1
|
Ingelson-Filpula WA, Breedon SA, Storey KB. MicroRNA, Myostatin, and Metabolic Rate Depression: Skeletal Muscle Atrophy Resistance in Hibernating Myotis lucifugus. Cells 2024; 13:2074. [PMID: 39768165 PMCID: PMC11674624 DOI: 10.3390/cells13242074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/18/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Little brown bats (Myotis lucifugus) cluster in hibernacula sites over winter, in which they use metabolic rate depression (MRD) to facilitate entrance and exit of hibernation. This study used small RNA sequencing and bioinformatic analyses to identify differentially regulated microRNAs (miRNAs) and to predict their downstream effects on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) terms in the skeletal muscle of torpid M. lucifugus as compared to euthermic controls. We observed a subset of ten miRNAs whose expression changed during hibernation, with predicted functional roles linked to cell cycle processes, downregulation of protein degradation via ubiquitin-mediated proteolysis, downregulation of signaling pathways, including MAPK, p53, mTOR, and TGFβ, and downregulation of cytoskeletal and vesicle trafficking terms. Taken together, our results indicate miRNA regulation corresponding to both widely utilized MRD survival strategies, as well as more hibernation- and tissue-specific roles in M. lucifugus, including skeletal muscle atrophy resistance via myostatin inhibition and insulin signaling suppression.
Collapse
Affiliation(s)
- W. Aline Ingelson-Filpula
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada; (W.A.I.-F.); (K.B.S.)
| | - Sarah A. Breedon
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada; (W.A.I.-F.); (K.B.S.)
| | - Kenneth B. Storey
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada; (W.A.I.-F.); (K.B.S.)
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
2
|
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
|
3
|
Bucarey JL, Trujillo-González I, Paules EM, Espinosa A. Myokines and Their Potential Protective Role Against Oxidative Stress in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Antioxidants (Basel) 2024; 13:1363. [PMID: 39594505 PMCID: PMC11591161 DOI: 10.3390/antiox13111363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
Myokines, bioactive peptides released by skeletal muscle, have emerged as crucial regulators of metabolic and protective pathways in peripheral tissues, particularly in combating oxidative stress and inflammation. Their plasma concentration significantly increases following exercise, offering valuable insights into the role of physical activity in preventing sarcopenia and mitigating metabolic diseases, including obesity, diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD). This review focuses on discussing the roles of specific myokines in activating intracellular signaling pathways within the liver, which confer protection against steatosis and lipid peroxidation. We detail the mechanism underlying lipid peroxidation and highlight the liver's antioxidant defenses, such as glutathione (GSH) and glutathione peroxidase 4 (GPX4), which are pivotal in reducing ferroptosis. Furthermore, we provide an in-depth analysis of key myokines, including myostatin, brain-derived neurotrophic factor (BDNF), and irisin, among others, and their potential impact on liver function. Finally, we discuss the molecular mechanisms through which these myokines influence oxidate stress and lipid metabolism, emphasizing their capacity to modulate antioxidant responses in the liver. Finally, we underscore the therapeutic potential of exercise as a non-pharmacological intervention to enhance myokine release, thereby preventing the progression of MASD through improved hepatic antioxidant defenses. This review represents a comprehensive perspective on the intersection of exercise, myokine biology, and liver health.
Collapse
Affiliation(s)
- José Luis Bucarey
- School of Medicine, Faculty of Medicine, Universidad de Valparaíso, San Felipe 2172972, Chile;
| | - Isis Trujillo-González
- Nutrition Research Institute, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.T.-G.); (E.M.P.)
| | - Evan M. Paules
- Nutrition Research Institute, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.T.-G.); (E.M.P.)
| | - Alejandra Espinosa
- School of Medicine, Faculty of Medicine, Universidad de Valparaíso, San Felipe 2172972, Chile;
- Center of Interdisciplinary Biomedical and Engineering Research for Health, Universidad de Valparaíso, San Felipe 2172972, Chile
| |
Collapse
|
4
|
Kataoka R, Hammert WB, Yamada Y, Song JS, Seffrin A, Kang A, Spitz RW, Wong V, Loenneke JP. The Plateau in Muscle Growth with Resistance Training: An Exploration of Possible Mechanisms. Sports Med 2024; 54:31-48. [PMID: 37787845 DOI: 10.1007/s40279-023-01932-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2023] [Indexed: 10/04/2023]
Abstract
It is hypothesized that there is likely a finite ability for muscular adaptation. While it is difficult to distinguish between a true plateau following a long-term training period and short-term stalling in muscle growth, a plateau in muscle growth has been attributed to reaching a genetic potential, with limited discussion on what might physiologically contribute to this muscle growth plateau. The present paper explores potential physiological factors that may drive the decline in muscle growth after prolonged resistance training. Overall, with chronic training, the anabolic signaling pathways may become more refractory to loading. While measures of anabolic markers may have some predictive capabilities regarding muscle growth adaptation, they do not always demonstrate a clear connection. Catabolic processes may also constrain the ability to achieve further muscle growth, which is influenced by energy balance. Although speculative, muscle cells may also possess cell scaling mechanisms that sense and regulate their own size, along with molecular brakes that hinder growth rate over time. When considering muscle growth over the lifespan, there comes a point when the anabolic response is attenuated by aging, regardless of whether or not individuals approach their muscle growth potential. Our goal is that the current review opens avenues for future experimental studies to further elucidate potential mechanisms to explain why muscle growth may plateau.
Collapse
Affiliation(s)
- Ryo Kataoka
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - William B Hammert
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Yujiro Yamada
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Jun Seob Song
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Aldo Seffrin
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Anna Kang
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Robert W Spitz
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Vickie Wong
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA
| | - Jeremy P Loenneke
- Department of Health, Exercise Science, and Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, P.O. Box 1848, University, MS, 38677, USA.
| |
Collapse
|
5
|
Knapp M, Supruniuk E, Górski J. Myostatin and the Heart. Biomolecules 2023; 13:1777. [PMID: 38136649 PMCID: PMC10741510 DOI: 10.3390/biom13121777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Myostatin (growth differentiation factor 8) is a member of the transforming growth factor-β superfamily. It is secreted mostly by skeletal muscles, although small amounts of myostatin are produced by the myocardium and the adipose tissue as well. Myostatin binds to activin IIB membrane receptors to activate the downstream intracellular canonical Smad2/Smad3 pathway, and additionally acts on non-Smad (non-canonical) pathways. Studies on transgenic animals have shown that overexpression of myostatin reduces the heart mass, whereas removal of myostatin has an opposite effect. In this review, we summarize the potential diagnostic and prognostic value of this protein in heart-related conditions. First, in myostatin-null mice the left ventricular internal diameters along with the diastolic and systolic volumes are larger than the respective values in wild-type mice. Myostatin is potentially secreted as part of a negative feedback loop that reduces the effects of the release of growth-promoting factors and energy reprogramming in response to hypertrophic stimuli. On the other hand, both human and animal data indicate that myostatin is involved in the development of the cardiac cachexia and heart fibrosis in the course of chronic heart failure. The understanding of the role of myostatin in such conditions might initiate a development of targeted therapies based on myostatin signaling inhibition.
Collapse
Affiliation(s)
- Małgorzata Knapp
- Department of Cardiology, Medical University of Białystok, 15-276 Białystok, Poland
| | - Elżbieta Supruniuk
- Department of Physiology, Medical University of Białystok, 15-222 Białystok, Poland;
| | - Jan Górski
- Department of Health Sciences, University of Łomża, 18-400 Łomża, Poland;
| |
Collapse
|
6
|
Kim JS, Taaffe DR, Galvão DA, Clay TD, Redfern AD, Hart NH, Gray ES, Ryan CJ, Kenfield SA, Saad F, Newton RU. Acute effect of high-intensity interval aerobic exercise on serum myokine levels and resulting tumour-suppressive effect in trained patients with advanced prostate cancer. Prostate Cancer Prostatic Dis 2023; 26:795-801. [PMID: 36446945 DOI: 10.1038/s41391-022-00624-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE Although skeletal muscle releases cytokines called myokines during exercise, the kinetics of the acute myokine response to exercise (exercise-induced circulatory myokine level alteration) is unknown in patients with advanced prostate cancer. We measured myokine levels in serum obtained from patients with metastatic castrate-resistant prostate cancer (mCRPC) before and after exercise and assessed the growth-suppressive effect of the serum by applying it to a PCa cell line. METHODS Nine patients with mCRPC (age = 67.8 ± 10.1 years, time since mCRPC diagnosis 36.2 ± 22.5 months) undertook 34 min of a high-intensity interval exercise session on a cycle ergometer. Blood was collected immediately pre, post and 30 min post. Serum levels of secreted protein acidic and rich in cysteine (SPARC), oncostatin M (OSM), interleukin-6 (IL-6), interleukin-15 (IL-15), decorin, irisin, and IGF-1 were determined. Growth of the androgen-independent PCa cell line DU-145 exposed to serum collected at three points was measured. RESULTS There was a significant elevation of SPARC (19.9%, P = 0.048), OSM (11.5%, P = 0.001), IL-6 (10.2%, P = 0.02) and IL-15 (7.8%, P = 0.023) in serum collected immediately after exercise compared to baseline, returning to baseline after 30 min rest. A significant reduction in DU-145 Cell growth and the Cell Index area under the curve at 72 h incubation was observed with the presence of serum obtained immediately post-exercise (Cell Index at 72 h: 16.9%, P < 0.001; area under the curve: 15.2%, P < 0.001) and with the presence of serum obtained 30 min post-exercise compared to baseline (Cell Index at 72 h: 6.5%; area under the curve: 8.8%, P < 0.001). CONCLUSION This study provides preliminary evidence for an acute exercise-induced myokine response and tumour growth suppression in serum after a bout of high-intensity interval exercise in patients with advanced PCa.
Collapse
Affiliation(s)
- Jin-Soo Kim
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Dennis R Taaffe
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Daniel A Galvão
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Timothy D Clay
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Department of Oncology, St John of God Subiaco Hospital, Perth, WA, Australia
| | - Andrew D Redfern
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, WA, Australia
- School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia
| | - Nicolas H Hart
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- School of Sport, Exercise, and Rehabilitation, University of Technology Sydney, Moore Park, NSW, Australia
| | - Elin S Gray
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Centre of Precision Health, Edith Cowan University, Joondalup, WA, Australia
| | - Charles J Ryan
- Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Stacey A Kenfield
- Departments of Urology and Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Fred Saad
- Department of Urology, Centre Hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Robert U Newton
- Exercise Medicine Research Institute, Edith Cowan University, Joondalup, WA, Australia.
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia.
- School of Human Movement and Nutrition Sciences, University of Queensland, St Lucia, QLD, Australia.
| |
Collapse
|
7
|
Zhang T, Li J, Li X, Liu Y. Intermuscular adipose tissue in obesity and related disorders: cellular origins, biological characteristics and regulatory mechanisms. Front Endocrinol (Lausanne) 2023; 14:1280853. [PMID: 37920255 PMCID: PMC10619759 DOI: 10.3389/fendo.2023.1280853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/01/2023] [Indexed: 11/04/2023] Open
Abstract
Intermuscular adipose tissue (IMAT) is a unique adipose depot interspersed between muscle fibers (myofibers) or muscle groups. Numerous studies have shown that IMAT is strongly associated with insulin resistance and muscular dysfunction in people with metabolic disease, such as obesity and type 2 diabetes. Moreover, IMAT aggravates obesity-related muscle metabolism disorders via secretory factors. Interestingly, researchers have discovered that intermuscular brown adipocytes in rodent models provide new hope for obesity treatment by acting on energy dissipation, which inspired researchers to explore the underlying regulation of IMAT formation. However, the molecular and cellular properties and regulatory processes of IMAT remain debated. Previous studies have suggested that muscle-derived stem/progenitor cells and other adipose tissue progenitors contribute to the development of IMAT. Adipocytes within IMAT exhibit features that are similar to either white adipocytes or uncoupling protein 1 (UCP1)-positive brown adipocytes. Additionally, given the heterogeneity of skeletal muscle, which comprises myofibers, satellite cells, and resident mesenchymal progenitors, it is plausible that interplay between these cellular components actively participate in the regulation of intermuscular adipogenesis. In this context, we review recent studies associated with IMAT to offer insights into the cellular origins, biological properties, and regulatory mechanisms of IMAT. Our aim is to provide novel ideas for the therapeutic strategy of IMAT and the development of new drugs targeting IMAT-related metabolic diseases.
Collapse
Affiliation(s)
- Ting Zhang
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
- Medical Research Center, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| | - Jun Li
- Department of Orthopedics, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| | - Xi Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yanjun Liu
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, China
| |
Collapse
|
8
|
Félix-Soriano E, Stanford KI. Exerkines and redox homeostasis. Redox Biol 2023; 63:102748. [PMID: 37247469 PMCID: PMC10236471 DOI: 10.1016/j.redox.2023.102748] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/31/2023] Open
Abstract
Exercise physiology has gained increasing interest due to its wide effects to promote health. Recent years have seen a growth in this research field also due to the finding of several circulating factors that mediate the effects of exercise. These factors, termed exerkines, are metabolites, growth factors, and cytokines secreted by main metabolic organs during exercise to regulate exercise systemic and tissue-specific effects. The metabolic effects of exerkines have been broadly explored and entail a promising target to modulate beneficial effects of exercise in health and disease. However, exerkines also have broad effects to modulate redox signaling and homeostasis in several cellular processes to improve stress response. Since redox biology is central to exercise physiology, this review summarizes current evidence for the cross-talk between redox biology and exerkines actions. The role of exerkines in redox biology entails a response to oxidative stress-induced pathological cues to improve health outcomes and to modulate exercise adaptations that integrate redox signaling.
Collapse
Affiliation(s)
- Elisa Félix-Soriano
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
9
|
Bhasin S, Cawthon PM, Correa-de-Araujo R, Storer TW, Volpi E, Newman AB, Dioh W, Tourette C, Evans WJ, Fielding RA. Optimizing the Design of Clinical Trials to Evaluate the Efficacy of Function-Promoting Therapies. J Gerontol A Biol Sci Med Sci 2023; 78:86-93. [PMID: 37325959 PMCID: PMC10272979 DOI: 10.1093/gerona/glad024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Several candidate molecules that may have application in treating physical limitations associated with aging and chronic diseases are in development. Challenges in the framing of indications, eligibility criteria, and endpoints and the lack of regulatory guidance have hindered the development of function-promoting therapies. METHODS Experts from academia, pharmaceutical industry, National Institutes of Health (NIH), and Food and Drug Administration (FDA) discussed optimization of trial design including the framing of indications, eligibility criteria, and endpoints. RESULTS Mobility disability associated with aging and chronic diseases is an attractive indication because it is recognized by geriatricians as a common condition associated with adverse outcomes, and it can be ascertained reliably. Other conditions associated with functional limitation in older adults include hospitalization for acute illnesses, cancer cachexia, and fall injuries. Efforts are underway to harmonize definitions of sarcopenia and frailty. Eligibility criteria should reconcile the goals of selecting participants with the condition and ensuring generalizability and ease of recruitment. An accurate measure of muscle mass (eg, D3 creatine dilution) could be a good biomarker in early-phase trials. Performance-based and patient-reported measures of physical function are needed to demonstrate whether treatment improves how a person lives, functions, or feels. Multicomponent functional training that integrates training in balance, stability, strength, and functional tasks with cognitive and behavioral strategies may be needed to translate drug-induced muscle mass gains into functional improvements. CONCLUSIONS Collaborations among academic investigators, NIH, FDA, pharmaceutical industry, patients, and professional societies are needed to conduct well-designed trials of function-promoting pharmacological agents with and without multicomponent functional training.
Collapse
Affiliation(s)
- Shalender Bhasin
- Research Program in Men’s Health, Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peggy M Cawthon
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Rosaly Correa-de-Araujo
- Division of Geriatrics and Clinical Gerontology, National Institute on Aging, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, USA
| | - Thomas W Storer
- Research Program in Men’s Health, Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elena Volpi
- Sealy Center on Aging; UTMB Claude D. Pepper Older Americans Independence Center, University of Texas Medical Branch, Galveston, Texas, USA
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | | | | | - William J Evans
- Department of Nutritional Science and Toxicology, University of California at Berkely, Berkely, California, USA
| | - Roger A Fielding
- Nutrition, Exercise Physiology, and Sarcopenia Laboratory, Jean Mayer USDA Human Nutrition Research Center in Aging, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Yang M, Liu C, Jiang N, Liu Y, Luo S, Li C, Zhao H, Han Y, Chen W, Li L, Xiao L, Sun L. Myostatin: a potential therapeutic target for metabolic syndrome. Front Endocrinol (Lausanne) 2023; 14:1181913. [PMID: 37288303 PMCID: PMC10242177 DOI: 10.3389/fendo.2023.1181913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/04/2023] [Indexed: 06/09/2023] Open
Abstract
Metabolic syndrome is a complex metabolic disorder, its main clinical manifestations are obesity, hyperglycemia, hypertension and hyperlipidemia. Although metabolic syndrome has been the focus of research in recent decades, it has been proposed that the occurrence and development of metabolic syndrome is related to pathophysiological processes such as insulin resistance, adipose tissue dysfunction and chronic inflammation, but there is still a lack of favorable clinical prevention and treatment measures for metabolic syndrome. Multiple studies have shown that myostatin (MSTN), a member of the TGF-β family, is involved in the development and development of obesity, hyperlipidemia, diabetes, and hypertension (clinical manifestations of metabolic syndrome), and thus may be a potential therapeutic target for metabolic syndrome. In this review, we describe the transcriptional regulation and receptor binding pathway of MSTN, then introduce the role of MSTN in regulating mitochondrial function and autophagy, review the research progress of MSTN in metabolic syndrome. Finally summarize some MSTN inhibitors under clinical trial and proposed the use of MSTN inhibitor as a potential target for the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Ming Yang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chongbin Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Na Jiang
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yan Liu
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Shilu Luo
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chenrui Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Hao Zhao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Wei Chen
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Li Li
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Li Xiao
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| |
Collapse
|
11
|
Mieske P, Scheinpflug J, Yorgan TA, Brylka L, Palme R, Hobbiesiefken U, Preikschat J, Lewejohann L, Diederich K. Effects of more natural housing conditions on the muscular and skeletal characteristics of female C57BL/6J mice. Lab Anim Res 2023; 39:9. [PMID: 37189184 DOI: 10.1186/s42826-023-00160-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/17/2023] [Accepted: 04/30/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Enrichment of home cages in laboratory experiments offers clear advantages, but has been criticized in some respects. First, there is a lack of definition, which makes methodological uniformity difficult. Second, there is concern that the enrichment of home cages may increase the variance of results in experiments. Here, the influence of more natural housing conditions on physiological parameters of female C57BL/6J mice was investigated from an animal welfare point of view. For this purpose, the animals were kept in three different housing conditions: conventional cage housing, enriched housing and the semi naturalistic environment. The focus was on musculoskeletal changes after long-term environmental enrichment. RESULTS The housing conditions had a long-term effect on the body weight of the test animals. The more complex and natural the home cage, the heavier the animals. This was associated with increased adipose deposits in the animals. There were no significant changes in muscle and bone characteristics except for single clues (femur diameter, bone resorption marker CTX-1). Additionally, the animals in the semi naturalistic environment (SNE) were found to have the fewest bone anomalies. Housing in the SNE appears to have the least effect on stress hormone concentrations. The lowest oxygen uptake was observed in enriched cage housing. CONCLUSIONS Despite increasing values, observed body weights were in the normal and strain-typical range. Overall, musculoskeletal parameters were slightly improved and age-related effects appear to have been attenuated. The variances in the results were not increased by more natural housing. This confirms the suitability of the applied housing conditions to ensure and increase animal welfare in laboratory experiments.
Collapse
Affiliation(s)
- Paul Mieske
- German Center for the Protection of Laboratory Animals (Bf3R), Federal German Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany.
| | - Julia Scheinpflug
- German Center for the Protection of Laboratory Animals (Bf3R), Federal German Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Laura Brylka
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Rupert Palme
- Unit of Physiology, Pathophysiology, and Experimental Endocrinology, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210, Vienna, Austria
| | - Ute Hobbiesiefken
- German Center for the Protection of Laboratory Animals (Bf3R), Federal German Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Juliane Preikschat
- German Center for the Protection of Laboratory Animals (Bf3R), Federal German Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | - Lars Lewejohann
- German Center for the Protection of Laboratory Animals (Bf3R), Federal German Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
- Animal Behavior and Laboratory Animal Science, Institute of Animal Welfare, Freie Universität Berlin, Königsweg 67, 14163, Berlin, Germany
| | - Kai Diederich
- German Center for the Protection of Laboratory Animals (Bf3R), Federal German Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| |
Collapse
|
12
|
Shrestha A, Dani M, Kemp P, Fertleman M. Acute Sarcopenia after Elective and Emergency Surgery. Aging Dis 2022; 13:1759-1769. [PMID: 36465176 PMCID: PMC9662269 DOI: 10.14336/ad.2022.0404] [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: 03/10/2022] [Accepted: 04/04/2022] [Indexed: 04/12/2024] Open
Abstract
Sarcopenia is an increasingly recognised condition of loss of muscle mass and function. The European Working Group on Sarcopenia in Older People 2 (EWSOP2) updated their definition in 2018, emphasising the importance of low muscle strength in diagnosis. Acute sarcopenia has been arbitrarily defined as sarcopenia lasting less than 6 months. This review highlights the pathophysiology involved in muscle wasting following surgery, focussing on hormonal factors, inflammation, microRNAs, and oxidative stress. Biomarkers such as GDF-15, IGF-1 and various microRNAs may predict post-surgical muscle loss. The impact of existing sarcopenia on various types of surgery and incident muscle wasting following surgery is also described. The gaps in research found include the need for longitudinal studies looking in changes in muscle strength and quantity following surgery. Further work is needed to examine if biomarkers are replicated in other surgery to consolidate existing theories on the pathophysiology of muscle wasting.
Collapse
Affiliation(s)
- Alvin Shrestha
- Cutrale Perioperative and Ageing group, Imperial College London, London SW7 2BX, United Kingdom
| | - Melanie Dani
- Cutrale Perioperative and Ageing group, Imperial College London, London SW7 2BX, United Kingdom
| | - Paul Kemp
- National Lung and Health Institute, Imperial College London, London SW7 2BX, United Kingdom
| | - Michael Fertleman
- Cutrale Perioperative and Ageing group, Imperial College London, London SW7 2BX, United Kingdom
| |
Collapse
|
13
|
Mishra R, Jha R, Mishra B, Kim YS. Maternal immunization against myostatin suppresses post-hatch chicken growth. PLoS One 2022; 17:e0275753. [PMID: 36201511 PMCID: PMC9536644 DOI: 10.1371/journal.pone.0275753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/22/2022] [Indexed: 11/07/2022] Open
Abstract
Myostatin (MSTN) is a negative regulator of skeletal muscle growth, thus it was hypothesized that immunization of hens against MSTN would enhance post-hatch growth and muscle mass via suppression of MSTN activity by anti-MSTN IgY in fertilized eggs. This study investigated the effects of immunization of hens against chicken MSTN (chMSTN) or a MSTN fragment (Myo2) on the growth and muscle mass of offspring. In Experiment 1, hens mixed with roosters were divided into two groups and hens in the Control and chMSTN groups were immunized with 0 and 0.5 mg of chMSTN, respectively. In Experiment 2, hens in the chMSTN group were divided into chMSTN and Myo2 groups while the Control group remained the same. The Control and chMSTN groups were immunized in the same way as Experiment 1. The Myo2 group was immunized against MSTN peptide fragment (Myo2) conjugated to KLH. Eggs collected from each group were incubated, and chicks were reared to examine growth and carcass parameters. ELISA showed the production of IgYs against chMSTN and Myo2 and the presence of these antibodies in egg yolk. IgY from the chMSTN and Myo2 groups showed binding affinity to chMSTN, Myo2, and commercial MSTN in Western blot analysis but did not show MSTN-inhibitory capacity in a reporter gene assay. In Experiment 1, no difference was observed in the body weight and carcass parameters of offspring between the Control and chMSTN groups. In Experiment 2, the body weight of chicks from the Myo2 group was significantly lower than that of the Control or chMSTN groups. The dressing percentage and breast muscle mass of the chMSTN and Myo2 groups were significantly lower than those of the Control group, and the breast muscle mass of Myo2 was significantly lower than that of the chMSTN. In summary, in contrast to our hypothesis, maternal immunization of hens did not increase but decreased the body weight and muscle mass of offspring.
Collapse
Affiliation(s)
- Rajeev Mishra
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Rajesh Jha
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Birendra Mishra
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Yong Soo Kim
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
- * E-mail:
| |
Collapse
|
14
|
Dang DX, Zhou H, Lou Y, Liu X, Li D. Development of breast muscle parameters, glycogen reserves, and myogenic gene expression in goslings during pre- and post-hatching periods. Front Physiol 2022; 13:990715. [PMID: 36176777 PMCID: PMC9513458 DOI: 10.3389/fphys.2022.990715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022] Open
Abstract
This study aimed to better understand the development patterns of breast muscle and glycogen reserves in goslings during pre- and post-hatching periods. The timepoints for sampling were embryonic days 23 and 27 of hatching and days 1, 4, and 7 post hatching. We found that the body weight of goslings increased with age. The small intestine developed with age and remained reasonably constant on day 4 post hatching. The breast muscle development decreased with age and stayed relatively stable on day 1 post hatching. The diameter of myofiber increased prior to hatching and then decreased while hatching. The development patterns of breast muscle glycogen reserves were similar to the diameter of myofiber. In contrast, the contents of liver glycogen began to decrease before hatching and then increased rapidly after hatching. Moreover, the expression of Myf-5 increased with age. The expression of MSTN was maintained at high levels prior to hatching, dropped immediately after hatching, and then gradually increased with age. Additionally, we also observed that the glycogen content in the breast muscle was positively correlated with the diameter of the myofiber. The liver glycogen content was positively correlated to the relative weight of the breast muscle, the diameter of the myofiber, and the breast muscle glycogen content. The development pattern of the myofiber was synchronized with the change in the MSTN/Myf-5 ratio. This study provided a profile to understand the development patterns of breast muscle, glycogen reserves, and myogenic gene expression in goslings, which was beneficial to understanding the characteristics of energy reserves during the early life of goslings.
Collapse
Affiliation(s)
- De Xin Dang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
- Department of Animal Resources Science, Dankook University, Cheonan, South Korea
| | - Haizhu Zhou
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yujie Lou
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Xiao Liu
- Institute of Animal Nutrition, College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
- *Correspondence: Xiao Liu, ; Desheng Li,
| | - Desheng Li
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
- *Correspondence: Xiao Liu, ; Desheng Li,
| |
Collapse
|
15
|
Holder ER, Alibhai FJ, Caudle SL, McDermott JC, Tobin SW. The importance of biological sex in cardiac cachexia. Am J Physiol Heart Circ Physiol 2022; 323:H609-H627. [PMID: 35960634 DOI: 10.1152/ajpheart.00187.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac cachexia is a catabolic muscle wasting syndrome observed in approximately 1 in 10 heart failure patients. Increased skeletal muscle atrophy leads to frailty and limits mobility which impacts quality of life, exacerbates clinical care, and is associated with higher rates of mortality. Heart failure is known to exhibit a wide range of prevalence and severity when examined across individuals of different ages and with co-morbidities related to diabetes, renal failure and pulmonary dysfunction. It is also recognized that men and women exhibit striking differences in the pathophysiology of heart failure as well as skeletal muscle homeostasis. Given that both skeletal muscle and heart failure physiology are in-part sex dependent, the diagnosis and treatment of cachexia in heart failure patients may depend on a comprehensive examination of how these organs interact. In this review we explore the potential for sex-specific differences in cardiac cachexia. We summarize advantages and disadvantages of clinical methods used to measure muscle mass and function and provide alternative measurements that should be considered in preclinical studies. Additionally, we summarize sex-dependent effects on muscle wasting in preclinical models of heart failure, disuse, and cancer. Lastly, we discuss the endocrine function of the heart and outline unanswered questions that could directly impact patient care.
Collapse
|
16
|
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
|
17
|
Velez LM, Van C, Moore T, Zhou Z, Johnson C, Hevener AL, Seldin MM. Genetic variation of putative myokine signaling is dominated by biological sex and sex hormones. eLife 2022; 11:e76887. [PMID: 35416774 PMCID: PMC9094747 DOI: 10.7554/elife.76887] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022] Open
Abstract
Skeletal muscle plays an integral role in coordinating physiological homeostasis, where signaling to other tissues via myokines allows for coordination of complex processes. Here, we aimed to leverage natural genetic correlation structure of gene expression both within and across tissues to understand how muscle interacts with metabolic tissues. Specifically, we performed a survey of genetic correlations focused on myokine gene regulation, muscle cell composition, cross-tissue signaling, and interactions with genetic sex in humans. While expression levels of a majority of myokines and cell proportions within skeletal muscle showed little relative differences between males and females, nearly all significant cross-tissue enrichments operated in a sex-specific or hormone-dependent fashion; in particular, with estradiol. These sex- and hormone-specific effects were consistent across key metabolic tissues: liver, pancreas, hypothalamus, intestine, heart, visceral, and subcutaneous adipose tissue. To characterize the role of estradiol receptor signaling on myokine expression, we generated male and female mice which lack estrogen receptor α specifically in skeletal muscle (MERKO) and integrated with human data. These analyses highlighted potential mechanisms of sex-dependent myokine signaling conserved between species, such as myostatin enriched for divergent substrate utilization pathways between sexes. Several other putative sex-dependent mechanisms of myokine signaling were uncovered, such as muscle-derived tumor necrosis factor alpha (TNFA) enriched for stronger inflammatory signaling in females compared to males and GPX3 as a male-specific link between glycolytic fiber abundance and hepatic inflammation. Collectively, we provide a population genetics framework for inferring muscle signaling to metabolic tissues in humans. We further highlight sex and estradiol receptor signaling as critical variables when assaying myokine functions and how changes in cell composition are predicted to impact other metabolic organs.
Collapse
Affiliation(s)
- Leandro M Velez
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, University of California IrvineIrvineUnited States
| | - Cassandra Van
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, University of California IrvineIrvineUnited States
| | - Timothy Moore
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine at UCLALos AngelesUnited States
| | - Zhenqi Zhou
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine at UCLALos AngelesUnited States
| | - Casey Johnson
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, University of California IrvineIrvineUnited States
| | - Andrea L Hevener
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, David Geffen School of Medicine at UCLALos AngelesUnited States
- Iris Cantor-UCLA Women’s Health Research Center, David Geffen School of Medicine at UCLALos AngelesUnited States
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, IrvineIrvineUnited States
- Center for Epigenetics and Metabolism, University of California IrvineIrvineUnited States
| |
Collapse
|
18
|
Lee HJ, Hong SJ, Kim SS, Kwon YY, Choi BH, Choi KM, Sheen SH, Lee MJ, Hwang SY, Park K, Joo Y, Song H, Lee CK. CD4+/CD8+ Ratio and Growth Differentiation Factor 8 Levels in Peripheral Blood of Large Canine Males Are Useful Parameters to Build an Age Prediction Model. World J Mens Health 2022; 40:316-329. [PMID: 35021315 PMCID: PMC8987144 DOI: 10.5534/wjmh.210003] [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: 01/19/2021] [Revised: 09/14/2021] [Accepted: 10/08/2021] [Indexed: 11/21/2022] Open
Abstract
Purpose To build an age prediction model, we measured CD4+ and CD8+ cells, and humoral components in canine peripheral blood. Materials and Methods Large Belgian Malinois (BGM) and German Shepherd Dog (GSD) breeds (n=27), aged from 1 to 12 years, were used for this study. Peripheral bloods were obtained by venepuncture, then plasma and peripheral blood mononuclear cells (PBMCs) were separated immediately. Six myokines, including interleukin (IL)-6, IL-8, IL-15, leukemia inhibitory factor (LIF), growth differentiation factor 8 (GDF8), and GDF11 were measured from plasma and CD4+/CD8+ T-lymphocytes ratio were measured from PBMC. These parameters were then tested with age prediction models to find the best fit model. Results We found that the T-lymphocyte ratio (CD4+/CD8+) was significantly correlated with age (r=0.46, p=0.016). Among the six myokines, only GDF8 showed a significant correlation with age (r=0.52, p=0.005). Interestingly, these two markers showed better correlations in male dogs than females, and BGM breed than GSD. Using these two age biomarkers, we could obtain the best fit in a quadratic linear mixed model (r=0.77, p=3×10-6). Conclusions Age prediction is a challenging task because of complication with biological age. Our quadratic linear mixed model using CD4+/CD8+ ratio and GDF8 level showed a meaningful age prediction.
Collapse
Affiliation(s)
- Han-Jun Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Seok-Jin Hong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Seung-Soo Kim
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Korea
| | - Young-Yon Kwon
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Bong-Hwan Choi
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, Rural Development Administration, Wanju, Korea
| | - Kyung-Mi Choi
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, Korea
| | - Seo-Hyeong Sheen
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Myung-Jin Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Sun-Young Hwang
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | | | - Younghun Joo
- Military Working Dog Training Center, Chuncheon, Korea
| | - Hwayoung Song
- Military Working Dog Training Center, Chuncheon, Korea
| | - Cheol-Koo Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea.,Institute of Animal Molecular Biotechnology, Korea University, Seoul, Korea.
| |
Collapse
|
19
|
Association study between relative expression levels of eight genes and growth rate in Hungarian common carp ( Cyprinus carpio). Saudi J Biol Sci 2022; 29:630-639. [PMID: 35002460 PMCID: PMC8716967 DOI: 10.1016/j.sjbs.2021.09.036] [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: 05/19/2021] [Revised: 09/11/2021] [Accepted: 09/12/2021] [Indexed: 11/20/2022] Open
Abstract
One of the most important issues in improving the competitiveness of the fish production sector is to improve the growth rate of fish. The genetic background to this trait is at present poorly understood. In this study, we compared the relative gene expression levels of the Akt1s1, FGF, GH, IGF1, MSTN, TLR2, TLR4 and TLR5 genes in blood in groups of common carps (Cyprinus carpio), which belonged to different growth types and phenotypes. Fish were divided into groups based on growth rate (normal group: n = 6; slow group: n = 6) and phenotype (scaled group: n = 6; mirror group: n = 6). In the first 18 weeks, we measured significant differences (p < 0.05) between groups in terms of body weight and body length. Over the next 18 weeks, the fish in the slow group showed more intense development. In the same period, the slow group was characterized by lower expression levels for most genes, whereas GH and IGF1 mRNA levels were higher compared to the normal group. We found that phenotype was not a determining factor in differences of relative expression levels of the genes studied.
Collapse
|
20
|
Efthymiadou A, Vasilakis IA, Giannakopoulos A, Chrysis D. Myostatin serum levels in children with type 1 diabetes mellitus. Hormones (Athens) 2021; 20:777-782. [PMID: 34486100 DOI: 10.1007/s42000-021-00317-y] [Citation(s) in RCA: 8] [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: 04/05/2021] [Accepted: 08/27/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE Type 1 diabetes mellitus (T1DM) can cause several complications, among them myopathy, which can appear even in adolescents. This is of importance, since skeletal muscle is the largest of the insulin-sensitive tissues and thus plays a significant role in glucose homeostasis. A prime regulator of skeletal muscle mass is myostatin, a protein which has a negative role in skeletal muscle development but also in glucose homeostasis, causing insulin resistance. Since myopathy is a complication of T1DM and myostatin is a fundamental regulator of skeletal muscle and is also involved in glucose homeostasis, we investigated the serum levels of myostatin in children with T1DM. METHODS We determined myostatin serum levels using ELISA in 87 children with T1DM aged 10.62 ± 3.94 years, and in 75 healthy children aged 10.46 ± 3.32 years old. RESULTS Myοstatin was significantly elevated in T1DM compared to the healthy control children (23.60 ± 7.70 vs 16.74 ± 6.95 ng/ml, p < 0.0001). Myostatin was not correlated with body mass index (BMI) SD or hemoglobin A1c (HbA1c). CONCLUSION Children with T1DM have significantly higher serum levels of myostatin compared to healthy children of the same age and BMI SD. The elevated myostatin in T1DM could reflect impaired muscle function and/or glucose metabolism, or could represent a homeostatic mechanism.
Collapse
Affiliation(s)
- Alexandra Efthymiadou
- Department of Pediatrics, Division of Endocrinology, Medical School, University of Patras, Patras, Rion 26504, Greece
| | - Ioannis-Anargyros Vasilakis
- Department of Pediatrics, Medical School, University of Patras, Patras, Rion 26504, Greece
- First Department of Pediatrics, Division of Endocrinology, Diabetes and Metabolism, Medical School, National and Kapodistrian University of Athens, Aghia Sophia" Children's Hospital, Athens, Greece
| | - Aristeidis Giannakopoulos
- Department of Pediatrics, Division of Endocrinology, Medical School, University of Patras, Patras, Rion 26504, Greece
| | - Dionisios Chrysis
- Department of Pediatrics, Division of Endocrinology, Medical School, University of Patras, Patras, Rion 26504, Greece.
| |
Collapse
|
21
|
Cell Types Used for Cultured Meat Production and the Importance of Myokines. Foods 2021; 10:foods10102318. [PMID: 34681367 PMCID: PMC8534705 DOI: 10.3390/foods10102318] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/20/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022] Open
Abstract
The world’s population continues to increase, meaning we require more consistent protein supply to meet demand. Despite the availability of plant-based protein alternatives, animal meat remains a popular, high-quality protein source. Research studies have focused on cultured meat (meat grown in vitro) as a safe and more efficient alternative to traditional meat. Cultured meat is produced by in vitro myogenesis, which involves the processing of muscle satellite and mature muscle cells. Meat culture efficiency is largely determined by the culture conditions, such as the cell type and cell culture medium used and the biomolecular composition. Protein production can be enhanced by providing the optimum biochemical and physical conditions for skeletal muscle cell growth, while myoblasts play important roles in skeletal muscle formation and growth. This review describes the cell types used to produce cultured meat and the biological effects of various myokines and cytokines, such as interleukin-6, leukemia inhibitory factor, interleukin-4, interleukin-15, and interleukin-1β, on skeletal muscle and myogenesis and their potential roles in cultured meat production.
Collapse
|
22
|
Feraco A, Gorini S, Armani A, Camajani E, Rizzo M, Caprio M. Exploring the Role of Skeletal Muscle in Insulin Resistance: Lessons from Cultured Cells to Animal Models. Int J Mol Sci 2021; 22:ijms22179327. [PMID: 34502235 PMCID: PMC8430804 DOI: 10.3390/ijms22179327] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle is essential to maintain vital functions such as movement, breathing, and thermogenesis, and it is now recognized as an endocrine organ. Muscles release factors named myokines, which can regulate several physiological processes. Moreover, skeletal muscle is particularly important in maintaining body homeostasis, since it is responsible for more than 75% of all insulin-mediated glucose disposal. Alterations of skeletal muscle differentiation and function, with subsequent dysfunctional expression and secretion of myokines, play a key role in the pathogenesis of obesity, type 2 diabetes, and other metabolic diseases, finally leading to cardiometabolic complications. Hence, a deeper understanding of the molecular mechanisms regulating skeletal muscle function related to energy metabolism is critical for novel strategies to treat and prevent insulin resistance and its cardiometabolic complications. This review will be focused on both cellular and animal models currently available for exploring skeletal muscle metabolism and endocrine function.
Collapse
Affiliation(s)
- Alessandra Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
- PhD Programme in Endocrinological Sciences, Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Manfredi Rizzo
- Promise Department, School of Medicine, University of Palermo, 90127 Palermo, Italy;
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
- Correspondence: ; Tel.: +39-065-225-3419
| |
Collapse
|
23
|
MSTN is an important myokine for weight-bearing training to attenuate bone loss in ovariectomized rats. J Physiol Biochem 2021; 78:61-72. [PMID: 34453705 DOI: 10.1007/s13105-021-00838-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
Weight-bearing training, as one of resistance exercises, is beneficial to bone health. Myostatin (MSTN) is a negative regulator of skeletal muscle growth and development. Animals lacking MSTN show increased bone mineral density (BMD). The aim of this study was to investigate the preventive effect of weight-bearing training on bone loss in ovariectomized rats and whether it was related to MSTN. In this study, the rats were randomly assigned to three group: Sham-ovariectomized (Sham), ovariectomized (OVX), ovariectomized and weight-bearing training (OWT). The rats in the OWT group ran at 20-m/min bearing with 35% of their body weight for 6 days/week. After 10 weeks, compared with the OVX group, weight-bearing training increased the BMD of total femur and trabecular bone by 8.13% and 57.44%, respectively. The OVX-induced destruction of bone microarchitecture including the thickness and number of trabeculae and bone volume fraction was all significantly improved (9.26%, 47.68%, 63.03%) in the OWT group. The OVX-induced degradation of bone mechanical properties was significantly enhanced in the OWT group (maximum load increased by 35.46%, stiffness increased by 89.19%, energy absorption increased by 53.4%; elastic modulus increased by 26.3%). Ten-week weight-bearing training also significantly upregulated the mRNA and protein expression of Wnt1 and β-catenin, which is crucial in bone development. Compared with the Sham group, MSTN in serum and muscle increased in the OVX group, but it decreased in the OWT group compared with the OVX group. Its receptor ActRIIB and downstream molecules Smad2/3 in the OVX group were downregulated in bone by weight-bearing training. The results indicated that MSTN is an important myokine for weight-bearing training to attenuate bone loss in ovariectomized rats.
Collapse
|
24
|
Kern-Matschilles S, Gar C, Wanger L, Haschka SJ, Potzel AL, Hesse N, Then C, Seissler J, Lechner A. Association of Serum Myostatin with Body Weight, Visceral Fat Volume, and High Sensitivity C-Reactive Protein But Not With Muscle Mass and Physical Fitness in Premenopausal Women. Exp Clin Endocrinol Diabetes 2021; 130:393-399. [PMID: 34407549 DOI: 10.1055/a-1500-4605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The myokine myostatin regulates muscle mass and has been linked to insulin resistance and metabolic syndrome. However, data on its role in humans is still limited. We, therefore, investigated the associations of serum myostatin with muscle mass, physical fitness, and components of the metabolic syndrome in a cohort of premenopausal women. METHODS We undertook a cross-sectional analysis of 233 women from the monocenter study PPSDiab, conducted in Munich, Germany. Participants had recently completed a pregnancy with or without gestational diabetes. Our analysis included medical history, anthropometrics, oral glucose tolerance testing, laboratory chemistry, cardiopulmonary exercise testing, and magnetic resonance imaging (n=142) of visceral fat volume, left quadriceps muscle mass, and muscle fat content. Serum myostatin was quantified by a competitive enzyme-linked immunosorbent assay. RESULTS We observed positive correlations of serum myostatin with body mass index (ρ=0.235; p=0.0003), body fat percentage (ρ=0.166; p=0.011), waist circumference (ρ=0.206; p=0.002), intraabdominal fat volume (ρ=0.182; p=0.030) and high-sensitivity C-reactive protein (ρ=0.175; p=0.008). These correlations were reproduced in linear regression analyses with adjustment for age and time after delivery. We saw no correlations with muscle mass, physical fitness, insulin sensitivity, triglycerides, HDL cholesterol, and blood pressure. CONCLUSIONS Our observation of elevated serum myostatin in women with a higher body fat percentage, visceral obesity, and elevated c-reactive protein suggests that this myokine contributes to the altered muscle-adipose tissue crosstalk in metabolic syndrome. Elevated myostatin may advance this pathophysiologic process and could also impair the efficacy of exercise interventions. Further mechanistic studies, therefore, seem warranted.
Collapse
Affiliation(s)
- Stefanie Kern-Matschilles
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Christina Gar
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Lorena Wanger
- Klinik und Poliklinik für Radiologie, LMU Klinikum, München, Germany
| | - Stefanie J Haschka
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Anne L Potzel
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Nina Hesse
- Klinik und Poliklinik für Radiologie, LMU Klinikum, München, Germany
| | - Cornelia Then
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Jochen Seissler
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| | - Andreas Lechner
- Diabetes Research Group, Medizinische Klinik und Poliklinik IV, LMU Klinikum, München, Germany.,Clinical Cooperation Group Type 2 Diabetes, Helmholtz Zentrum München, Neuherberg, Germany.,German Center for Diabetes Research (DZD)
| |
Collapse
|
25
|
Anderson LB, Ravara B, Hameed S, Latour CD, Latour SM, Graham VM, Hashmi MN, Cobb B, Dethrow N, Urazaev AK, Davie JK, Albertin G, Carraro U, Zampieri S, Pond AL. MERG1A Protein Abundance Increases in the Atrophied Skeletal Muscle of Denervated Mice, But Does Not Affect NFκB Activity. J Neuropathol Exp Neurol 2021; 80:776-788. [PMID: 34363662 DOI: 10.1093/jnen/nlab062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Skeletal muscle atrophy may occur with disease, injury, decreased muscle use, starvation, and normal aging. No reliably effective treatments for atrophy are available, thus research into the mechanisms contributing to muscle loss is essential. The ERG1A K+ channel contributes to muscle loss by increasing ubiquitin proteasome proteolysis (UPP) in the skeletal muscle of both unweighted and cachectic mice. Because the mechanisms which produce atrophy vary based upon the initiating factor, here we investigate atrophy produced by denervation. Using immunohistochemistry and immunoblots, we demonstrate that ERG1A protein abundance increases significantly in the Gastrocnemius muscle of rodents 7 days after both sciatic nerve transection and hind limb unweighting. Further, we reveal that ectopic expression of a Merg1a encoded plasmid in normal mouse Gastrocnemius muscle has no effect on activity of the NFκB transcription factor family, a group of proteins which contribute to muscle atrophy by modulation of the UPP. Further, although NFκB activity increases significantly after denervation, we show that expression of a plasmid encoding a dominant negative Merg1a mutant in Gastrocnemius muscle prior to denervation, has no effect on NFκB activity. Thus, although the ERG1A K+ channel increases UPP, it does not do so through modulation of NFκB transcription factors.
Collapse
Affiliation(s)
- Luke B Anderson
- Anatomy Department, Southern Illinois University, Carbondale, Illinois, USA (LBA, SH, MNH, BC, ND, ALP)
| | - Barbara Ravara
- Department of Surgery, Oncology, and Gastroenterology and Department of Biomedical Sciences, University of Padova, Padova, Italy (BR, GA, SZ).,Department of Neuroscience (DNS), University of Padova, Padova, Italy (BR, GA).,A&C M-C Foundation for Translational Myology, Padova, Italy (BR, UC)
| | - Sohaib Hameed
- Anatomy Department, Southern Illinois University, Carbondale, Illinois, USA (LBA, SH, MNH, BC, ND, ALP)
| | - Chase D Latour
- Gillings School of Public Health, University of North Carolina, Chapel Hill, North Carolina, USA (CDL)
| | - Sawyer M Latour
- Doisey School of Health, Saint Louis University, St. Louis, Missouri, USA (SML, VMG)
| | - Valerie M Graham
- Doisey School of Health, Saint Louis University, St. Louis, Missouri, USA (SML, VMG)
| | - Mariam N Hashmi
- Anatomy Department, Southern Illinois University, Carbondale, Illinois, USA (LBA, SH, MNH, BC, ND, ALP)
| | - Brittan Cobb
- Anatomy Department, Southern Illinois University, Carbondale, Illinois, USA (LBA, SH, MNH, BC, ND, ALP)
| | - Nicole Dethrow
- Anatomy Department, Southern Illinois University, Carbondale, Illinois, USA (LBA, SH, MNH, BC, ND, ALP)
| | - Albert K Urazaev
- School of Arts, Sciences and Education, Ivy Technical Community College, Lafayette, Indiana, USA (AKU)
| | - Judy K Davie
- Biochemistry Department, Southern Illinois University, Carbondale, Illinois, USA(JKD)
| | - Giovanna Albertin
- Department of Surgery, Oncology, and Gastroenterology and Department of Biomedical Sciences, University of Padova, Padova, Italy (BR, GA, SZ).,Department of Neuroscience (DNS), University of Padova, Padova, Italy (BR, GA)
| | - Ugo Carraro
- A&C M-C Foundation for Translational Myology, Padova, Italy (BR, UC)
| | - Sandra Zampieri
- Department of Surgery, Oncology, and Gastroenterology and Department of Biomedical Sciences, University of Padova, Padova, Italy (BR, GA, SZ)
| | - Amber L Pond
- Anatomy Department, Southern Illinois University, Carbondale, Illinois, USA (LBA, SH, MNH, BC, ND, ALP)
| |
Collapse
|
26
|
Xiong J, Le Y, Rao Y, Zhou L, Hu Y, Guo S, Sun Y. RANKL Mediates Muscle Atrophy and Dysfunction in a Cigarette Smoke-induced Model of Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2021; 64:617-628. [PMID: 33689672 DOI: 10.1165/rcmb.2020-0449oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle dysfunction is one of the important comorbidities of chronic obstructive pulmonary disease (COPD); however, the underlying mechanisms remain largely unknown. RANKL (receptor activator of nuclear factor κB ligand), a key mediator in osteoclast differentiation, was also found to play a role in skeletal muscle pathogenesis. Whether RANKL is involved in COPD-related skeletal muscle dysfunction is as-of-yet unknown. We examined the expression of RANKL/RANK in skeletal muscles from mice exposed to cigarette smoke (CS) for 24 weeks. Grip strength and exercise capacity as well as muscular morphology were evaluated in CS-exposed mice with or without anti-RANKL treatment. The expressions of protein synthesis- or muscle growth-related molecules (IGF-1, myogenin, and myostatin), muscle-specific ubiquitin E3 ligases (MuRF1 and atrogin-1), and the NF-κb inflammatory pathway were also evaluated in skeletal muscles. The effect of CS extract on RANKL/RANK expression and that of exogenous RANKL on the ubiquitin-proteasome pathway in C2C12 myotubes were investigated in vitro. Long-term CS exposure induced skeletal muscle dysfunction and atrophy together with upregulation of RANKL/RANK expression in a well-established mouse model of COPD. RANKL neutralization prevented skeletal muscle dysfunction and atrophy. RANKL inhibition decreased expressions of myostatin and MuRF1/Atrogin1 and suppressed the NF-κb pathway in skeletal muscles from CS-exposed mice. In in vitro experiments with C2C12 myotubes, CS extract induced expression of RANKL/RANK, and exogenous RANKL induced activation of the ubiquitin-proteasome pathway and NF-κb pathway via RANK. Our results revealed an important role of the RANKL/RANK pathway in muscle atrophy induced by CS exposure, suggesting that RANKL may be a potential therapeutic target in COPD-related skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; and
| | - Yanqing Le
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; and
| | - Yafei Rao
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; and
| | - Lu Zhou
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; and
| | - Yuhan Hu
- Department of Respiratory Medicine, and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Suliang Guo
- Department of Respiratory Medicine, and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China; and
| |
Collapse
|
27
|
Seto JT, Roeszler KN, Meehan LR, Wood HD, Tiong C, Bek L, Lee SF, Shah M, Quinlan KGR, Gregorevic P, Houweling PJ, North KN. ACTN3 genotype influences skeletal muscle mass regulation and response to dexamethasone. SCIENCE ADVANCES 2021; 7:eabg0088. [PMID: 34215586 PMCID: PMC11060041 DOI: 10.1126/sciadv.abg0088] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/19/2021] [Indexed: 06/13/2023]
Abstract
Homozygosity for the common ACTN3 null polymorphism (ACTN3 577X) results in α-actinin-3 deficiency in ~20% of humans worldwide and is linked to reduced sprint and power performance in both elite athletes and the general population. α-Actinin-3 deficiency is also associated with reduced muscle mass, increased risk of sarcopenia, and altered muscle wasting response induced by denervation and immobilization. Here, we show that α-actinin-3 plays a key role in the regulation of protein synthesis and breakdown signaling in skeletal muscle and influences muscle mass from early postnatal development. We also show that α-actinin-3 deficiency reduces the atrophic and anti-inflammatory response to the glucocorticoid dexamethasone in muscle and protects against dexamethasone-induced muscle wasting in female but not male mice. The effects of α-actinin-3 deficiency on muscle mass regulation and response to muscle wasting provide an additional mechanistic explanation for the positive selection of the ACTN3 577X allele in recent human history.
Collapse
Affiliation(s)
- Jane T Seto
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Kelly N Roeszler
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Lyra R Meehan
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Harrison D Wood
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Chrystal Tiong
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Lucinda Bek
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Siaw F Lee
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Manan Shah
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Paul Gregorevic
- Centre for Muscle Research, Department of Physiology, 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, University of Washington, Seattle, WA, USA
| | - Peter J Houweling
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
- Department of Paediatrics, University of Melbourne, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Kathryn N North
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia.
- Department of Paediatrics, University of Melbourne, The Royal Children's Hospital, Melbourne, VIC, Australia
| |
Collapse
|
28
|
Abstract
We aimed to assess the effects of spirulina supplementation during gradual weight loss on serum concentrations of follistatin (FST), myostatin (MST), insulin-like growth factor 1 (IGF-1), aspartate aminotransferase (AST), alanine aminotransferase (ALT) and body composition in competitive wrestlers. Forty competitive wrestlers (age: 22 (sem 2) years) were randomly assigned to one of two groups: gradual weight loss + spirulina (SP; n 20) or gradual weight loss + placebo (PL; n 20). Subjects in both groups lost weight according to a designed diet over 12 d and were required to reduce baseline body mass (BM) by 4%. Subjects in the SP group received two tablets of spirulina, while subjects in the PL received two tablets of placebo before each meal. Concentrations of mentioned serum markers and body composition were measured before and after the interventions. BM (SP = -3·1 kg and PL = -2·9 kg), body fat percentage (BFP) (SP = -2·1 % and PL = -0·6 %), fat mass (FM) (SP = -2·2 kg and PL = -0·9 kg) and skeletal muscle mass (SP = -1·4 kg and PL = -1·5 kg) significantly decreased in both groups (P < 0·05). The changes in BFP and FM were significantly greater in SP compared with the PL group (P < 0·001). Additionally, MST (SP = -0·1 ng/ml), AST (SP = -2·1 u/l) and ALT (SP = -2·7 u/l) concentrations significantly diminished in SP group (P = 0·005), while FST (PL = -0·1 ng/ml) and IGF-1 (PL = -2·6 ng/ml) concentrations significantly decreased in PL group (P < 0·05). Spirulina supplementation during gradual weight loss is beneficial in reducing BFP, FM, MST and liver enzymes while maintaining IGF-1 and FST concentrations in competitive wrestlers.
Collapse
|
29
|
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
|
30
|
Bagheri R, Ashtary-Larky D, Elliott BT, Willoughby DS, Kargarfard M, Alipour M, Lamuchi-Deli N, Kooti W, Asbaghi O, Wong A. The effects of gradual vs. rapid weight loss on serum concentrations of myokines and body composition in overweight and obese females. Arch Physiol Biochem 2021; 129:821-828. [PMID: 33502906 DOI: 10.1080/13813455.2021.1874020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Context: Research has shown the modulations of Follistatin (FST) and Myostatin (MST) following weight loss.Objective: We evaluated the effects of gradual weight loss (GWL) and rapid weight loss (RWL) on serum MST, FST, and body composition in overweight and obese females.Materials and methods: Thirty-six overweight and obese females successfully completed the study interventions: GWL (n = 18) or RWL (n = 18). Serum MST and FST concentrations, as well as anthropometric measurements, were collected at baseline and at the conclusion of each weight loss intervention.Results: MST concentration significantly (p < .05) decreased in the GWL; while FST concentration, body fat percentage and skeletal muscle mass significantly declined in both conditions. The loss in skeletal muscle mass was significantly greater in RWL relative to GWL.Discussion and conclusion: GWL was more effective than RWL in preserving skeletal muscle mass in overweight and obese females. Moreover, GWL leads to declines in MST concentrations.
Collapse
Affiliation(s)
- Reza Bagheri
- Department of Exercise Physiology, University of Isfahan, Isfahan, Iran
| | - Damoon Ashtary-Larky
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bradley T Elliott
- Translational Physiology Research Group, School of Life Sciences, University of Westminster, London, UK
| | - Darryn S Willoughby
- School of Exercise and Sport Science, University of Mary Hardin-Baylor, Belton, TX, USA
| | - Mehdi Kargarfard
- Department of Exercise Physiology, University of Isfahan, Isfahan, Iran
| | - Meysam Alipour
- Alimentary Tract Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nasrin Lamuchi-Deli
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Wesam Kooti
- Lung Diseases & Allergy Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Omid Asbaghi
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Alexei Wong
- Department of Health and Human Performance, Marymount University, Arlington, United States
| |
Collapse
|
31
|
Bai L, Wang W, Xiang Y, Wang S, Wan S, Zhu Y. Aberrant elevation of GDF8 impairs granulosa cell glucose metabolism via upregulating SERPINE1 expression in patients with PCOS. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 23:294-309. [PMID: 33425488 PMCID: PMC7779537 DOI: 10.1016/j.omtn.2020.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023]
Abstract
Clinical investigations have demonstrated that polycystic ovary syndrome (PCOS) is often accompanied by insulin resistance (IR) in more than 70% of women with PCOS. However, the etiology of PCOS with IR remains to be characterized. Growth differentiation factor 8 (GDF8) is an intraovarian factor that plays a vital role in the regulation of follicle development and ovulation. Previous studies have reported that GDF8 is a pathogenic factor in glucose metabolism disorder in IR patients. To date, the role of GDF8 on glucose metabolism of granulosa cell in PCOS patients remains to be determined. In the current study, we demonstrated that the expression and accumulation of GDF8 in human granulosa-lutein (hGL) cells and follicular fluid from PCOS patients were higher compared with those of non-PCOS women. GDF8 treatment caused glucose metabolism defects in hGL cells. Transcriptome sequencing results showed that SERPINE1 mediated GDF8-induced impairment of hGL glucose metabolism defects. Using pharmacological and small interfering RNA (siRNA)-mediated knockdown approaches, we demonstrated that GDF8 upregulated the expression of SERPINE1 via the ALK5-mediated SMAD2/3-SMAD4 signaling pathway. Interestingly, the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway was also activated with GDF8 treatment but did not participate in the effect of GDF8 on SERPINE1 expression. Our results also showed that TP53 was required for the GDF8-stimulated increase in SERPINE1 expression. Importantly, our study demonstrated that SB-431542 treatment significantly improved DHEA-induced PCOS-like ovaries. These findings support a potential role for GDF8 in metabolic disorders in PCOS.
Collapse
Affiliation(s)
- Long Bai
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China.,Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China
| | - Wei Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China.,Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China
| | - Yu Xiang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China.,Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China
| | - Shuyi Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China.,Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China
| | - Shan Wan
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China.,Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China
| | - Yimin Zhu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310002, China.,Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310002, China
| |
Collapse
|
32
|
Kakehi S, Tamura Y, Kubota A, Takeno K, Kawaguchi M, Sakuraba K, Kawamori R, Watada H. Effects of blood flow restriction on muscle size and gene expression in muscle during immobilization: A pilot study. Physiol Rep 2020; 8:e14516. [PMID: 32725695 PMCID: PMC7387888 DOI: 10.14814/phy2.14516] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Muscle mass is known to rapidly decrease with muscle disuse. Previous reports suggest that repetitive blood flow restriction (BFR) mitigates the reduction of muscle mass with disuse. However, the effects of BFR on muscle atrophy and gene expression levels in muscle during cast immobilization have not been clarified. METHODS To investigate the effect of BFR on muscle atrophy and gene expression levels during cast immobilization in humans, we recruited 10 healthy males who were randomly divided into the control and BFR treatment groups. All subjects were immobilized with a cast for 14 days. BFR treatment was conducted only in the BFR group. We evaluated cross sectional area (CSA) of thigh muscles by magnetic resonance imaging before and 14 days after cast immobilization. A percutaneous biopsy of the vastus lateralis muscle (VL) was performed before and 1, 7, and 14 days after cast immobilization. Expression of genes related to muscle atrophy and synthesis were evaluated using real-time PCR. RESULTS The CSA of the VL and the thigh flexor muscles were significantly decreased in both groups; however, percent decrease in CSA was significantly smaller in the BFR group compared with the control group. In two-way repeated ANOVA analysis, the time × treatment interaction in gene expression of the muscle-specific ubiquitin ligases muscle ring finger 1 (MuRF1) was significant, and elevated MURF1 expression level by cast immobilization was seemed to be suppressed by the BFR treatment. CONCLUSION BFR treatment may prevent reduced VL and thigh flexor muscles and increased MuRF1 expression level during cast immobilization. Further study is required to confirm these results.
Collapse
Affiliation(s)
- Saori Kakehi
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
- Sportology CenterJuntendo University Graduate School of MedicineTokyoJapan
| | - Yoshifumi Tamura
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
- Sportology CenterJuntendo University Graduate School of MedicineTokyoJapan
| | - Atsushi Kubota
- Department of Sports MedicineJuntendo University Graduate School of Health and Sports ScienceChibaJapan
| | - Kageumi Takeno
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
- Sportology CenterJuntendo University Graduate School of MedicineTokyoJapan
| | - Minako Kawaguchi
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
| | - Keishoku Sakuraba
- Department of Sports MedicineJuntendo University Graduate School of Health and Sports ScienceChibaJapan
| | - Ryuzo Kawamori
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
- Sportology CenterJuntendo University Graduate School of MedicineTokyoJapan
| | - Hirotaka Watada
- Department of Metabolism & EndocrinologyJuntendo University Graduate School of MedicineTokyoJapan
- Sportology CenterJuntendo University Graduate School of MedicineTokyoJapan
- Center for Therapeutic Innovations in DiabetesJuntendo University Graduate School of MedicineTokyoJapan
- Center for Molecular DiabetologyJuntendo University Graduate School of MedicineTokyoJapan
| |
Collapse
|
33
|
Pydi SP, Jain S, Barella LF, Zhu L, Sakamoto W, Meister J, Wang L, Lu H, Cui Y, Gavrilova O, Wess J. β-arrestin-1 suppresses myogenic reprogramming of brown fat to maintain euglycemia. SCIENCE ADVANCES 2020; 6:eaba1733. [PMID: 32548266 PMCID: PMC7274797 DOI: 10.1126/sciadv.aba1733] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/16/2020] [Indexed: 05/05/2023]
Abstract
A better understanding of the signaling pathways regulating adipocyte function is required for the development of new classes of antidiabetic/obesity drugs. We here report that mice lacking β-arrestin-1 (barr1), a cytoplasmic and nuclear signaling protein, selectively in adipocytes showed greatly impaired glucose tolerance and insulin sensitivity when consuming an obesogenic diet. In contrast, transgenic mice overexpressing barr1 in adipocytes were protected against the metabolic deficits caused by a high-calorie diet. Barr1 deficiency led to a myogenic reprogramming of brown adipose tissue (BAT), causing elevated plasma myostatin (Mstn) levels, which in turn led to impaired insulin signaling in multiple peripheral tissues. Additional in vivo studies indicated that barr1-mediated suppression of Mstn expression by BAT is required for maintaining euglycemia. These findings convincingly identify barr1 as a critical regulator of BAT function. Strategies aimed at enhancing barr1 activity in BAT may prove beneficial for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Sai P. Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
- Corresponding author. (J.W.); (S.P.P.)
| | - Shanu Jain
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Luiz F. Barella
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Lu Zhu
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Wataru Sakamoto
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Lei Wang
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Huiyan Lu
- Mouse Transgenic Core Facility, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Yinghong Cui
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
- Corresponding author. (J.W.); (S.P.P.)
| |
Collapse
|
34
|
Dagbay KB, Treece E, Streich FC, Jackson JW, Faucette RR, Nikiforov A, Lin SC, Boston CJ, Nicholls SB, Capili AD, Carven GJ. Structural basis of specific inhibition of extracellular activation of pro- or latent myostatin by the monoclonal antibody SRK-015. J Biol Chem 2020; 295:5404-5418. [PMID: 32075906 PMCID: PMC7170532 DOI: 10.1074/jbc.ra119.012293] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/05/2020] [Indexed: 12/27/2022] Open
Abstract
Myostatin (or growth/differentiation factor 8 (GDF8)) is a member of the transforming growth factor β superfamily of growth factors and negatively regulates skeletal muscle growth. Its dysregulation is implicated in muscle wasting diseases. SRK-015 is a clinical-stage mAb that prevents extracellular proteolytic activation of pro- and latent myostatin. Here we used integrated structural and biochemical approaches to elucidate the molecular mechanism of antibody-mediated neutralization of pro-myostatin activation. The crystal structure of pro-myostatin in complex with 29H4-16 Fab, a high-affinity variant of SRK-015, at 2.79 Å resolution revealed that the antibody binds to a conformational epitope in the arm region of the prodomain distant from the proteolytic cleavage sites. This epitope is highly sequence-divergent, having only limited similarity to other closely related members of the transforming growth factor β superfamily. Hydrogen/deuterium exchange MS experiments indicated that antibody binding induces conformational changes in pro- and latent myostatin that span the arm region, the loops contiguous to the protease cleavage sites, and the latency-associated structural elements. Moreover, negative-stain EM with full-length antibodies disclosed a stable, ring-like antigen-antibody structure in which the two Fab arms of a single antibody occupy the two arm regions of the prodomain in the pro- and latent myostatin homodimers, suggesting a 1:1 (antibody:myostatin homodimer) binding stoichiometry. These results suggest that SRK-015 binding stabilizes the latent conformation and limits the accessibility of protease cleavage sites within the prodomain. These findings shed light on approaches that specifically block the extracellular activation of growth factors by targeting their precursor forms.
Collapse
Affiliation(s)
| | - Erin Treece
- Scholar Rock Inc., Cambridge, Massachusetts 02139
| | | | | | | | | | - Susan C Lin
- Scholar Rock Inc., Cambridge, Massachusetts 02139
| | | | | | | | | |
Collapse
|
35
|
Abe T, Bell ZW, Wong V, Spitz RW, Loenneke JP. Why is low body fat rarely seen in large-sized male athletes? Am J Hum Biol 2020; 32:e23399. [PMID: 32022361 DOI: 10.1002/ajhb.23399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES It is unknown why low body fat is rarely seen in large-sized athletes (>100 kg body mass). The aim of this review was to examine the relationship between body mass and body composition (fat mass and fat-free mass) in elite male athletes, and to discuss the possible reasons why low body fat is rarely seen in large-sized male athletes. METHODS A search using two electronic databases was conducted. Eighteen studies estimated body composition in elite athletes by dual-energy X-ray absorptiometry, totaling 2249 elite male athletes and 72 data points. RESULTS Our results indicated that low body fat (eg, less than 10% body fat) was rarely seen in large-sized male athletes over 100 kg body mass. The larger the body mass, the higher the fat-free mass, with fat-free mass leveling off when body mass exceeds approximately 120 kg. CONCLUSION Possible reasons for this are unknown but we provide some ideas for why this might occur. The two different stages to consider with respect to skeletal muscle growth: the amount of growth during development and the amount of growth as a result of long-term resistance training. In certain sporting events, a large body mass may be favored. However, the large-sized athletes have to balance any potential positive influence of body mass on sports performance with the potential negative factors associated with body fat accumulation. Further research is warranted, as there is currently limited evidence on this topic.
Collapse
Affiliation(s)
- Takashi Abe
- Department of Health, Exercise Science, & Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, Mississippi
| | - Zachary W Bell
- Department of Health, Exercise Science, & Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, Mississippi
| | - Vickie Wong
- Department of Health, Exercise Science, & Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, Mississippi
| | - Robert W Spitz
- Department of Health, Exercise Science, & Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, Mississippi
| | - Jeremy P Loenneke
- Department of Health, Exercise Science, & Recreation Management, Kevser Ermin Applied Physiology Laboratory, The University of Mississippi, University, Mississippi
| |
Collapse
|
36
|
Castillero E, Akashi H, Najjar M, Ji R, Brandstetter LM, Wang C, Liao X, Zhang X, Sperry A, Gailes M, Guaman K, Recht A, Schlosberg I, Sweeney HL, Ali ZA, Homma S, Colombo PC, Ferrari G, Schulze PC, George I. Activin type II receptor ligand signaling inhibition after experimental ischemic heart failure attenuates cardiac remodeling and prevents fibrosis. Am J Physiol Heart Circ Physiol 2019; 318:H378-H390. [PMID: 31886717 DOI: 10.1152/ajpheart.00302.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Myostatin (MSTN) is a transforming growth factor (TGF)-β superfamily member that acts as a negative regulator of muscle growth and may play a role in cardiac remodeling. We hypothesized that inhibition of activin type II receptors (ACTRII) to reduce MSTN signaling would reduce pathological cardiac remodeling in experimental heart failure (HF). C57BL/6J mice underwent left anterior descending coronary artery ligation under anesthesia to induce myocardial infarction (MI) or no ligation (sham). MI and sham animals were each randomly divided into groups (n ≥ 10 mice/group) receiving an ACTRII or ACTRII/TGFβ receptor-signaling inhibiting strategy: 1) myo-Fc group (weekly 10 mg/kg Myo-Fc) or 2) Fol + TGFi group (daily 12 µg/kg follistatin plus 2 mg/kg TGFβ receptor inhibitor), versus controls. ACTRII/TGFBR signaling inhibition preserved cardiac function by echocardiography and prevented an increase in brain natriuretic peptide (BNP). ACTRII/TGFBR inhibition resulted in increased phosphorylation (P) of Akt and decreased P-p38 mitogen-activated protein kinase (MAPK) in MI mice. In vitro, Akt contributed to P-SMAD2,3, P-p38, and BNP regulation in cardiomyocytes. ACTRII/TGFBR inhibition increased sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) levels and decreased unfolded protein response (UPR) markers in MI mice. ACTRII/TGFBR inhibition was associated with a decrease in cardiac fibrosis and fibrosis markers, connective tissue growth factor (CTGF), type I collagen, fibronectin, α-smooth muscle actin, and matrix metalloproteinase (MMP)-12 in MI mice. MSTN exerted a direct regulation on the UPR marker eukaryotic translation initiation factor-2α (eIf2α) in cardiomyocytes. Our study suggests that ACTRII ligand inhibition has beneficial effects on cardiac signaling and fibrosis after ischemic HF.NEW & NOTEWORTHY Activin type II receptor ligand inhibition resulted in preserved cardiac function, a decrease in cardiac fibrosis, improved SERCA2a levels, and a prevention of the unfolded protein response in mice with myocardial infarction.
Collapse
Affiliation(s)
- Estibaliz Castillero
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Hirokazu Akashi
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Marc Najjar
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Ruiping Ji
- Division of Cardiology, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Lea Maria Brandstetter
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Catherine Wang
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Xianghai Liao
- Division of Cardiology, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Xiaokan Zhang
- Division of Cardiology, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Alexandra Sperry
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Marcia Gailes
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Karina Guaman
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Adam Recht
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Ira Schlosberg
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - H Lee Sweeney
- Department of Pharmacology, University of Florida, Gainesville, Florida
| | - Ziad A Ali
- Division of Cardiology, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Shunichi Homma
- Division of Cardiology, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Paolo C Colombo
- Division of Cardiology, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Giovanni Ferrari
- Division of Surgical Science, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| | - P Christian Schulze
- Division of Cardiology, Department of Medicine, College of Physicians and Surgeons of Columbia University, New York, New York
| | - Isaac George
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons of Columbia University, New York, New York
| |
Collapse
|
37
|
Tavoian D, Arnold WD, Mort SC, de Lacalle S. Sex differences in body composition but not neuromuscular function following long-term, doxycycline-induced reduction in circulating levels of myostatin in mice. PLoS One 2019; 14:e0225283. [PMID: 31751423 PMCID: PMC6872155 DOI: 10.1371/journal.pone.0225283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/31/2019] [Indexed: 02/05/2023] Open
Abstract
Age-related declines in muscle function result from changes in muscle structure and contractile properties, as well as from neural adaptations. Blocking myostatin to drive muscle growth is one potential therapeutic approach. While the effects of myostatin depletion on muscle characteristics are well established, we have very little understanding of its effects on the neural system. Here we assess the effects of long-term, post-developmental myostatin reduction on electrophysiological motor unit characteristics and body composition in aging mice. We used male (N = 21) and female (N = 26) mice containing a tetracycline-inducible system to delete the myostatin gene in skeletal muscle. Starting at 12 months of age, half of the mice were administered doxycycline (tetracycline) through their chow for one year. During that time we measured food intake, body composition, and hindlimb electromyographic responses. Doxycycline-induced myostatin reduction had no effect on motor unit properties for either sex, though significant age-dependent declines in motor unit number occurred in all mice. However, treatment with doxycycline induced different changes in body composition between sexes. All female mice increased in total, lean and fat mass, but doxycycline-treated female mice experienced a significantly larger increase in lean mass than controls. All male mice also increased total and lean mass, but administration of doxycycline had no effect. Additionally, doxycycline-treated male mice maintained their fat mass at baseline levels, while the control group experienced a significant increase from baseline and compared to the doxycycline treated group. Our results show that long-term administration of doxycycline results in body composition adaptations that are distinctive between male and female mice, and that the effects of myostatin reduction are most pronounced during the first three months of treatment. We also report that age-related changes in motor unit number are not offset by reduced myostatin levels, despite increased lean mass exhibited by female mice.
Collapse
Affiliation(s)
- Dallin Tavoian
- Program in Translational Biomedical Sciences, 1 Ohio University, Athens, OH, United States of America
| | - W. David Arnold
- Departments of Neurology, PM&R, and Neuroscience, and Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States of America
| | - Sophia C. Mort
- Program in Translational Biomedical Sciences, 1 Ohio University, Athens, OH, United States of America
| | - Sonsoles de Lacalle
- Sonsoles de Lacalle, Department of Biomedical Sciences,1 Ohio University, Athens, OH, United States of America
- * E-mail:
| |
Collapse
|
38
|
Molecular characterization, expression analysis of myostatin gene and its negative regulation by miR-29b-3p in Chinese concave-eared frogs (Odorrana tormota). Comp Biochem Physiol B Biochem Mol Biol 2019; 240:110369. [PMID: 31676334 DOI: 10.1016/j.cbpb.2019.110369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/08/2019] [Accepted: 09/26/2019] [Indexed: 01/09/2023]
Abstract
The molecular characteristics, expression patterns and functions of the amphibian myostatin (MSTN) gene are unknown. Here, we isolated a full-length Odorrana tormota MSTN cDNA sequence of 1701 bp (Ot-MSTN), containing a putative N-terminal signal peptide, a TGF-β propeptide domain and an active peptide. Ot-MSTN was expressed in 9 selected tissues examined, and the highest level of expression was in thigh muscle, followed by brain and female gonadal tissue. The expression of Ot-MSTN in multiple O. tormota tissues supported that the activities of MSTN may be not limited to skeletal muscle. Ot-MSTN expression was decreased from stage 31 to stage 40, while the growth rate was increased. The expression of Ot-MSTN in adult male frogs increased with age, indicating that adult male frogs may inhibit the continued hypertrophy of thigh muscle fibers and decrease the growth rate of thigh muscle to ensure muscles do not grow too large. Luciferase reporter assays showed that miR-29b-3p directly targeted the 3'-UTR of Ot-MSTN. miR-29b-3p expression in the thigh muscle of 2 yrs. females who grew faster was significantly lower than that of the slow-growing 2 yrs. male individuals, which showed an opposite trend with Ot-MSTN expression. In addition,miR-29b-3p expression reversed trends of Ot-MSTN expression at different developmental stages in thigh muscle. Therefore, these data indicate that miR-29-3p may negatively regulate the expression of MSTN and regulate thigh muscle growth and development in O. tormota.
Collapse
|
39
|
Chimeric Myostatin — Tetanic Toxin Epitopes and Heterologous Prime-boost Immunization Improve Immune Response Stimulating Muscle Growth in Mice. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-019-0092-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
40
|
Ikeda K, Horie-Inoue K, Inoue S. Functions of estrogen and estrogen receptor signaling on skeletal muscle. J Steroid Biochem Mol Biol 2019; 191:105375. [PMID: 31067490 DOI: 10.1016/j.jsbmb.2019.105375] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/03/2019] [Indexed: 12/23/2022]
Abstract
Activity of estrogen, a sex steroid hormone, is not only limited to the reproductive organs but also involves other organs and tissues, including skeletal muscle. In postmenopausal women, estrogen decline causes endocrine and metabolic dysfunction, leading to a predisposition to osteoporosis, metabolic syndrome, and decreased muscle mass and strength. The decline in skeletal muscle mass often associates with sarcopenia, a popular condition observed in fragile elder people. In addition, varying estrogen levels associated with the menstrual phases may modulate exercise performance in women. Estrogen is thus considered to play a crucial role in skeletal muscle homeostasis and exercise capacity, although its precise mechanisms remain to be elucidated. In this article, we review the role of estrogen in the skeletal muscle, outlining the proposed molecular mechanisms. We especially focus on the current understanding of estrogen actions on mitochondria metabolism in skeletal muscle.
Collapse
Affiliation(s)
- Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan; Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.
| |
Collapse
|
41
|
Garbern J, Kristl AC, Bassaneze V, Vujic A, Schoemaker H, Sereda R, Peng L, Ricci-Blair EM, Goldstein JM, Walker RG, Bhasin S, Wagers AJ, Lee RT. Analysis of Cre-mediated genetic deletion of Gdf11 in cardiomyocytes of young mice. Am J Physiol Heart Circ Physiol 2019; 317:H201-H212. [PMID: 31125255 PMCID: PMC6692736 DOI: 10.1152/ajpheart.00615.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 01/19/2023]
Abstract
Administration of active growth differentiation factor 11 (GDF11) to aged mice can reduce cardiac hypertrophy, and low serum levels of GDF11 measured together with the related protein, myostatin (also known as GDF8), predict future morbidity and mortality in coronary heart patients. Using mice with a loxP-flanked ("floxed") allele of Gdf11 and Myh6-driven expression of Cre recombinase to delete Gdf11 in cardiomyocytes, we tested the hypothesis that cardiac-specific Gdf11 deficiency might lead to cardiac hypertrophy in young adulthood. We observed that targeted deletion of Gdf11 in cardiomyocytes does not cause cardiac hypertrophy but rather leads to left ventricular dilation when compared with control mice carrying only the Myh6-cre or Gdf11-floxed alleles, suggesting a possible etiology for dilated cardiomyopathy. However, the mechanism underlying this finding remains unclear because of multiple confounding effects associated with the selected model. First, whole heart Gdf11 expression did not decrease in Myh6-cre; Gdf11-floxed mice, possibly because of upregulation of Gdf11 in noncardiomyocytes in the heart. Second, we observed Cre-associated toxicity, with lower body weights and increased global fibrosis, in Cre-only control male mice compared with flox-only controls, making it challenging to infer which changes in Myh6-cre;Gdf11-floxed mice were the result of Cre toxicity versus deletion of Gdf11. Third, we observed differential expression of cre mRNA in Cre-only controls compared with the cardiomyocyte-specific knockout mice, also making comparison between these two groups difficult. Thus, targeted Gdf11 deletion in cardiomyocytes may lead to left ventricular dilation without hypertrophy, but alternative animal models are necessary to understand the mechanism for these findings. NEW & NOTEWORTHY We observed that targeted deletion of growth differentiation factor 11 in cardiomyocytes does not cause cardiac hypertrophy but rather leads to left ventricular dilation compared with control mice carrying only the Myh6-cre or growth differentiation factor 11-floxed alleles. However, the mechanism underlying this finding remains unclear because of multiple confounding effects associated with the selected mouse model.
Collapse
Affiliation(s)
- Jessica Garbern
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
- Department of Cardiology, Boston Children's Hospital , Boston, Massachusetts
| | - Amy C Kristl
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Vinicius Bassaneze
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Ana Vujic
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Henk Schoemaker
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| | - Rebecca Sereda
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Liming Peng
- Brigham Research Assay Core, Brigham and Women's Hospital , Boston, Massachusetts
| | - Elisabeth M Ricci-Blair
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Jill M Goldstein
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Ryan G Walker
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
| | - Shalender Bhasin
- Brigham Research Assay Core, Brigham and Women's Hospital , Boston, Massachusetts
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School , Boston, Massachusetts
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center , Boston, Massachusetts
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University , Cambridge, Massachusetts
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts
| |
Collapse
|
42
|
Paul RG, Hennebry AS, Elston MS, Conaglen JV, McMahon CD. Regulation of murine skeletal muscle growth by STAT5B is age- and sex-specific. Skelet Muscle 2019; 9:19. [PMID: 31230596 PMCID: PMC6589877 DOI: 10.1186/s13395-019-0204-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/11/2019] [Indexed: 01/22/2023] Open
Abstract
Background Sexually dimorphic growth has been attributed to the growth hormone (GH)/insulin-like growth factor 1 (IGF1) axis, particularly GH-induced activation of the intracellular signal transducer and activator of transcription 5B (STAT5B), because deletion of STAT5B reduces body mass and the mass of skeletal muscles in male mice to that in female mice. However, it remains unclear why these effects are sex- and species-specific, because the loss of STAT5B retards growth in girls, but not in male mice. Our objectives were to determine whether sexually dimorphic growth of skeletal muscle persisted in STAT5B−/− mice and investigate the mechanisms by which STAT5B regulates sexually dimorphic growth. Methods Blood and skeletal muscle were harvested from male and female STAT5B−/− mice and their wild-type littermates from the onset of puberty to adulthood. Results Growth of the skeleton and skeletal muscles was retarded in both sexes of STAT5B−/− mice, but more so in males. Although reduced, sexually dimorphic growth of skeletal muscle persisted in STAT5B−/− mice with an oxidative shift in the composition of myofibres in both sexes. Concentrations of IGF1 in blood and skeletal muscle were reduced in male STAT5B−/− mice at all ages, but only in female STAT5B−/− mice at the onset of puberty. Expression of androgen receptor (AR) and oestrogen receptor alpha (ERα) mRNA and protein was reduced in skeletal muscles of male and female STAT5B−/− mice, respectively. Loss of STAT5B abolished the sexually dimorphic expression of myostatin protein and Igf1, Ar, Erα, suppressor of cytokine signalling 2 (Socs2), and cytokine-inducible SH2-containing protein (Cis) mRNA in skeletal muscle. Conclusions STAT5B appears to mediate GH signalling in skeletal muscles of male mice at all ages, but only until puberty in female mice. STAT5B also appears to mediate the actions of androgens and oestrogens in both male and female mice, but sexually dimorphic growth persists in STAT5B−/− mice. Electronic supplementary material The online version of this article (10.1186/s13395-019-0204-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ryan G Paul
- AgResearch Ltd, Ruakura Research Centre, Private Bag 3123, Hamilton, New Zealand. .,Faculty of Medical & Health Sciences, Waikato Clinical Campus, University of Auckland, Private Bag 3200, Hamilton, 3240, New Zealand.
| | - Alex S Hennebry
- AgResearch Ltd, Ruakura Research Centre, Private Bag 3123, Hamilton, New Zealand
| | - Marianne S Elston
- Faculty of Medical & Health Sciences, Waikato Clinical Campus, University of Auckland, Private Bag 3200, Hamilton, 3240, New Zealand
| | - John V Conaglen
- Faculty of Medical & Health Sciences, Waikato Clinical Campus, University of Auckland, Private Bag 3200, Hamilton, 3240, New Zealand
| | - Chris D McMahon
- AgResearch Ltd, Ruakura Research Centre, Private Bag 3123, Hamilton, New Zealand
| |
Collapse
|
43
|
Kim JH, Kim JH, Sutikno LA, Lee SB, Jin DH, Hong YK, Kim YS, Jin HJ. Identification of the minimum region of flatfish myostatin propeptide (Pep45-65) for myostatin inhibition and its potential to enhance muscle growth and performance in animals. PLoS One 2019; 14:e0215298. [PMID: 30998775 PMCID: PMC6472743 DOI: 10.1371/journal.pone.0215298] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 03/30/2019] [Indexed: 12/31/2022] Open
Abstract
Myostatin (MSTN) negatively regulates skeletal muscle growth, and its activity is inhibited by the binding of MSTN propeptide (MSTNpro), the N-terminal domain of proMSTN that is proteolytically cleaved from the proMSTN. Partial sequences from the N-terminal side of MSTNpro have shown to be sufficient to inhibit MSTN activity. In this study, to determine the minimum size of flatfish MSTNpro for MSTN inhibition, various truncated forms of flatfish MSTNpro with N-terminal maltose binding protein (MBP) fusion were expressed in E. coli and purified. MSTNpro regions consisting of residues 45–68, -69, and -70 with MBP fusion suppressed MSTN activity with a potency comparable to that of full-sequence flatfish MSTNpro in a pGL3-(CAGA)12-luciferase reporter assay. Even though the MSTN-inhibitory potency was about 1,000-fold lower, the flatfish MSTNpro region containing residues 45–65 (MBP-Pro45-65) showed MSTN-inhibitory capacity but not the MBP-Pro45-64, indicating that the region 45–65 is the minimum domain required for MSTN binding and suppression of its activity. To examine the in vivo effect of MBP-fused, truncated flatfish MSTNpro, MBP-Pro45-70-His6 (20 mg/kg body wt) was subcutaneously injected 5 times for 14 days in mice. Body wt gain and bone mass were not affected by the administration. Grip strength and swimming time were significantly enhanced at 7 d after the administration. At 14 d, the effect on grip strength disappeared, and the extent of the effect on swimming time significantly diminished. The presence of antibody against MBP-Pro45-70-His6 was observed at both 7 and 14 d after the administration with the titer value at 14 d being much greater than that at 7 d, suggesting that antibodies against MBP-Pro45-70-His6 neutralized the MSTN-inhibitory effect of MBP-Pro45-70-His6. We, thus, examined the MSTN-inhibitory capacity and in vivo effect of flatfish MSTNpro region 45–65 peptide (Pep45-65-NH2), which was predicted to have no immunogenicity in silico analysis. Pep45-65-NH2 suppressed MSTN activity with a potency similar to that of MBP-Pro45-65 but did not suppress GDF11, or activin A. Pep45-65-NH2 blocked MSTN-induced Smad2 phosphorylation in HepG2 cells. The administration of Pep45-65 (20 mg/kg body wt, 5 times for 2 weeks) increased the body wt gain with a greater gain at 14 d than at 7 d and muscle wt. Grip strength and swimming time were also significantly enhanced by the administration. Antibody titer against Pep45-65 was not detected. In conclusion, current results indicate that MSTN-inhibitory proteins with heterologous fusion partner may not be effective in suppressing MSTN activity in vivo due to an immune response against the proteins. Current results also show that the region of flatfish MSTNpro consisting of 45–65 (Pep45-65) can suppress mouse MSTN activity and increase muscle mass and function without invoking an immune response, implying that Pep45-65 would be a potential agent to enhance skeletal muscle growth and function in animals or to treat muscle atrophy caused by various clinical conditions.
Collapse
Affiliation(s)
- Jeong Hwan Kim
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung-si, Ganwon-do, Korea
| | - Jeong Han Kim
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung-si, Ganwon-do, Korea
| | | | - Sang Beum Lee
- Department of Human Nurtrition, Food and Animal Sciences, University of Hawaii, Honolulu, Hawaii, United States of America
| | - Deuk-Hee Jin
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung-si, Ganwon-do, Korea
| | - Yong-Ki Hong
- Department of Biotechnology, Pukyong National University, Namgu, Busan, Korea
| | - Yong Soo Kim
- Department of Human Nurtrition, Food and Animal Sciences, University of Hawaii, Honolulu, Hawaii, United States of America
- * E-mail: (YK); (HJ)
| | - Hyung-Joo Jin
- Department of Marine Molecular Bioscience, Gangneung-Wonju National University, Gangneung-si, Ganwon-do, Korea
- * E-mail: (YK); (HJ)
| |
Collapse
|
44
|
Zhang Y, Wang Y, Yulin B, Tang B, Wang M, Zhang C, Zhang W, Jin J, Li T, Zhao R, Yu X, Zuo Q, Li B. CRISPR/Cas9-mediated sheep MSTN gene knockout and promote sSMSCs differentiation. J Cell Biochem 2019; 120:1794-1806. [PMID: 30242885 DOI: 10.1002/jcb.27474] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 07/20/2018] [Indexed: 01/24/2023]
Abstract
Myostatin (MSTN) is an important gene involved in the regulation of embryonic muscle cells and adult muscle development; it has a good application prospect in transgenic animal production by improving the yield of muscle. The purpose of this study is to construct MSTN gene knockout vector using clustered regularly interspaced short palindromic repeats ( CRISPR)/CRISPR-associated protein 9 ( Cas9). The knockout efficiency was evaluated in sheep ear fibroblasts (SEFs) by cleavage activity of transcription of guide RNA ( gRNA), luciferase-single-strand annealing assay, T7 endonuclease I assay (T7E1), and TA clone sequence (10/38); and above all, detection showed that the cleavage activity of CRISPR/Cas9-mediated MSTN reached 29%. MSTN-Cas9/gRNA4 was transfected into sheep skeletal muscle satellite cell (sSMSC) to confirm the function of MSTN in myotomes formation induced by starvation in low-serum medium. The results showed that myotubes formation efficiency were 11.2 ± 1.3% and 19.5 ± 2.1% in the control group and knockout group, respectively. The average length of myotomes was 22 ± 5.3 and 47 ± 3.6 μm, displaying that MSTN knockout can promote sSMSC differentiation in number and length. The unlabeled MSTN-Cas9/gRNA4 was transfected into SEFs and monoclonal positive cells was obtained after 48 hours transfection. The MSTN-positive cells were used as donor cells to perform somatic cell nuclear transplantation to produce transgenic sheep. A total of 20 embryos were transplanted into surrogate mothers, four of them normally produce offspring. The genomic DNA of surviving lambs were used as a template, three positive individuals were identified by T7E1 digestion. All the results demonstrated that the CRISPR/Cas9 system has the potential to become an important and applicable gene engineering tool in animal breeding.
Collapse
Affiliation(s)
- Yani Zhang
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Yingjie Wang
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Bi Yulin
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Beibei Tang
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Man Wang
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Chen Zhang
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Wenhui Zhang
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Jing Jin
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | | | - Ruifeng Zhao
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Xinjian Yu
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Qisheng Zuo
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| | - Bichun Li
- Institutes of Agricultural Science and Technology Development, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology,Yangzhou University, Yangzhou, China
| |
Collapse
|
45
|
Kemp PR, Griffiths M, Polkey MI. Muscle wasting in the presence of disease, why is it so variable? Biol Rev Camb Philos Soc 2018; 94:1038-1055. [PMID: 30588725 DOI: 10.1111/brv.12489] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 11/22/2018] [Accepted: 11/27/2018] [Indexed: 12/16/2022]
Abstract
Skeletal muscle wasting is a common clinical feature of many chronic diseases and also occurs in response to single acute events. The accompanying loss of strength can lead to significant disability, increased care needs and have profound negative effects on quality of life. As muscle is the most abundant source of amino acids in the body, it appears to function as a buffer for fuel and substrates that can be used to repair damage elsewhere and to feed the immune system. In essence, the fundamentals of muscle wasting are simple: less muscle is made than is broken down. However, although well-described mechanisms modulate muscle protein turnover, significant individual differences in the amount of muscle lost in the presence of a given severity of disease complicate the understanding of underlying mechanisms and suggest that individuals have different sensitivities to signals for muscle loss. Furthermore, the rate at which muscle protein is turned over under normal conditions means that clinically significant muscle loss can occur with changes in the rate of protein synthesis and/or breakdown that are too small to be measurable. Consequently, the changes in expression of factors regulating muscle turnover required to cause a decline in muscle mass are small and, except in cases of rapid wasting, there is no consistent pattern of change in the expression of factors that regulate muscle mass. MicroRNAs are fine tuners of cell phenotype and are therefore ideally suited to cause the subtle changes in proteome required to tilt the balance between synthesis and degradation in a way that causes clinically significant wasting. Herein we present a model in which muscle loss as a consequence of disease in non-muscle tissue is modulated by a set of microRNAs, the muscle expression of which is associated with severity of disease in the non-muscle tissue. These microRNAs alter fundamental biological processes including the synthesis of ribosomes and mitochondria leading to reduced protein synthesis and increased protein breakdown, thereby freeing amino acids from the muscle. We argue that the variability in muscle loss observed in the human population arises from at least two sources. The first is from pre-existing or disease-induced variation in the expression of microRNAs controlling the sensitivity of muscle to the atrophic signal and the second is from the expression of microRNAs from imprinted loci (i.e. only expressed from the maternally or paternally inherited allele) and may control the rate of myonuclear recruitment. In the absence of disease, these factors do not correlate with muscle mass, since there is no challenge to the established balance. However, in the presence of such a challenge, these microRNAs determine the rate of decline for a given disease severity. Together these mechanisms provide novel insight into the loss of muscle mass and its variation in the human population. The involvement of imprinted loci also suggests that genes that regulate early development also contribute to the ability of individuals to resist muscle loss in response to disease.
Collapse
Affiliation(s)
- Paul R Kemp
- National Heart & Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, U.K
| | - Mark Griffiths
- National Heart & Lung Institute, Imperial College London, South Kensington Campus, London, SW7 2AZ, U.K
| | - Michael I Polkey
- National Institute for Health Research Respiratory Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, Sydney Street, London SW3 6NP, U.K
| |
Collapse
|
46
|
Zhou Y, Zhang Y, Zhu D. Myostatin promotes the epithelial-to-mesenchymal transition of the dermomyotome during somitogenesis. Dev Dyn 2018; 247:1241-1252. [PMID: 30325085 DOI: 10.1002/dvdy.24681] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 09/30/2018] [Accepted: 10/10/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Myostatin (MSTN), a member of the transforming growth factor-β (TGF-β) superfamily, has been implicated in the negative regulation of skeletal myogenesis. However, the molecular mechanism through which MSTN regulates early embryonic myogenesis is not well understood. RESULTS We demonstrate that MSTN regulates early embryonic myogenesis by promoting the epithelial-to-mesenchymal transition (EMT) of the dermomyotome during somitogenesis in chicks. We show that the MSTN gene is first expressed at the center of the dermomyotome. As somitogenesis progresses, its expression extends dorsally and ventrally along the plane of the dermomyotome. By combining in situ hybridization and immunofluorescence assays, we demonstrate that the expression pattern of MSTN is spatiotemporally well correlated with EMT of the dermomyotome. Our gain- and loss-of-function experiments further reveal that MSTN can induce EMT of the chick dermomyotome. We also show that MSTN induces EMT of a nonsmall cell lung carcinoma cell line (A549) and Madin-Darby canine kidney cells in vitro. CONCLUSIONS Our experimental data suggest that MSTN regulates myogenesis by promoting EMT during somitogenesis. These findings provide novel insights into the functions of MSTN during early embryonic myogenesis. Developmental Dynamics 247:1241-1252, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yuchang Zhou
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, P R China
| | - Yong Zhang
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, P R China
| | - Dahai Zhu
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, P R China
| |
Collapse
|
47
|
Golan T, Geva R, Richards D, Madhusudan S, Lin BK, Wang HT, Walgren RA, Stemmer SM. LY2495655, an antimyostatin antibody, in pancreatic cancer: a randomized, phase 2 trial. J Cachexia Sarcopenia Muscle 2018; 9:871-879. [PMID: 30051975 PMCID: PMC6204586 DOI: 10.1002/jcsm.12331] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 06/10/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Cachexia is a formidable clinical challenge in pancreatic cancer. We assessed LY2495655 (antimyostatin antibody) plus standard-of-care chemotherapy in pancreatic cancer using cachexia status as a stratifier. METHODS In this randomized, phase 2 trial, patients with stage II-IV pancreatic cancer were randomized to 300 mg LY2495655, 100 mg LY2495655, or placebo, plus physician-choice chemotherapy from a prespecified list of standard-of-care regimens for first and later lines of care. Investigational treatment was continued during second-line treatment. The primary endpoint was overall survival. RESULTS Overall, 125 patients were randomized. In August 2014, 300 mg LY2495655 was terminated due to imbalance in death rates between the treatment arms; in January 2015, 100 mg LY2495655 treatment was terminated due to futility. LY2495655 did not improve overall survival: the hazard ratio was 1.70 (90% confidence interval, 1.1-2.7) for 300 mg vs. placebo and 1.3 (0.82-2.1) for 100 mg vs. placebo (recommended doses). Progression-free survival results were consistent with the overall survival results. A numerically higher hazard ratio was observed in patients with weight loss (WL) of ≥5% (cachexia) than with <5% WL within 6 months before randomization. Subgroup analyses for patients stratified by WL in the 6 months preceding enrollment suggested that functional responses to LY2495655 (either dose) may have been superior in patients with <5% WL vs. patients with ≥5% WL. Among possibly drug-related adverse events, fatigue, diarrhoea, and anorexia were more common in LY2495655-treated than in placebo-treated patients. CONCLUSIONS In the intention-to-treat analysis, LY2495655 did not confer clinical benefit in pancreatic cancer. Our data highlight the importance of assessing survival when investigating therapeutic management of cachexia and support the use of WL as a stratifier (independent of performance status).
Collapse
Affiliation(s)
| | - Ravit Geva
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | | | - Srinivasan Madhusudan
- Academic Oncology, University of Nottingham, School of Medicine, Nottingham University Hospitals, City Hospital Campus, Nottingham, UK
| | | | | | | | - Salomon M Stemmer
- Rabin Medical Center, Petach Tikva, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
48
|
Renzini A, Marroncelli N, Noviello C, Moresi V, Adamo S. HDAC4 Regulates Skeletal Muscle Regeneration via Soluble Factors. Front Physiol 2018; 9:1387. [PMID: 30319457 PMCID: PMC6171007 DOI: 10.3389/fphys.2018.01387] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle possesses a high ability to regenerate after an insult or in pathological conditions, relying on satellite cells, the skeletal muscle stem cells. Satellite cell behavior is tightly regulated by the surrounding microenvironment, which provides multiple signals derived from local cells and systemic factors. Among epigenetic mechanisms, histone deacetylation has been proved to affect muscle regeneration. Indeed, pan-histone deacetylase inhibitors were found to improve muscle regeneration, while deletion of histone deacetylase 4 (HDAC4) in satellite cells inhibits their proliferation and differentiation, leading to compromised muscle regeneration. In this study, we delineated the HDAC4 function in adult skeletal muscle, following injury, by using a tissue-specific null mouse line. We showed that HDAC4 is crucial for skeletal muscle regeneration by mediating soluble factors that influence muscle-derived cell proliferation and differentiation. These findings add new biological functions to HDAC4 in skeletal muscle that need considering when administering histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Alessandra Renzini
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Nicoletta Marroncelli
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Chiara Noviello
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| | - Viviana Moresi
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy.,Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Sergio Adamo
- DAHFMO Unit of Histology and Medical Embryology, Interuniversity Institute of Myology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
49
|
Butcher JT, Ali MI, Ma MW, McCarthy CG, Islam BN, Fox LG, Mintz JD, Larion S, Fulton DJ, Stepp DW. Effect of myostatin deletion on cardiac and microvascular function. Physiol Rep 2018; 5. [PMID: 29192067 PMCID: PMC5727279 DOI: 10.14814/phy2.13525] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 10/13/2017] [Accepted: 10/20/2017] [Indexed: 12/20/2022] Open
Abstract
The objective of this study is to test the hypothesis that increased muscle mass has positive effects on cardiovascular function. Specifically, we tested the hypothesis that increases in lean body mass caused by deletion of myostatin improves cardiac performance and vascular function. Echocardiography was used to quantify left ventricular function at baseline and after acute administration of propranolol and isoproterenol to assess β‐adrenergic reactivity. Additionally, resistance vessels in several beds were removed, cannulated, pressurized to 60 mmHg and reactivity to vasoactive stimuli was assessed. Hemodynamics were measured using in vivo radiotelemetry. Myostatin deletion results in increased fractional shortening at baseline. Additionally, arterioles in the coronary and muscular microcirculations are more sensitive to endothelial‐dependent dilation while nonmuscular beds or the aorta were unaffected. β‐adrenergic dilation was increased in both coronary and conduit arteries, suggesting a systemic effect of increased muscle mass on vascular function. Overall hemodynamics and physical characteristics (heart weight and size) remained unchanged. Myostatin deletion mimics in part the effects of exercise on cardiovascular function. It significantly increases lean muscle mass and results in muscle‐specific increases in endothelium‐dependent vasodilation. This suggests that increases in muscle mass may serve as a buffer against pathological states that specifically target cardiac function (heart failure), the β‐adrenergic system (age), and nitric oxide bio‐availability (atherosclerosis). Taken together, pharmacological inhibition of the myostatin pathway could prove an excellent mechanism by which the benefits of exercise can be conferred in patients that are unable to exercise.
Collapse
Affiliation(s)
- Joshua T Butcher
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| | - M Irfan Ali
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Merry W Ma
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Cameron G McCarthy
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Bianca N Islam
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Lauren G Fox
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| | - James D Mintz
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Sebastian Larion
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| | - David J Fulton
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| | - David W Stepp
- Department of Pharmacology, Department of Physiology, Vascular Biology Center, Augusta University, Augusta, Georgia
| |
Collapse
|
50
|
Grafe I, Alexander S, Peterson JR, Snider TN, Levi B, Lee B, Mishina Y. TGF-β Family Signaling in Mesenchymal Differentiation. Cold Spring Harb Perspect Biol 2018; 10:a022202. [PMID: 28507020 PMCID: PMC5932590 DOI: 10.1101/cshperspect.a022202] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) can differentiate into several lineages during development and also contribute to tissue homeostasis and regeneration, although the requirements for both may be distinct. MSC lineage commitment and progression in differentiation are regulated by members of the transforming growth factor-β (TGF-β) family. This review focuses on the roles of TGF-β family signaling in mesenchymal lineage commitment and differentiation into osteoblasts, chondrocytes, myoblasts, adipocytes, and tenocytes. We summarize the reported findings of cell culture studies, animal models, and interactions with other signaling pathways and highlight how aberrations in TGF-β family signaling can drive human disease by affecting mesenchymal differentiation.
Collapse
Affiliation(s)
- Ingo Grafe
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Stefanie Alexander
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Jonathan R Peterson
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Taylor Nicholas Snider
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Benjamin Levi
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|