradoxical effects of GCs on beta cell mass and insulin secretion by the pancreas with a specific focus on the direct and indirect (through insulin-sensitive organs) effects of GCs. Overall, a better knowledge of the specific action of GCs on several organs and their molecular targets may help foster the understanding of GCs’ side effects and design new drugs that possess therapeutic benefits without metabolic adverse effects.
Collapse
|
34
|
Lawrence MM, Zwetsloot KA, Arthur ST, Sherman CA, Huot JR, Badmaev V, Grace M, Lila MA, Nieman DC, Shanely RA. Phytoecdysteroids Do Not Have Anabolic Effects in Skeletal Muscle in Sedentary Aging Mice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18020370. [PMID: 33418916 PMCID: PMC7825148 DOI: 10.3390/ijerph18020370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 01/07/2023]
Abstract
Skeletal muscle mass and strength are lost with aging. Phytoecdysteroids, in particular 20-hydroxyecdysone (20E), increase protein synthesis in C2C12 skeletal muscle cells and muscle strength in young rats. The objective of this study was to determine whether an extract from Ajuga turkestanica (ATE), enriched in phytoecdysteroids, and 20E affect skeletal muscle mass and fiber size, fiber type, activation of the PI3K–Akt signaling pathway, and the mRNA levels of MAFbx, MuRF-1, and myostatin in sedentary aging mice. Aging male C57BL/6 mice (20 months old) received ATE, 20E, or vehicle (CT) once per day for 28 days or a single acute dose. Treatment did not alter body, muscle, or organ mass; fiber cross-sectional area; or fiber type in the triceps brachii or plantaris muscles. Likewise, protein synthesis signaling markers (i.e., phosphorylation of AktSer473 and p70S6kThr389) measured after either 28 days or acutely were unchanged. Neither ATE nor 20E treatment for 28 days affected the mRNA levels of MAFbx, MuRF-1, and myostatin. In conclusion, these data indicate that phytoecdysteroid treatment does not alter muscle mass or fiber type, nor does it activate protein synthesis signaling in the skeletal muscle of sedentary aging mice.
Collapse
Affiliation(s)
- Marcus M. Lawrence
- Department of Health and Exercise Science, Appalachian State University, Boone, NC 28608, USA; (M.M.L.); (K.A.Z.); (C.A.S.)
- Human Performance Laboratory, North Carolina Research Campus, Kannapolis, NC 28081, USA;
- Integrated Muscle Physiology Laboratory, Boone, NC 28607, USA
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, UT 84720, USA
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina Charlotte, Charlotte, NC 28223, USA; (S.T.A.); (J.R.H.)
| | - Kevin A. Zwetsloot
- Department of Health and Exercise Science, Appalachian State University, Boone, NC 28608, USA; (M.M.L.); (K.A.Z.); (C.A.S.)
- Integrated Muscle Physiology Laboratory, Boone, NC 28607, USA
- Department of Biology, Appalachian State University, Boone, NC 20608, USA
| | - Susan T. Arthur
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina Charlotte, Charlotte, NC 28223, USA; (S.T.A.); (J.R.H.)
| | - Chase A. Sherman
- Department of Health and Exercise Science, Appalachian State University, Boone, NC 28608, USA; (M.M.L.); (K.A.Z.); (C.A.S.)
- Integrated Muscle Physiology Laboratory, Boone, NC 28607, USA
| | - Joshua R. Huot
- Laboratory of Systems Physiology, Department of Kinesiology, University of North Carolina Charlotte, Charlotte, NC 28223, USA; (S.T.A.); (J.R.H.)
| | | | - Mary Grace
- Plants for Human Health Institute, North Carolina Research Campus, North Carolina State University, Kannapolis, NC 28081, USA; (M.G.); (M.A.L.)
| | - Mary Ann Lila
- Plants for Human Health Institute, North Carolina Research Campus, North Carolina State University, Kannapolis, NC 28081, USA; (M.G.); (M.A.L.)
| | - David C. Nieman
- Human Performance Laboratory, North Carolina Research Campus, Kannapolis, NC 28081, USA;
- Department of Biology, Appalachian State University, Boone, NC 20608, USA
| | - R. Andrew Shanely
- Department of Health and Exercise Science, Appalachian State University, Boone, NC 28608, USA; (M.M.L.); (K.A.Z.); (C.A.S.)
- Human Performance Laboratory, North Carolina Research Campus, Kannapolis, NC 28081, USA;
- Integrated Muscle Physiology Laboratory, Boone, NC 28607, USA
- Correspondence: ; Tel.: +1-828-262-6319
| |
Collapse
|
35
|
Wiedmer P, Jung T, Castro JP, Pomatto LC, Sun PY, Davies KJ, Grune T. Sarcopenia - Molecular mechanisms and open questions. Ageing Res Rev 2021; 65:101200. [PMID: 33130247 DOI: 10.1016/j.arr.2020.101200] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
Sarcopenia represents a muscle-wasting syndrome characterized by progressive and generalized degenerative loss of skeletal muscle mass, quality, and strength occurring during normal aging. Sarcopenia patients are mainly suffering from the loss in muscle strength and are faced with mobility disorders reducing their quality of life and are, therefore, at higher risk for morbidity (falls, bone fracture, metabolic diseases) and mortality. Several molecular mechanisms have been described as causes for sarcopenia that refer to very different levels of muscle physiology. These mechanisms cover e. g. function of hormones (e. g. IGF-1 and Insulin), muscle fiber composition and neuromuscular drive, myo-satellite cell potential to differentiate and proliferate, inflammatory pathways as well as intracellular mechanisms in the processes of proteostasis and mitochondrial function. In this review, we describe sarcopenia as a muscle-wasting syndrome distinct from other atrophic diseases and summarize the current view on molecular causes of sarcopenia development as well as open questions provoking further research efforts for establishing efficient lifestyle and therapeutic interventions.
Collapse
|
36
|
Zhao X, Zhu R, Wang Y, Qi J, Wang J, Bai L, Wang H, Wu Y, Hu H. Differentiation proliferative capacity of skeletal muscle satellite cells from Dapulian and Landrace pigs. ITALIAN JOURNAL OF ANIMAL SCIENCE 2020. [DOI: 10.1080/1828051x.2020.1769511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Xueyan Zhao
- Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Rongsheng Zhu
- Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Yanping Wang
- Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jing Qi
- Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jiying Wang
- Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Liya Bai
- Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Huaizhong Wang
- Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Ying Wu
- Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Hongmei Hu
- Shandong Provincial Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| |
Collapse
|
37
|
Knapp F, Niemann B, Li L, Molenda N, Kracht M, Schulz R, Rohrbach S. Differential effects of right and left heart failure on skeletal muscle in rats. J Cachexia Sarcopenia Muscle 2020; 11:1830-1849. [PMID: 32985798 PMCID: PMC7749622 DOI: 10.1002/jcsm.12612] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/17/2020] [Accepted: 07/07/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Exercise intolerance is a cardinal symptom in right (RV) and left ventricular (LV) failure. The underlying skeletal muscle contributes to increased morbidity in patients. Here, we compared skeletal muscle sarcopenia in a novel two-stage model of RV failure to an established model of LV failure. METHODS Pulmonary artery banding (PAB) or aortic banding (AOB) was performed in weanling rats, inducing a transition from compensated cardiac hypertrophy (after 7 weeks) to heart failure (after 22-26 weeks). Cardiac function was characterized by echocardiography. Skeletal muscle catabolic/anabolic balance and energy metabolism were analysed by histological and biochemical methods, real-time PCR, and western blot. RESULTS Two clearly distinguishable stages of left or right heart disease with a comparable severity were reached. However, skeletal muscle impairment was significantly more pronounced in LV failure. While the compensatory stage resulted only in minor changes, soleus and gastrocnemius muscle of AOB rats at the decompensated stage demonstrated reduced weight and fibre diameter, higher proteasome activity and expression of the muscle-specific ubiquitin E3 ligases muscle-specific RING finger 1 and atrogin-1, increased expression of the atrophy marker myostatin, increased autophagy activation, and impaired mitochondrial function and respiratory chain gene expression. Soleus and gastrocnemius muscle of PAB rats did not show significant changes in muscle weight and proteasome or autophagy activation, but mitochondrial function was mildly impaired as well. The diaphragm did not demonstrate differences in any model or disease stage except for myostatin expression, which was altered at the decompensated stage in both models. Plasma interleukin (IL)-6 and angiotensin II were strongly increased at the decompensated stage (AOB > > PAB). Soleus and gastrocnemius muscle itself demonstrated an increase in IL-6 expression independent from blood-derived cytokines only in AOB animals. In vitro experiments in rat skeletal muscle cells suggested a direct impact of IL-6 and angiotensin II on distinctive atrophic changes. CONCLUSIONS Manifold skeletal muscle alterations are more pronounced in LV failure compared with RV failure despite a similar ventricular impairment. Most of the catabolic changes were observed in soleus or gastrocnemius muscle rather than in the constantly active diaphragm. Mitochondrial dysfunction and up-regulation of myostatin were identified as the earliest signs of skeletal muscle impairment.
Collapse
Affiliation(s)
- Fabienne Knapp
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Bernd Niemann
- Department of Adult and Pediatric Cardiac and Vascular Surgery, University Hospital Giessen and Marburg, Justus Liebig University Giessen, Rudolf-Buchheim-Strasse 7, Giessen, 35392, Germany
| | - Ling Li
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Nicole Molenda
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Schubertstrasse 81, Giessen, 35392, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
38
|
Hagg A, Kharoud S, Goodchild G, Goodman CA, Chen JL, Thomson RE, Qian H, Gregorevic P, Harrison CA, Walton KL. TMEPAI/PMEPA1 Is a Positive Regulator of Skeletal Muscle Mass. Front Physiol 2020; 11:560225. [PMID: 33250771 PMCID: PMC7672205 DOI: 10.3389/fphys.2020.560225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/12/2020] [Indexed: 12/20/2022] Open
Abstract
Inhibition of myostatin- and activin-mediated SMAD2/3 signaling using ligand traps, such as soluble receptors, ligand-targeting propeptides and antibodies, or follistatin can increase skeletal muscle mass in healthy mice and ameliorate wasting in models of cancer cachexia and muscular dystrophy. However, clinical translation of these extracellular approaches targeting myostatin and activin has been hindered by the challenges of achieving efficacy without potential effects in other tissues. Toward the goal of developing tissue-specific myostatin/activin interventions, we explored the ability of transmembrane prostate androgen-induced (TMEPAI), an inhibitor of transforming growth factor-β (TGF-β1)-mediated SMAD2/3 signaling, to promote growth, and counter atrophy, in skeletal muscle. In this study, we show that TMEPAI can block activin A, activin B, myostatin and GDF-11 activity in vitro. To determine the physiological significance of TMEPAI, we employed Adeno-associated viral vector (AAV) delivery of a TMEPAI expression cassette to the muscles of healthy mice, which increased mass by as much as 30%, due to hypertrophy of muscle fibers. To demonstrate that TMEPAI mediates its effects via inhibition of the SMAD2/3 pathway, tibialis anterior (TA) muscles of mice were co-injected with AAV vectors expressing activin A and TMEPAI. In this setting, TMEPAI blocked skeletal muscle wasting driven by activin-induced phosphorylation of SMAD3. In a model of cancer cachexia associated with elevated circulating activin A, delivery of AAV:TMEPAI into TA muscles of mice bearing C26 colon tumors ameliorated the muscle atrophy normally associated with cancer progression. Collectively, the findings indicate that muscle-directed TMEPAI gene delivery can inactivate the activin/myostatin-SMAD3 pathway to positively regulate muscle mass in healthy settings and models of disease.
Collapse
Affiliation(s)
- Adam Hagg
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Swati Kharoud
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Georgia Goodchild
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Craig A Goodman
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science, Sunshine Hospital, The University of Melbourne, St Albans, VIC, Australia
| | - Justin L Chen
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Rachel E Thomson
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Hongwei Qian
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.,Department of Neurology, The University of Washington School of Medicine, Seattle, WA, United States
| | - Craig A Harrison
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Kelly L Walton
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Hudson Institute of Medical Research, Clayton, VIC, Australia
| |
Collapse
|
39
|
Myostatin-1 Inhibits Cell Proliferation by Inhibiting the mTOR Signal Pathway and MRFs, and Activating the Ubiquitin-Proteasomal System in Skeletal Muscle Cells of Japanese Flounder Paralichthys olivaceus. Cells 2020; 9:cells9112376. [PMID: 33138208 PMCID: PMC7692286 DOI: 10.3390/cells9112376] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 11/17/2022] Open
Abstract
Myostatin (MSTN) is a negative regulator of skeletal muscle growth and development. The mechanisms of fish MSTN involved in muscle growth are not fully understood. In the present study, knockdown and overexpression of mstn-1 was performed in cultured Japanese flounder muscle cells to investigate the molecular function and the underlying mechanism of fish MSTN-1. Results showed that mstn-1 knockdown significantly induced cell proliferation and the mRNA expression of myogenic regulatory factors (MRFs), while overexpression of mstn-1 led to a significant decrease of cell proliferation and a suppression of the MRFs mRNA expression. The overexpression of mstn-1 also significantly increased the mRNA expression of ubiquitin–proteasomal pathway of proteolysis genes including muscle RING-finger protein 1 (murf-1) by 204.1% (p = 0.024) and muscle atrophy F-box protein (mafbx) by 165.7% (p = 0.011). However, mystn-1 overexpression inhibited the activation of mTOR signal pathway and the AKT/FoxO1 pathway through decreasing phosphorylation of AKT at Ser 473 by 56.0% (p = 0.001). Meanwhile, mystn-1 overexpression increased the dephosphorylation and nuclear localization of FoxO1 by 394.9% (p = 0.005). These results demonstrate that mstn-1 in Japanese flounder has the effects of inhibiting cell proliferation and growth, and the mTOR and AKT/FoxO1 pathways participated in these biological effects.
Collapse
|
40
|
Myostatin as a Biomarker of Muscle Wasting and other Pathologies-State of the Art and Knowledge Gaps. Nutrients 2020; 12:nu12082401. [PMID: 32796600 PMCID: PMC7469036 DOI: 10.3390/nu12082401] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 12/14/2022] Open
Abstract
Sarcopenia is a geriatric syndrome with a significant impact on older patients’ quality of life, morbidity and mortality. Despite the new available criteria, its early diagnosis remains difficult, highlighting the necessity of looking for a valid muscle wasting biomarker. Myostatin, a muscle mass negative regulator, is one of the potential candidates. The aim of this work is to point out various factors affecting the potential of myostatin as a biomarker of muscle wasting. Based on the literature review, we can say that recent studies produced conflicting results and revealed a number of potential confounding factors influencing their use in sarcopenia diagnosing. These factors include physiological variables (such as age, sex and physical activity) as well as a variety of disorders (including heart failure, metabolic syndrome, kidney failure and inflammatory diseases) and differences in laboratory measurement methodology. Our conclusion is that although myostatin alone might not prove to be a feasible biomarker, it could become an important part of a recently proposed panel of muscle wasting biomarkers. However, a thorough understanding of the interrelationship of these markers, as well as establishing a valid measurement methodology for myostatin and revising current research data in the light of new criteria of sarcopenia, is needed.
Collapse
|
41
|
Effect of Quercetin on Dexamethasone-Induced C2C12 Skeletal Muscle Cell Injury. Molecules 2020; 25:molecules25143267. [PMID: 32709024 PMCID: PMC7397304 DOI: 10.3390/molecules25143267] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/09/2020] [Accepted: 07/15/2020] [Indexed: 01/09/2023] Open
Abstract
Glucocorticoids are widely used anti-inflammatory drugs in clinical settings. However, they can induce skeletal muscle atrophy by reducing fiber cross-sectional area and myofibrillar protein content. Studies have proven that antioxidants can improve glucocorticoid-induced skeletal muscle atrophy. Quercetin is a potent antioxidant flavonoid widely distributed in fruits and vegetables and has shown protective effects against dexamethasone-induced skeletal muscle atrophy. In this study, we demonstrated that dexamethasone significantly inhibited cell growth and induced cell apoptosis by stimulating hydroxyl free radical production in C2C12 skeletal muscle cells. Our results evidenced that quercetin increased C2C12 skeletal cell viability and exerted antiapoptotic effects on dexamethasone-treated C2C12 cells by regulating mitochondrial membrane potential (ΔΨm) and reducing oxidative species. Quercetin can protect against dexamethasone-induced muscle atrophy by regulating the Bax/Bcl-2 ratio at the protein level and abnormal ΔΨm, which leads to the suppression of apoptosis.
Collapse
|
42
|
Shad BJ, Thompson JL, Holwerda AM, Stocks B, Elhassan YS, Philp A, VAN Loon LJC, Wallis GA. One Week of Step Reduction Lowers Myofibrillar Protein Synthesis Rates in Young Men. Med Sci Sports Exerc 2020; 51:2125-2134. [PMID: 31083048 DOI: 10.1249/mss.0000000000002034] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE Across the lifespan, physical activity levels decrease and time spent sedentary typically increases. However, little is known about the impact that these behavioral changes have on skeletal muscle mass regulation. The primary aim of this study was to use a step reduction model to determine the impact of reduced physical activity and increased sedentary time on daily myofibrillar protein synthesis rates in healthy young men. METHODS Eleven men (22 ± 2 yr) completed 7 d of habitual physical activity (HPA) followed by 7 d of step reduction (SR). Myofibrillar protein synthesis rates were determined during HPA and SR using the deuterated water (H2O) method combined with the collection of skeletal muscle biopsies and daily saliva samples. Gene expression of selected proteins related to muscle mass regulation and oxidative metabolism were determined via real time reverse transcription-quantitative polymerase chain reaction (RT-qPCR). RESULTS Daily step count was reduced by approximately 91% during SR (from 13,054 ± 2763 steps per day to 1192 ± 330 steps per day; P < 0.001) and this led to an increased contribution of sedentary time to daily activity (73% ± 6% to 90% ± 3%; P < 0.001). Daily myofibrillar protein synthesis decreased by approximately 27% from 1.39 ± 0.32%·d during HPA to 1.01 ± 0.38%·d during SR (P < 0.05). Muscle atrophy F-box and myostatin mRNA expression were upregulated, whereas mechanistic target of rapamycin, p53, and PDK4 mRNA expression were downregulated after SR (P < 0.05). CONCLUSIONS One week of reduced physical activity and increased sedentary time substantially lowers daily myofibrillar protein synthesis rates in healthy young men.
Collapse
Affiliation(s)
- Brandon J Shad
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UNITED KINGDOM
| | - Janice L Thompson
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UNITED KINGDOM
| | - Andrew M Holwerda
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, THE NETHERLANDS
| | - Ben Stocks
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UNITED KINGDOM
| | - Yasir S Elhassan
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UNITED KINGDOM.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UNITED KINGDOM
| | - Andrew Philp
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, New South Wales, AUSTRALIA
| | - Luc J C VAN Loon
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, THE NETHERLANDS
| | - Gareth A Wallis
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UNITED KINGDOM
| |
Collapse
|
43
|
Goldbraikh D, Neufeld D, Eid‐Mutlak Y, Lasry I, Gilda JE, Parnis A, Cohen S. USP1 deubiquitinates Akt to inhibit PI3K-Akt-FoxO signaling in muscle during prolonged starvation. EMBO Rep 2020; 21:e48791. [PMID: 32133736 PMCID: PMC7132338 DOI: 10.15252/embr.201948791] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 01/29/2020] [Accepted: 02/11/2020] [Indexed: 01/11/2023] Open
Abstract
PI3K-Akt-FoxO-mTOR signaling is the central pathway controlling growth and metabolism in all cells. Ubiquitination of the protein kinase Akt prior to its phosphorylation is required for PI3K-Akt activity. Here, we found that the deubiquitinating (DUB) enzyme USP1 removes K63-linked polyubiquitin chains on Akt to restrict PI3K-Akt-FoxO signaling in mouse muscle during prolonged starvation. DUB screening platform identified USP1 as a direct DUB for Akt, and USP1 depletion in mouse muscle increased Akt ubiquitination, PI3K-Akt-FoxO signaling, and glucose uptake during fasting. Co-immunoprecipitation and mass spectrometry identified disabled homolog-2 (Dab2), the tuberous sclerosis complex TSC1/TSC2, and PHLPP1 as USP1 bound proteins. During starvation, Dab2 is essential for Akt recruitment to USP1-TSC1-PHLPP1 complex, and for PI3K-Akt-FoxO inhibition. Surprisingly, USP1 limits TSC1 levels to sustain mTOR-mediated basal protein synthesis rates and maintain its own protein levels. We propose that Dab2 recruits Akt to USP1-TSC1-PHLPP1 complex to efficiently terminate the transmission of growth signals when cellular energy level is low.
Collapse
Affiliation(s)
- Dana Goldbraikh
- Faculty of BiologyTechnion Institute of TechnologyHaifaIsrael
| | | | - Yara Eid‐Mutlak
- Faculty of BiologyTechnion Institute of TechnologyHaifaIsrael
| | - Inbal Lasry
- Faculty of BiologyTechnion Institute of TechnologyHaifaIsrael
| | | | - Anna Parnis
- Faculty of BiologyTechnion Institute of TechnologyHaifaIsrael
| | - Shenhav Cohen
- Faculty of BiologyTechnion Institute of TechnologyHaifaIsrael
| |
Collapse
|
44
|
Kang W, Tong T, Park T. Corticotropin releasing factor-overexpressing mouse is a model of chronic stress-induced muscle atrophy. PLoS One 2020; 15:e0229048. [PMID: 32049987 PMCID: PMC7015416 DOI: 10.1371/journal.pone.0229048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/28/2020] [Indexed: 12/20/2022] Open
Abstract
Chronic stress and continually high glucocorticoid levels can induce muscle atrophy. Unfortunately, there is a lack of appropriate animal models for stress-induced muscle atrophy research. Corticotropin releasing factor-overexpressing (CRF-OE) mice are a transgenic model of chronic stress that exhibit increased plasma corticosterone levels and Cushing’s syndrome; however, the skeletal muscle pathology of the CRF-OE mouse has not been well studied. We observed that male, 19-week-old CRF-OE mice had significantly lower skeletal muscle mass, average cross-sectional myofiber area, and total muscle protein content than their wild type (WT) littermates. Muscle function determined by grip strength, wire-hang, and open field tests showed that 19-week-old male CRF-OE mice had impaired physical ability. Additionally, the skeletal muscles of CRF-mice exhibited decreased expression of factors involved in the IGF-1/AKT/mTOR protein synthesis pathway and increased ubiquitin proteasome pathway activity compared to the WT control mice. In conclusion, 19-week-old CRF-OE mice display numerous features of muscle atrophy and thus serve as a model for investigating stress-induced muscle atrophy and interventions to target the deleterious effects of stress on skeletal muscle.
Collapse
Affiliation(s)
- Wesuk Kang
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, Yonsei University, Seodaemun-gu, Seoul, Korea
| | - Tao Tong
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, Yonsei University, Seodaemun-gu, Seoul, Korea
| | - Taesun Park
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, Yonsei University, Seodaemun-gu, Seoul, Korea
- * E-mail:
| |
Collapse
|
45
|
Aravena J, Abrigo J, Gonzalez F, Aguirre F, Gonzalez A, Simon F, Cabello-Verrugio C. Angiotensin (1-7) Decreases Myostatin-Induced NF-κB Signaling and Skeletal Muscle Atrophy. Int J Mol Sci 2020; 21:ijms21031167. [PMID: 32050585 PMCID: PMC7037856 DOI: 10.3390/ijms21031167] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 02/07/2023] Open
Abstract
Myostatin is a myokine that regulates muscle function and mass, producing muscle atrophy. Myostatin induces the degradation of myofibrillar proteins, such as myosin heavy chain or troponin. The main pathway that mediates protein degradation during muscle atrophy is the ubiquitin proteasome system, by increasing the expression of atrogin-1 and MuRF-1. In addition, myostatin activates the NF-κB signaling pathway. Renin–angiotensin system (RAS) also regulates muscle mass. Angiotensin (1-7) (Ang-(1-7)) has anti-atrophic properties in skeletal muscle. In this paper, we evaluated the effect of Ang-(1-7) on muscle atrophy and signaling induced by myostatin. The results show that Ang-(1-7) prevented the decrease of the myotube diameter and myofibrillar protein levels induced by myostatin. Ang-(1-7) also abolished the increase of myostatin-induced reactive oxygen species production, atrogin-1, MuRF-1, and TNF-α gene expressions and NF-κB signaling activation. Ang-(1-7) inhibited the activity mediated by myostatin through Mas receptor, as is demonstrated by the loss of all Ang-(1-7)-induced effects when the Mas receptor antagonist A779 was used. Our results show that the effects of Ang-(1-7) on the myostatin-dependent muscle atrophy and signaling are blocked by MK-2206, an inhibitor of Akt/PKB. Together, these data indicate that Ang-(1-7) inhibited muscle atrophy and signaling induced by myostatin through a mechanism dependent on Mas receptor and Akt/PKB.
Collapse
Affiliation(s)
- Javier Aravena
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Johanna Abrigo
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Francisco Gonzalez
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Francisco Aguirre
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Andrea Gonzalez
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
- Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD), Universidad de Chile, Santiago 8370146, Chile
- Laboratory of Integrative Physiopathology, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Department of Biological Sciences, Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8370146, Chile
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago 8350709, Chile
- Correspondence: ; Tel.: +5622-770-3665
| |
Collapse
|
46
|
Chronic uphill and downhill exercise protocols do not lead to sarcomerogenesis in mouse skeletal muscle. J Biomech 2020; 98:109469. [PMID: 31732175 DOI: 10.1016/j.jbiomech.2019.109469] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 01/13/2023]
|
47
|
Penniman CM, Suarez Beltran PA, Bhardwaj G, Junck TL, Jena J, Poro K, Hirshman MF, Goodyear LJ, O'Neill BT. Loss of FoxOs in muscle reveals sex-based differences in insulin sensitivity but mitigates diet-induced obesity. Mol Metab 2019; 30:203-220. [PMID: 31767172 PMCID: PMC6819874 DOI: 10.1016/j.molmet.2019.10.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Gender influences obesity-related complications, including diabetes. Females are more protected from insulin resistance after diet-induced obesity, which may be related to fat accumulation and muscle insulin sensitivity. FoxOs regulate muscle atrophy and are targets of insulin action, but their role in muscle insulin sensitivity and mitochondrial metabolism is unknown. METHODS We measured muscle insulin signaling, mitochondrial energetics, and metabolic responses to a high-fat diet (HFD) in male and female muscle-specific FoxO1/3/4 triple knock-out (TKO) mice. RESULTS In male TKO muscle, insulin-stimulated AKT activation was decreased. AKT2 protein and mRNA levels were reduced and insulin receptor protein and IRS-2 mRNA decreased. These changes contributed to decreased insulin-stimulated glucose uptake in glycolytic muscle in males. In contrast, female TKOs maintain normal insulin-mediated AKT phosphorylation, normal AKT2 levels, and normal glucose uptake in glycolytic muscle. When challenged with a HFD, fat gain was attenuated in both male and female TKO mice, and associated with decreased glucose levels, improved glucose homeostasis, and reduced muscle triglyceride accumulation. Furthermore, female TKO mice showed increased energy expenditure, relative to controls, due to increased lean mass and maintenance of mitochondrial function in muscle. CONCLUSIONS FoxO deletion in muscle uncovers sexually dimorphic regulation of AKT2, which impairs insulin signaling in male mice, but not females. However, loss of FoxOs in muscle from both males and females also leads to muscle hypertrophy and increases in metabolic rate. These factors mitigate fat gain and attenuate metabolic abnormalities in response to a HFD.
Collapse
Affiliation(s)
- Christie M Penniman
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Pablo A Suarez Beltran
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Gourav Bhardwaj
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Taylor L Junck
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jayashree Jena
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Kennedy Poro
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Michael F Hirshman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Laurie J Goodyear
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Brian T O'Neill
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
48
|
Li Y, Zhang F, Modrak S, Little A, Zhang H. Chronic Alcohol Consumption Enhances Skeletal Muscle Wasting in Mice Bearing Cachectic Cancers: The Role of TNFα/Myostatin Axis. Alcohol Clin Exp Res 2019; 44:66-77. [PMID: 31657476 DOI: 10.1111/acer.14221] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/03/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chronic alcohol consumption enhances cancer-associated cachexia, which is one of the major causes of decreased survival. The precise molecular mechanism of how alcohol consumption enhances cancer-associated cachexia, especially skeletal muscle loss, remains to be elucidated. METHODS We used a mouse model of chronic alcohol consumption, in which 20% (w/v) alcohol was provided as sole drinking fluid, and Lewis lung carcinoma to study the underlying mechanisms. RESULTS We found that alcohol consumption up-regulated the expression of MAFbx, MuRF-1, and LC3 in skeletal muscle, suggesting that alcohol enhanced ubiquitin-mediated proteolysis and LC3-mediated autophagy. Alcohol consumption enhanced phosphorylation of Smad2/3, p38, and ERK and decreased the phosphorylation of FOXO1. These are the signaling molecules governing protein degradation pathways. Moreover, alcohol consumption slightly up-regulated the expression of insulin receptor substrate-1, did not affect phosphatidylinositol-3 kinase, but decreased the phosphorylation of Akt and mammalian target of rapamycin (mTOR), and down-regulated the expression of Raptor and p70 ribosomal kinase S6 kinase, suggesting that alcohol impaired protein synthesis signaling pathway in skeletal muscle of tumor-bearing mice. Alcohol consumption enhanced the expression of myostatin in skeletal muscle, plasma, and tumor, but did not affect the expression of myostatin in non-tumor-bearing mice. In TNFα knockout mice, the effects of alcohol-enhanced expression of myostatin and protein degradation-related signaling molecules, and decreased protein synthesis signaling in skeletal muscle were abolished. Consequently, alcohol consumption neither affected cancer-associated cachexia nor decreased the survival of TNFα KO mice bearing cachectic cancer. CONCLUSIONS Chronic alcohol consumption enhances cancer-associated skeletal muscle loss through suppressing Akt/mTOR-mediated protein synthesis pathway and enhancing protein degradation pathways. This process is initiated by TNFα and mediated by myostatin.
Collapse
Affiliation(s)
- Yuanfei Li
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington.,Department of Oncology, (YL), The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Faya Zhang
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Samantha Modrak
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Alex Little
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| | - Hui Zhang
- From the Department of Pharmaceutical Sciences (YL, FZ, SM, AL, HZ) College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington
| |
Collapse
|
49
|
Saneyasu T, Honda K, Kamisoyama H. Myostatin Increases Smad2 Phosphorylation and Atrogin-1 Expression in Chick Embryonic Myotubes. J Poult Sci 2019; 56:224-230. [PMID: 32055218 PMCID: PMC7005388 DOI: 10.2141/jpsa.0180092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/12/2018] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle mass is an important trait in poultry meat production. In mammals, myostatin, a negative regulator of skeletal muscle growth, activates Smad transcription factors and induces the expression of atrogin-1 by regulating the Akt/FOXO pathway. Although the amino acid sequence of chicken myostatin is known to be completely identical to its mammalian counterpart, previous studies in chicken skeletal muscles have implied that the physiological roles of chicken myostatin are different from those of mammals. Furthermore, it remains to be elucidated whether myostatin affects cellular signaling factors and atrogin-1 expression. In this study, using chick embryonic myotubes, we found that myostatin significantly increased the phosphorylation rate of Smad2 and mRNA levels of atrogin-1. No significant change was observed in the phosphorylation of Akt and FOXO1. These in vitro results suggest that the molecular mechanisms underlying myostatin-induced expression of atrogin-1 might be different between chickens and mammals.
Collapse
Affiliation(s)
- Takaoki Saneyasu
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Kazuhisa Honda
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Hiroshi Kamisoyama
- Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| |
Collapse
|
50
|
Kim YM, Ji ES, Ko IG, Jin JJ, Cho YH, Seo TB. Combination of treadmill exercise with bone marrow stromal cells transplantation activates protein synthesis-related molecules in soleus muscle of the spinal cord injured rats. J Exerc Rehabil 2019; 15:377-382. [PMID: 31316929 PMCID: PMC6614772 DOI: 10.12965/jer.1938284.142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/03/2019] [Indexed: 12/21/2022] Open
Abstract
The present study investigated whether treadmill exercise with bone marrow stromal cells (BMSCs) transplantation increase expression level of protein synthesis-related molecules in the soleus muscle after spinal cord injury (SCI). The spinal cord contusion injury was performed at the T9-10 level using the impactor (10 g×25 mm). BMSCs were cultured from femur and tibia of 4-week-old rats and then transplanted directly into the lesion 1-week post injury. The rats in exercise group were walking on treadmill device for 6 days per a week during 6 weeks. Prepared soleus muscles were used for examining mechanisms of protein synthesis after SCI. Myostatin induction level was increased by SCI, but BMSCs engrafting after SCI decreased compared to SCI group. Combination of treadmill exercise with BMSCs showed more potent decrement on myostatin expression. Protein kinase B (Akt) and mammalian target of rapamycin (mTOR) levels were significantly increased in SCI and BMSCs transplantation group compared to SCI group. Combination of treadmill exercise with BMSCs further facilitated expression levels of Akt and mTOR. Insulin-like growth factor-I (IGF-I) and phosphorylated cyclic adenosine monophosphate response element-binding protein (p-CREB) induction levels were more increased in SCI and BMSC transplantation group compared to SCI group. Combination of treadmill exercise with BMSCs further increased expression levels of IGF-I and p-CREB, although statistical significance was not appeared. Combining treadmill exercise with BMSCs transplantation might accelerate protein synthesis and hypertrophy in the soleus muscle after SCI through activation of IGF-I/mTOR signaling pathway.
Collapse
Affiliation(s)
- You-Mi Kim
- Sports Science Research Institution, Korea National Sport University, Seoul, Korea
| | - Eun-Sang Ji
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Il-Gyu Ko
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jun-Jang Jin
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Yeong-Hyun Cho
- Department of Kinesiology, College of Natural Science, Jeju National University, Jeju, Korea
| | - Tae-Beom Seo
- Department of Kinesiology, College of Natural Science, Jeju National University, Jeju, Korea
| |
Collapse
|