1
|
Kirk B, Lombardi G, Duque G. Bone and muscle crosstalk in ageing and disease. Nat Rev Endocrinol 2025:10.1038/s41574-025-01088-x. [PMID: 40011751 DOI: 10.1038/s41574-025-01088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 02/28/2025]
Abstract
Interorgan communication between bone and skeletal muscle is central to human health. A dysregulation of bone-muscle crosstalk is implicated in several age-related diseases. Ageing-associated changes in endocrine, inflammatory, nutritional and biomechanical stimuli can influence the differentiation capacity, function and survival of mesenchymal stem cells and bone-forming and muscle-forming cells. Consequently, the secretome phenotype of bone and muscle cells is altered, leading to impaired crosstalk and, ultimately, catabolism of both tissues. Adipose tissue acts as a third player in the bone-muscle interaction by secreting factors that affect bone and muscle cells. Physical exercise remains the key biological stimulus for bone-muscle crosstalk, either directly via the release of cytokines from bone, muscle or adipocytes, or indirectly through extracellular vesicles. Overall, bone-muscle crosstalk is considered an inherent process necessary to maintain the structure and function of both tissues across the life cycle. This Review summarizes the latest biomedical advances in bone-muscle crosstalk as it pertains to human ageing and disease. We also outline future research priorities to accommodate the understanding of this rapidly emerging field.
Collapse
Affiliation(s)
- Ben Kirk
- Department of Medicine, Western Health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, Melbourne, Victoria, Australia
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Gustavo Duque
- Department of Medicine, Western Health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia.
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, Melbourne, Victoria, Australia.
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
- Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
2
|
Barber TM, Kabisch S, Pfeiffer AFH, Weickert MO. Optimised Skeletal Muscle Mass as a Key Strategy for Obesity Management. Metabolites 2025; 15:85. [PMID: 39997710 PMCID: PMC11857510 DOI: 10.3390/metabo15020085] [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: 01/07/2025] [Revised: 01/17/2025] [Accepted: 01/19/2025] [Indexed: 02/26/2025] Open
Abstract
The 'Body Mass Index' (BMI) is an anachronistic and outdated ratio that is used as an internationally accepted diagnostic criterion for obesity, and to prioritise, stratify, and outcome-assess its management options. On an individual level, the BMI has the potential to mislead, including inaccuracies in cardiovascular risk assessment. Furthermore, the BMI places excessive emphasis on a reduction in overall body weight (rather than optimised body composition) and contributes towards a misunderstanding of the quiddity of obesity and a dispassionate societal perspective and response to the global obesity problem. The overall objective of this review is to provide an overview of obesity that transitions away from the BMI and towards a novel vista: viewing obesity from the perspective of the skeletal muscle (SM). We resurrect the SM as a tissue hidden in plain sight and provide an overview of the key role that the SM plays in influencing metabolic health and efficiency. We discuss the complex interlinks between the SM and the adipose tissue (AT) through key myokines and adipokines, and argue that rather than two separate tissues, the SM and AT should be considered as a single entity: the 'Adipo-Muscle Axis'. We discuss the vicious circle of sarcopenic obesity, in which aging- and obesity-related decline in SM mass contributes to a worsened metabolic status and insulin resistance, which in turn further compounds SM mass and function. We provide an overview of the approaches that can mitigate against the decline in SM mass in the context of negative energy balance, including the optimisation of dietary protein intake and resistance physical exercises, and of novel molecules in development that target the SM, which will play an important role in the future management of obesity. Finally, we argue that the Adipo-Muscle Ratio (AMR) would provide a more clinically meaningful descriptor and definition of obesity than the BMI and would help to shift our focus regarding its effective management away from merely inducing weight loss and towards optimising the AMR with proper attention to the maintenance and augmentation of SM mass and function.
Collapse
Affiliation(s)
- Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK;
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV1 5FB, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
| | - Stefan Kabisch
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany (A.F.H.P.)
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany (A.F.H.P.)
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße, 85764 Neuherberg, Germany
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK;
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV1 5FB, UK
- NIHR CRF Human Metabolism Research Unit, University Hospitals Coventry and Warwickshire, Clifford Bridge Road, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| |
Collapse
|
3
|
Timofte DV, Tudor RC, Mocanu V, Labusca L. Obesity, Osteoarthritis, and Myokines: Balancing Weight Management Strategies, Myokine Regulation, and Muscle Health. Nutrients 2024; 16:4231. [PMID: 39683624 PMCID: PMC11644804 DOI: 10.3390/nu16234231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity and osteoarthritis (OA) are increasingly prevalent conditions that are intricately linked, with each exacerbating the other's pathogenesis and worsening patient outcomes. This review explores the dual impact of obesity on OA, highlighting the role of excessive weight in aggravating joint degeneration and the limitations OA imposes on physical activity, which further perpetuates obesity. The role of muscle tissue, particularly the release of myokines during physical activity, is examined in the context of OA and obesity. Myokines such as irisin, IL-6, and myostatin are discussed for their roles in metabolic regulation, inflammation, and tissue repair, offering insights into their potential therapeutic targets. This review emphasizes the importance of supervised weight management methods in parallel with muscle rehabilitation in improving joint health and metabolic balance. The potential for myokine modulation through targeted exercise and weight loss interventions to mitigate the adverse effects of obesity and OA is also discussed, suggesting avenues for future research and therapy development to reduce the burden of these chronic conditions.
Collapse
Affiliation(s)
- Daniel Vasile Timofte
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.V.T.); (R.C.T.)
| | - Razvan Cosmin Tudor
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.V.T.); (R.C.T.)
- Dr. Iacob Czihac Military Emergency Hospital Iasi, General Henri Mathias Berthelot Str. 7-9, 700483 Iași, Romania
| | - Veronica Mocanu
- Department of Morpho-Functional Sciences II (Pathophysiology), Center for Obesity BioBehavioral Experimental Research, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Luminita Labusca
- Department of Orthopedics and Traumatology, “Sf. Spiridon” Emergency Clinical Hospital, 700111 Iasi, Romania;
- National Institute of Research and Development in Technical Physics Iasi, 700050 Iasi, Romania
| |
Collapse
|
4
|
Duan Z, Yang Y, Qin M, Yi X. Interleukin 15: A new intermediary in the effects of exercise and training on skeletal muscle and bone function. J Cell Mol Med 2024; 28:e70136. [PMID: 39601091 PMCID: PMC11599876 DOI: 10.1111/jcmm.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 11/29/2024] Open
Abstract
Interleukin-15 (IL-15), a pro-inflammatory cytokine, is produced mainly by skeletal muscle cells, macrophages and epithelial cells. Recent research has demonstrated that IL-15 is closely related to the functions of bone and skeletal muscle in the locomotor system. There is growing evidence that exercise, an important means to regulate the immune and locomotor systems, influences IL-15 content in various tissues, thereby indirectly affecting the function of bones and muscles. Furthermore, the form, intensity, and duration of exercise determine the degree of change in IL-15 and downstream effects. This paper reviews the structure, synthesis and secretion of IL-15, the role of IL-15 in regulating the metabolism of bone tissue cells and myofibers through binding to the IL-15 receptor-α (IL-15Rα), and the response of IL-15 to different types of exercise. This review provides a reference for further analyses of the role and mechanism of action of IL-15 in the regulation of metabolism during exercise.
Collapse
Affiliation(s)
- Ziqiang Duan
- School of Sports HealthShenyang Sport UniversityShenyangChina
| | - Yang Yang
- School of KinesiologyShanghai University of SportShanghaiChina
| | - Mianhong Qin
- School of Sports HealthShenyang Sport UniversityShenyangChina
| | - Xuejie Yi
- Social Science Research CenterShenyang Sport UniversityShenyangChina
| |
Collapse
|
5
|
Das S, Preethi B, Kushwaha S, Shrivastava R. Therapeutic strategies to modulate gut microbial health: Approaches for sarcopenia management. Histol Histopathol 2024; 39:1395-1425. [PMID: 38497338 DOI: 10.14670/hh-18-730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Sarcopenia is a progressive and generalized loss of skeletal muscle and functions associated with ageing with currently no definitive treatment. Alterations in gut microbial composition have emerged as a significant contributor to the pathophysiology of multiple diseases. Recently, its association with muscle health has pointed to its potential role in mediating sarcopenia. The current review focuses on the association of gut microbiota and mediators of muscle health, connecting the dots between the influence of gut microbiota and their metabolites on biomarkers of sarcopenia. It further delineates the mechanism by which the gut microbiota affects muscle health with progressing age, aiding the formulation of a multi-modal treatment plan involving nutritional supplements and pharmacological interventions along with lifestyle changes compiled in the review. Nutritional supplements containing proteins, vitamin D, omega-3 fatty acids, creatine, curcumin, kefir, and ursolic acid positively impact the gut microbiome. Dietary fibres foster a conducive environment for the growth of beneficial microbes such as Bifidobacterium, Faecalibacterium, Ruminococcus, and Lactobacillus. Probiotics and prebiotics act by protecting against reactive oxygen species (ROS) and inflammatory cytokines. They also increase the production of gut microbiota metabolites like short-chain fatty acids (SCFAs), which aid in improving muscle health. Foods rich in polyphenols are anti-inflammatory and have an antioxidant effect, contributing to a healthier gut. Pharmacological interventions like faecal microbiota transplantation (FMT), non-steroidal anti-inflammatory drugs (NSAIDs), ghrelin mimetics, angiotensin-converting enzyme inhibitors (ACEIs), and butyrate precursors lead to the production of anti-inflammatory fatty acids and regulate appetite, gut motility, and microbial impact on gut health. Further research is warranted to deepen our understanding of the interaction between gut microbiota and muscle health for developing therapeutic strategies for ameliorating sarcopenic muscle loss.
Collapse
Affiliation(s)
- Shreya Das
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, India
| | - B Preethi
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, India
| | - Sapana Kushwaha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, India.
| | - Richa Shrivastava
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, India.
| |
Collapse
|
6
|
Farley MJ, Boytar AN, Adlard KN, Salisbury CE, Hart NH, Schaumberg MA, Jenkins DG, Skinner TL. Interleukin-15 and high-intensity exercise: relationship with inflammation, body composition and fitness in cancer survivors. J Physiol 2024; 602:5203-5215. [PMID: 39303144 DOI: 10.1113/jp286043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
Pre-clinical murine and in vitro models have demonstrated that exercise suppresses tumour and cancer cell growth. These anti-oncogenic effects of exercise were associated with the exercise-mediated release of myokines such as interleukin (IL)-15. However, no study has quantified the acute IL-15 response in human cancer survivors, and whether physiological adaptations to exercise training (i.e. body composition and cardiorespiratory fitness) influence this response. In the present study breast, prostate and colorectal cancer survivors (n = 14) completed a single bout of high-intensity interval exercise (HIIE) [4×4 min at 85-95% heart rate (HR) peak, 3 min at 50-70% HR peak] before and after 7 months of three times weekly high-intensity interval training (HIIT) on a cycle ergometer. At each time point venous blood was sampled before and immediately after HIIE to assess the acute myokine (IL-15, IL-6, IL-10, IL-1ra) responses. Markers of inflammation, cardiorespiratory fitness and measures of body composition were obtained at baseline and 7 months. An acute bout of HIIE resulted in a significant increase in IL-15 concentrations (pre-intervention: 113%; P = 0.013, post-intervention: 102%; P = 0.005). Post-exercise IL-15 concentrations were associated with all other post-exercise myokine concentrations, lean mass (P = 0.031), visceral adipose tissue (P = 0.039) and absoluteV ̇ O 2 ${{\dot{V}}_{{{{\mathrm{O}}}_{\mathrm{2}}}}}$ peak (P = 0.032). There was no significant effect of 7 months of HIIT on pre- or post-HIIE IL-15 concentrations (P > 0.05). This study demonstrates HIIE is a sufficient stimulus to increase circulating IL-15 and other myokines including IL-6, IL-10 and IL-1ra which may be clinically relevant in the anti-oncogenic effect of exercise and repetitive exposure to these effects may contribute to the positive relationship between exercise and cancer recurrence. KEY POINTS: Exercise has been demonstrated to reduce the risk of cancer recurrence. Pre-clinical murine and in vitro models have demonstrated that exercise suppresses tumour and cancer cell growth, mediated by exercise-induced myokines (IL-6 and IL-15). High-intensity interval exercise significantly increased myokines associated with the anti-oncogenic effect of exercise and the magnitude of response was associated with lean mass, but training did not appear to influence this response. Given IL-15 has been implicated in the anti-oncogenic effect of exercise and is being explored as an immunotherapy agent, high-intensity interval exercise may improve outcomes for people living beyond cancer through IL-15-mediated pathways. Interventions that increase lean mass may also enhance this response.
Collapse
Affiliation(s)
- Morgan J Farley
- Human Performance Research Centre, INSIGHT Research Institute, Faculty of Health, University of Technology Sydney (UTS), Sydney, New South Wales, Australia
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
- Cancer and Palliative Care Outcomes Centre, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Alexander N Boytar
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Kirsten N Adlard
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Chloe E Salisbury
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Nicolas H Hart
- Human Performance Research Centre, INSIGHT Research Institute, Faculty of Health, University of Technology Sydney (UTS), Sydney, New South Wales, Australia
- Cancer and Palliative Care Outcomes Centre, Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
- Caring Futures Institute, College of Nursing and Health Sciences, Flinders University, Adelaide, South Australia, Australia
- Exercise Medicine Research Institute, School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
- Institute for Health Research, University of Notre Dame Australia, Perth, Western Australia, Australia
| | - Mia A Schaumberg
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
- School of Health, University of the Sunshine Coast, Sunshine Coast, Queensland, Australia
- Sunshine Coast Health Institute, Sunshine Coast Hospital and Health Service, Birtinya, Queensland, Australia
- Manna Institute, University of New England, Armidale, New South Wales, Australia
| | - David G Jenkins
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
- School of Health, University of the Sunshine Coast, Sunshine Coast, Queensland, Australia
- Sunshine Coast Health Institute, Sunshine Coast Hospital and Health Service, Birtinya, Queensland, Australia
- Applied Sports Science Technology and Medicine Research Centre, Swansea University, Swansea, UK
| | - Tina L Skinner
- Human Performance Research Centre, INSIGHT Research Institute, Faculty of Health, University of Technology Sydney (UTS), Sydney, New South Wales, Australia
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
- School of Health, University of the Sunshine Coast, Sunshine Coast, Queensland, Australia
- School of Health Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
7
|
Liu J, Chen Y. Cell-cell crosstalk between fat cells and immune cells. Am J Physiol Endocrinol Metab 2024; 327:E371-E383. [PMID: 39082899 DOI: 10.1152/ajpendo.00024.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/06/2024]
Abstract
Obesity is a metabolic disorder with pandemic-like implications, lacking viable pharmaceutical treatments currently. Thermogenic adipose tissues, including brown and beige adipose tissues, play an essential role in regulating systemic energy homeostasis and have emerged as appealing therapeutic targets for the treatment of obesity and obesity-related diseases. The function of adipocytes is subject to complex regulation by a cellular network of immune signaling pathways in response to environmental signals. However, the specific regulatory roles of immune cells in thermogenesis and relevant involving mechanisms are still not well understood. Here, we concentrate on our present knowledge of the interaction between thermogenic adipocytes and immune cells and present an overview of cellular and molecular mechanisms underlying immunometabolism in adipose tissues. We discuss cytokines, especially interleukins, which originate from widely variable sources, and their impacts on the development and function of thermogenic adipocytes. Moreover, we summarize the neuroimmune regulation in heat production and expand a new mode of intercellular communication mediated by mitochondrial transfer. The crosstalk between immune cells and adipocytes achieves adipose tissue homeostasis and systemic energy balance. A deep understanding of this intricate interaction would provide evidence for improving thermogenic efficiency by remodeling the immune microenvironment. Interventions based on these factors show a high potential to prevent adverse metabolic outcomes in patients with obesity.
Collapse
Affiliation(s)
- Jiadai Liu
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Laboratory of Endocrinology and Metabolism, Ministry of Education Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong Chen
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Laboratory of Endocrinology and Metabolism, Ministry of Education Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, People's Republic of China
| |
Collapse
|
8
|
Rathor R, Suryakumar G. Myokines: A central point in managing redox homeostasis and quality of life. Biofactors 2024; 50:885-909. [PMID: 38572958 DOI: 10.1002/biof.2054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 03/15/2024] [Indexed: 04/05/2024]
Abstract
Redox homeostasis is a crucial phenomenon that is obligatory for maintaining the healthy status of cells. However, the loss of redox homeostasis may lead to numerous diseases that ultimately result in a compromised quality of life. Skeletal muscle is an endocrine organ that secretes hundreds of myokines. Myokines are peptides and cytokines produced and released by muscle fibers. Skeletal muscle secreted myokines act as a robust modulator for regulating cellular metabolism and redox homeostasis which play a prime role in managing and improving metabolic function in multiple organs. Further, the secretory myokines maintain redox homeostasis not only in muscles but also in other organs of the body via stabilizing oxidants and antioxidant levels. Myokines are also engaged in maintaining mitochondrial dynamics as mitochondria is a central point for the generation of reactive oxygen species (ROS). Ergo, myokines also act as a central player in communicating signals to other organs, including the pancreas, gut, liver, bone, adipose tissue, brain, and skin via their autocrine, paracrine, or endocrine effects. The present review provides a comprehensive overview of skeletal muscle-secreted myokines in managing redox homeostasis and quality of life. Additionally, probable strategies will be discussed that provide a solution for a better quality of life.
Collapse
Affiliation(s)
- Richa Rathor
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Ministry of Defence, Delhi, India
| | - Geetha Suryakumar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Ministry of Defence, Delhi, India
| |
Collapse
|
9
|
Walzik D, Wences Chirino TY, Zimmer P, Joisten N. Molecular insights of exercise therapy in disease prevention and treatment. Signal Transduct Target Ther 2024; 9:138. [PMID: 38806473 PMCID: PMC11133400 DOI: 10.1038/s41392-024-01841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Despite substantial evidence emphasizing the pleiotropic benefits of exercise for the prevention and treatment of various diseases, the underlying biological mechanisms have not been fully elucidated. Several exercise benefits have been attributed to signaling molecules that are released in response to exercise by different tissues such as skeletal muscle, cardiac muscle, adipose, and liver tissue. These signaling molecules, which are collectively termed exerkines, form a heterogenous group of bioactive substances, mediating inter-organ crosstalk as well as structural and functional tissue adaption. Numerous scientific endeavors have focused on identifying and characterizing new biological mediators with such properties. Additionally, some investigations have focused on the molecular targets of exerkines and the cellular signaling cascades that trigger adaption processes. A detailed understanding of the tissue-specific downstream effects of exerkines is crucial to harness the health-related benefits mediated by exercise and improve targeted exercise programs in health and disease. Herein, we review the current in vivo evidence on exerkine-induced signal transduction across multiple target tissues and highlight the preventive and therapeutic value of exerkine signaling in various diseases. By emphasizing different aspects of exerkine research, we provide a comprehensive overview of (i) the molecular underpinnings of exerkine secretion, (ii) the receptor-dependent and receptor-independent signaling cascades mediating tissue adaption, and (iii) the clinical implications of these mechanisms in disease prevention and treatment.
Collapse
Affiliation(s)
- David Walzik
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Tiffany Y Wences Chirino
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany
| | - Philipp Zimmer
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
| | - Niklas Joisten
- Division of Performance and Health (Sports Medicine), Institute for Sport and Sport Science, TU Dortmund University, 44227, Dortmund, North Rhine-Westphalia, Germany.
- Division of Exercise and Movement Science, Institute for Sport Science, University of Göttingen, 37075, Göttingen, Lower Saxony, Germany.
| |
Collapse
|
10
|
Mou K, Chan SMH, Vlahos R. Musculoskeletal crosstalk in chronic obstructive pulmonary disease and comorbidities: Emerging roles and therapeutic potentials. Pharmacol Ther 2024; 257:108635. [PMID: 38508342 DOI: 10.1016/j.pharmthera.2024.108635] [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: 11/06/2023] [Revised: 02/13/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a multifaceted respiratory disorder characterized by progressive airflow limitation and systemic implications. It has become increasingly apparent that COPD exerts its influence far beyond the respiratory system, extending its impact to various organ systems. Among these, the musculoskeletal system emerges as a central player in both the pathogenesis and management of COPD and its associated comorbidities. Muscle dysfunction and osteoporosis are prevalent musculoskeletal disorders in COPD patients, leading to a substantial decline in exercise capacity and overall health. These manifestations are influenced by systemic inflammation, oxidative stress, and hormonal imbalances, all hallmarks of COPD. Recent research has uncovered an intricate interplay between COPD and musculoskeletal comorbidities, suggesting that muscle and bone tissues may cross-communicate through the release of signalling molecules, known as "myokines" and "osteokines". We explored this dynamic relationship, with a particular focus on the role of the immune system in mediating the cross-communication between muscle and bone in COPD. Moreover, we delved into existing and emerging therapeutic strategies for managing musculoskeletal disorders in COPD. It underscores the development of personalized treatment approaches that target both the respiratory and musculoskeletal aspects of COPD, offering the promise of improved well-being and quality of life for individuals grappling with this complex condition. This comprehensive review underscores the significance of recognizing the profound impact of COPD on the musculoskeletal system and its comorbidities. By unravelling the intricate connections between these systems and exploring innovative treatment avenues, we can aspire to enhance the overall care and outcomes for COPD patients, ultimately offering hope for improved health and well-being.
Collapse
Affiliation(s)
- Kevin Mou
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Stanley M H Chan
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
11
|
Tanaka M, Sugimoto K, Akasaka H, Yoshida S, Takahashi T, Fujimoto T, Xie K, Yasunobe Y, Yamamoto K, Hirabayashi T, Nakanishi R, Fujino H, Rakugi H. Effects of interleukin-15 on autophagy regulation in the skeletal muscle of mice. Am J Physiol Endocrinol Metab 2024; 326:E326-E340. [PMID: 38294696 DOI: 10.1152/ajpendo.00311.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 02/01/2024]
Abstract
This study aimed to evaluate the role of skeletal muscle-derived interleukin (IL)-15 in the regulation of skeletal muscle autophagy using IL-15 knockout (KO) and transgenic (TG) mice. Male C57BL/6 wild-type (WT), IL-15 KO, and IL-15 TG mice were used in this study. Changes in muscle mass, forelimb grip strength, succinate dehydrogenase (SDH) activity, gene and protein expression levels of major regulators and indicators of autophagy, comprehensive gene expression, and DNA methylation in the gastrocnemius muscle were analyzed. Enrichment pathway analyses revealed that the pathology of IL-15 gene deficiency was related to the autophagosome pathway. Moreover, although IL-15 KO mice maintained gastrocnemius muscle mass, they exhibited a decrease in autophagy induction. IL-15 TG mice exhibited a decrease in gastrocnemius muscle mass and an increase in forelimb grip strength and SDH activity in skeletal muscle. In the gastrocnemius muscle, the ratio of phosphorylated adenosine monophosphate-activated protein kinase α (AMPKα) to total AMPKα and unc-51-like autophagy activating kinase 1 and Beclin1 protein expression were higher in the IL-15 TG group than in the WT group. IL-15 gene deficiency induces a decrease in autophagy induction. In contrast, IL-15 overexpression could improve muscle quality by activating autophagy induction while decreasing muscle mass. The regulation of IL-15 in autophagy in skeletal muscles may lead to the development of therapies for the autophagy-induced regulation of skeletal muscle mass and cellular quality control.NEW & NOTEWORTHY IL-15 gene deficiency can decrease autophagy induction. However, although IL-15 overexpression induced a decrease in muscle mass, it led to an improvement in muscle quality. Based on these results, understanding the role of IL-15 in regulating autophagy pathways within skeletal muscle may lead to the development of therapies for the autophagy-induced regulation of skeletal muscle mass and cellular quality control.
Collapse
Affiliation(s)
- Minoru Tanaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
- Department of Rehabilitation Science, Osaka Health Science University, Osaka, Japan
| | - Ken Sugimoto
- Department of General and Geriatric Medicine, Kawasaki Medical School, Okayama, Japan
| | - Hiroshi Akasaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shino Yoshida
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshimasa Takahashi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Taku Fujimoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keyu Xie
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yukiko Yasunobe
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takumi Hirabayashi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Ryosuke Nakanishi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Osaka Rosai Hospital, Osaka, Japan
| |
Collapse
|
12
|
Khalafi M, Maleki AH, Symonds ME, Sakhaei MH, Rosenkranz SK, Ehsanifar M, Korivi M, Liu Y. Interleukin-15 responses to acute and chronic exercise in adults: a systematic review and meta-analysis. Front Immunol 2024; 14:1288537. [PMID: 38235143 PMCID: PMC10791876 DOI: 10.3389/fimmu.2023.1288537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024] Open
Abstract
PURPOSE Interlukin-15 (IL-15) is an inflammatory cytokine that plays a vital role in immunology and obesity-associated metabolic syndrome. We performed this systematic review and meta-analysis to investigate whether exercise promotes circulating IL-15 concentrations in adults. METHODS We searched PubMed, Web of Science, and Scopus from inception to May, 2023 and identified original studies that investigated the effectiveness of acute and/or chronic exercise on serum/plasma IL-15 levels in adults. Standardized mean differences (SMD) and 95% confidence intervals (CI) were calculated using random effect models. Subgroup analyses were performed based on type of exercise, and training status, health status and body mass indexes (BMI) of participants. RESULTS Fifteen studies involving 411 participants and 12 studies involving 899 participants were included in the acute and chronic exercise analyses, respectively. Our findings showed that acute exercise increased circulating IL-15 concentrations immediately after exercise compared with baseline [SMD=0.90 (95% CI: 0.47 to 1.32), p=0.001], regardless of exercise type and participants' training status. Similarly, acute exercise was also associated with increased IL-15 concentrations even one-hour after exercise [SMD=0.50 (95% CI: 0.00 to 0.99), p=0.04]. Nevertheless, chronic exercise did not have a significant effect on IL-15 concentrations [SMD=0.40 (95% CI: -0.08 to 0.88), p=0.10]. CONCLUSION Our results confirm that acute exercise is effective in increasing the IL-15 concentrations immediately and one-hour after exercise intervention, and thereby playing a potential role in improving metabolism in adults. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=445634, identifier CRD42023445634.
Collapse
Affiliation(s)
- Mousa Khalafi
- Department of Physical Education and Sport Sciences, Faculty of Humanities, University of Kashan, Kashan, Iran
| | - Aref Habibi Maleki
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Michael E. Symonds
- Academic Unit of Population and Lifespan Sciences, Centre for Perinatal Research, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Mohammad Hossein Sakhaei
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, Guilan, Iran
| | - Sara K. Rosenkranz
- Department of Kinesiology and Nutrition Sciences, University of Nevada Las Vegas, Las Vegas, NV, United States
| | - Mahsa Ehsanifar
- Department of Exercise Physiology and Corrective Exercises, Faculty of Sport Sciences, Urmia University, Urmia, Iran
| | - Mallikarjuna Korivi
- Institute of Human Movement and Sports Engineering, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Yubo Liu
- Institute of Human Movement and Sports Engineering, Zhejiang Normal University, Jinhua, Zhejiang, China
| |
Collapse
|
13
|
Chang KV, Wu WT, Chen YH, Chen LR, Hsu WH, Lin YL, Han DS. Enhanced serum levels of tumor necrosis factor-α, interleukin-1β, and -6 in sarcopenia: alleviation through exercise and nutrition intervention. Aging (Albany NY) 2023; 15:13471-13485. [PMID: 38032288 DOI: 10.18632/aging.205254] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Limited research has been conducted on the post-intervention inflammatory status in sarcopenic patients, despite previous studies revealing elevated pro-inflammatory markers. This study aimed to investigate the potential elevation of specific pro-inflammatory cytokines in sarcopenic patients and evaluate the effects of exercise and nutritional support interventions on these cytokine levels. METHODS In this post-hoc analysis of a randomized controlled trial (RCT), 57 individuals with sarcopenia from the RCT and 57 non-sarcopenic participants from the same geriatric community cohort that did not participate in the RCT were enrolled. Grip strength and body composition measurements were recorded. Tumor necrotizing factor (TNF)-α, interleukin (IL)-1β, IL-6, and IL-15 levels were assessed at baseline for both groups and after a 12-week intervention consisting of resistive exercise and supplementation with branched-chain amino acids, calcium, and vitamin D3 in the patients with sarcopenia. RESULTS The sarcopenic group demonstrated significantly lower body weight, body mass index, grip strength, and skeletal muscle mass index. Moreover, sarcopenic patients exhibited higher levels of TNF-α (p=0.007), IL-1β (p<0.001), and IL-6 (p<0.001), while no significant difference was observed in IL-15 (p=0.345) between participants with and those without sarcopenia. Following the intervention, the sarcopenic group experienced significant improvements in grip strength and skeletal muscle mass index with a notable reduction in TNF-α (p=0.003), IL-1β (p=0.012) and IL-6 (p=0.001) levels. CONCLUSIONS Sarcopenic patients exhibit elevated levels of TNF-α, IL-1β, and IL-6, which declined after nutrition support and exercise interventions. However, further research is necessary to evaluate the long-term impact of these interventions on cytokine levels.
Collapse
Affiliation(s)
- Ke-Vin Chang
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
- Center for Regional Anesthesia and Pain Medicine, Taipei Municipal Wang-Fang Hospital, Taipei, Taiwan
| | - Wei-Ting Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| | - Yu-Hsin Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| | - Lan-Rong Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| | - Wei-Hsiang Hsu
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | - Yun-Lian Lin
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
- Department of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Der-Sheng Han
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei, Taiwan
| |
Collapse
|
14
|
Matthews I, Birnbaum A, Gromova A, Huang AW, Liu K, Liu EA, Coutinho K, McGraw M, Patterson DC, Banks MT, Nobles AC, Nguyen N, Merrihew GE, Wang L, Baeuerle E, Fernandez E, Musi N, MacCoss MJ, Miranda HC, La Spada AR, Cortes CJ. Skeletal muscle TFEB signaling promotes central nervous system function and reduces neuroinflammation during aging and neurodegenerative disease. Cell Rep 2023; 42:113436. [PMID: 37952157 PMCID: PMC10841857 DOI: 10.1016/j.celrep.2023.113436] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/12/2023] [Accepted: 10/28/2023] [Indexed: 11/14/2023] Open
Abstract
Skeletal muscle has recently arisen as a regulator of central nervous system (CNS) function and aging, secreting bioactive molecules known as myokines with metabolism-modifying functions in targeted tissues, including the CNS. Here, we report the generation of a transgenic mouse with enhanced skeletal muscle lysosomal and mitochondrial function via targeted overexpression of transcription factor E-B (TFEB). We discovered that the resulting geroprotective effects in skeletal muscle reduce neuroinflammation and the accumulation of tau-associated pathological hallmarks in a mouse model of tauopathy. Muscle-specific TFEB overexpression significantly ameliorates proteotoxicity, reduces neuroinflammation, and promotes transcriptional remodeling of the aged CNS, preserving cognition and memory in aged mice. Our results implicate the maintenance of skeletal muscle function throughout aging in direct regulation of CNS health and disease and suggest that skeletal muscle originating factors may act as therapeutic targets against age-associated neurodegenerative disorders.
Collapse
Affiliation(s)
- Ian Matthews
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Allison Birnbaum
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Anastasia Gromova
- Department of Pathology and Laboratory Medicine, UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA
| | - Amy W Huang
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Kailin Liu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Eleanor A Liu
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Kristen Coutinho
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Megan McGraw
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dalton C Patterson
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Macy T Banks
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amber C Nobles
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nhat Nguyen
- Department of Pathology and Laboratory Medicine, UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA
| | - Gennifer E Merrihew
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Lu Wang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Eric Baeuerle
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care Network, San Antonio, TX 78229, USA
| | - Elizabeth Fernandez
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care Network, San Antonio, TX 78229, USA
| | - Nicolas Musi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Helen C Miranda
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; RNA Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Albert R La Spada
- Department of Pathology and Laboratory Medicine, UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA; Department of Neurology and Department of Biological Chemistry, UCI Institute for Neurotherapeutics, University of California, Irvine, Irvine, CA 92697, USA.
| | - Constanza J Cortes
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA.
| |
Collapse
|
15
|
Hjortshoej MH, Aagaard P, Storgaard CD, Juneja H, Lundbye‐Jensen J, Magnusson SP, Couppé C. Hormonal, immune, and oxidative stress responses to blood flow-restricted exercise. Acta Physiol (Oxf) 2023; 239:e14030. [PMID: 37732509 PMCID: PMC10909497 DOI: 10.1111/apha.14030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/12/2023] [Accepted: 07/30/2023] [Indexed: 09/22/2023]
Abstract
INTRODUCTION Heavy-load free-flow resistance exercise (HL-FFRE) is a widely used training modality. Recently, low-load blood-flow restricted resistance exercise (LL-BFRRE) has gained attention in both athletic and clinical settings as an alternative when conventional HL-FFRE is contraindicated or not tolerated. LL-BFRRE has been shown to result in physiological adaptations in muscle and connective tissue that are comparable to those induced by HL-FFRE. The underlying mechanisms remain unclear; however, evidence suggests that LL-BFRRE involves elevated metabolic stress compared to conventional free-flow resistance exercise (FFRE). AIM The aim was to evaluate the initial (<10 min post-exercise), intermediate (10-20 min), and late (>30 min) hormonal, immune, and oxidative stress responses observed following acute sessions of LL-BFRRE compared to FFRE in healthy adults. METHODS A systematic literature search of randomized and non-randomized studies was conducted in PubMed, Embase, Cochrane Central, CINAHL, and SPORTDiscus. The Cochrane Risk of Bias (RoB2, ROBINS-1) and TESTEX were used to evaluate risk of bias and study quality. Data extractions were based on mean change within groups. RESULTS A total of 12525 hits were identified, of which 29 articles were included. LL-BFRRE demonstrated greater acute increases in growth hormone responses when compared to overall FFRE at intermediate (SMD 2.04; 95% CI 0.87, 3.22) and late (SMD 2.64; 95% CI 1.13, 4.16) post-exercise phases. LL-BFRRE also demonstrated greater increase in testosterone responses compared to late LL-FFRE. CONCLUSION These results indicate that LL-BFRRE can induce increased or similar hormone and immune responses compared to LL-FFRE and HL-FFRE along with attenuated oxidative stress responses compared to HL-FFRE.
Collapse
Affiliation(s)
- M. H. Hjortshoej
- Institute of Sports Medicine Copenhagen, Department of Orthopedic SurgeryCopenhagen University Hospital Bispebjerg and FrederiksbergCopenhagenDenmark
- Center for Healthy Aging, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
- Department of Physical and Occupational TherapyBispebjerg and Frederiksberg University HospitalCopenhagenDenmark
- Centre for Health and RehabilitationUniversity College AbsalonSlagelseDenmark
| | - P. Aagaard
- Department of Sports Science and Clinical BiomechanicsUniversity of Southern DenmarkOdenseDenmark
| | - C. D. Storgaard
- Institute of Sports Medicine Copenhagen, Department of Orthopedic SurgeryCopenhagen University Hospital Bispebjerg and FrederiksbergCopenhagenDenmark
- Center for Healthy Aging, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
- Department of Nutrition, Exercise and Sports, Section of Integrative PhysiologyUniversity of CopenhagenCopenhagenDenmark
| | - H. Juneja
- Centre for Health and RehabilitationUniversity College AbsalonSlagelseDenmark
| | - J. Lundbye‐Jensen
- Department of Nutrition, Exercise and Sports, Section of Integrative PhysiologyUniversity of CopenhagenCopenhagenDenmark
| | - S. P. Magnusson
- Institute of Sports Medicine Copenhagen, Department of Orthopedic SurgeryCopenhagen University Hospital Bispebjerg and FrederiksbergCopenhagenDenmark
- Center for Healthy Aging, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
- Department of Physical and Occupational TherapyBispebjerg and Frederiksberg University HospitalCopenhagenDenmark
| | - C. Couppé
- Institute of Sports Medicine Copenhagen, Department of Orthopedic SurgeryCopenhagen University Hospital Bispebjerg and FrederiksbergCopenhagenDenmark
- Center for Healthy Aging, Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
- Department of Physical and Occupational TherapyBispebjerg and Frederiksberg University HospitalCopenhagenDenmark
| |
Collapse
|
16
|
Szűcs G, Pipicz M, Szabó MR, Csont T, Török L, Csonka C. Effect of Eccentric Exercise on Metabolic Health in Diabetes and Obesity. SPORTS MEDICINE - OPEN 2023; 9:91. [PMID: 37775653 PMCID: PMC10541389 DOI: 10.1186/s40798-023-00596-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/12/2023] [Indexed: 10/01/2023]
Abstract
There is a growing body of evidence showing the importance of physical activity against civilization-induced metabolic diseases, including type 2 diabetes (T2DM) and obesity. Eccentric contraction, when skeletal muscles generate force by lengthening, is a unique type of skeletal muscle activity. Eccentric contraction may lead to better power production characteristics of the muscle because eccentric contraction requires less energy and can result in higher tension. Therefore, it is an ideal tool in the rehabilitation program of patients. However, the complex metabolic effect (i.e., fat mass reduction, increased lipid oxidation, improvement in blood lipid profile, and increased insulin sensitivity) of the eccentric contraction alone has scarcely been investigated. This paper aims to review the current literature to provide information on whether eccentric contraction can influence metabolic health and body composition in T2DM or obesity. We also discussed the potential role of myokines in mediating the effects of eccentric exercise. A better understanding of the mechanism of eccentric training and particularly their participation in the regulation of metabolic diseases may widen their possible therapeutic use and, thereby, may support the fight against the leading global risks for mortality in the world.
Collapse
Affiliation(s)
- Gergő Szűcs
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
- Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, 6720, Szeged, Hungary
| | - Márton Pipicz
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
- Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, 6720, Szeged, Hungary
| | - Márton Richárd Szabó
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
- Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, 6720, Szeged, Hungary
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
- Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, 6720, Szeged, Hungary
| | - László Török
- Department of Traumatology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, Szeged, 6720, Hungary
- Department of Sports Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Tisza Lajos krt. 107, Szeged, 6720, Hungary
| | - Csaba Csonka
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary.
- Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, 6720, Szeged, Hungary.
- Department of Sports Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Tisza Lajos krt. 107, Szeged, 6720, Hungary.
| |
Collapse
|
17
|
Lim JY, Kim E. The Role of Organokines in Obesity and Type 2 Diabetes and Their Functions as Molecular Transducers of Nutrition and Exercise. Metabolites 2023; 13:979. [PMID: 37755259 PMCID: PMC10537761 DOI: 10.3390/metabo13090979] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
Maintaining systemic homeostasis requires the coordination of different organs and tissues in the body. Our bodies rely on complex inter-organ communications to adapt to perturbations or changes in metabolic homeostasis. Consequently, the liver, muscle, and adipose tissues produce and secrete specific organokines such as hepatokines, myokines, and adipokines in response to nutritional and environmental stimuli. Emerging evidence suggests that dysregulation of the interplay of organokines between organs is associated with the pathophysiology of obesity and type 2 diabetes (T2D). Strategies aimed at remodeling organokines may be effective therapeutic interventions. Diet modification and exercise have been established as the first-line therapeutic intervention to prevent or treat metabolic diseases. This review summarizes the current knowledge on organokines secreted by the liver, muscle, and adipose tissues in obesity and T2D. Additionally, we highlighted the effects of diet/nutrition and exercise on the remodeling of organokines in obesity and T2D. Specifically, we investigated the ameliorative effects of caloric restriction, selective nutrients including ω3 PUFAs, selenium, vitamins, and metabolites of vitamins, and acute/chronic exercise on the dysregulation of organokines in obesity and T2D. Finally, this study dissected the underlying molecular mechanisms by which nutrition and exercise regulate the expression and secretion of organokines in specific tissues.
Collapse
Affiliation(s)
- Ji Ye Lim
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), 6431 Fannin St., Houston, TX 77030, USA
| | - Eunju Kim
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), 6431 Fannin St., Houston, TX 77030, USA
| |
Collapse
|
18
|
Zhang Y, Zhao Z, Huang LA, Liu Y, Yao J, Sun C, Li Y, Zhang Z, Ye Y, Yuan F, Nguyen TK, Garlapati NR, Wu A, Egranov SD, Caudle AS, Sahin AA, Lim B, Beretta L, Calin GA, Yu D, Hung MC, Curran MA, Rezvani K, Gan B, Tan Z, Han L, Lin C, Yang L. Molecular mechanisms of snoRNA-IL-15 crosstalk in adipocyte lipolysis and NK cell rejuvenation. Cell Metab 2023; 35:1457-1473.e13. [PMID: 37329887 PMCID: PMC10712687 DOI: 10.1016/j.cmet.2023.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/03/2023] [Accepted: 05/18/2023] [Indexed: 06/19/2023]
Abstract
Obesity, in which the functional importance of small nucleolar RNAs (snoRNAs) remains elusive, correlates with risk for many cancer types. Here, we identify that the serum copies of adipocyte-expressed SNORD46 correlate with body mass index (BMI), and serum SNORD46 antagonizes interleukin-15 (IL-15) signaling. Mechanically, SNORD46 binds IL-15 via G11, and G11A (a mutation that significantly enhances binding affinity) knockin drives obesity in mice. Functionally, SNORD46 blocks IL-15-induced, FER kinase-dependent phosphorylation of platelet glycoprotein 4 (CD36) and monoglyceride lipase (MGLL) in adipocytes, leading to inhibited lipolysis and browning. In natural killer (NK) cells, SNORD46 suppresses the IL-15-dependent autophagy, leading to reduced viability of obese NK. SNORD46 power inhibitors exhibit anti-obesity effects, concurring with improved viability of obese NK and anti-tumor immunity of CAR-NK cell therapy. Hence, our findings demonstrate the functional importance of snoRNAs in obesity and the utility of snoRNA power inhibitors for antagonizing obesity-associated immune resistance.
Collapse
Affiliation(s)
- Yaohua Zhang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zilong Zhao
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lisa A Huang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuan Liu
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Jun Yao
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chengcao Sun
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yajuan Li
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhao Zhang
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Youqiong Ye
- Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX 77030, USA
| | - Fei Yuan
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tina K Nguyen
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nikhil Reddy Garlapati
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andrew Wu
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sergey D Egranov
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Abigail S Caudle
- Department of Breast Surgical Oncology, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Aysegul A Sahin
- Department of Pathology, Division of Pathology and Laboratory Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bora Lim
- Oncology/Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George A Calin
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNAs, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung 406, Taiwan
| | - Michael A Curran
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Katayoun Rezvani
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Boyi Gan
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Experimental Radiation Oncology, Division of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhi Tan
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Leng Han
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA; Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Liuqing Yang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNAs, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
19
|
Greeves JP, Beck B, Nindl BC, O'Leary TJ. Current risks factors and emerging biomarkers for bone stress injuries in military personnel. J Sci Med Sport 2023:S1440-2440(23)00075-0. [PMID: 37188615 DOI: 10.1016/j.jsams.2023.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 04/01/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Bone stress injuries (BSIs) have plagued the military for over 150 years; they afflict around 5 to 10% of military recruits, more so in women, and continue to place a medical and financial burden on defence. While the tibia generally adapts to the rigours of basic military training, the putative mechanisms for bone maladaptation are still unclear. METHODS This paper provides a review of the published literature on current risk factors and emerging biomarkers for BSIs in military personnel; the potential for biochemical markers of bone metabolism to monitor the response to military training; and, the association of novel biochemical 'exerkines' with bone health. RESULTS The primary risk factor for BSI in military (and athletic) populations is too much training, too soon. Appropriate physical preparation before training will likely be most protective, but routine biomarkers will not yet identify those at risk. Nutritional interventions will support a bone anabolic response to training, but exposure to stress, sleep loss, and medication is likely harmful to bone. Monitoring physiology using wearables-ovulation, sleep and stress-offer potential to inform prevention strategies. CONCLUSIONS The risk factors for BSIs are well described, but their aetiology is very complex particularly in the multi-stressor military environment. Our understanding of the skeletal responses to military training is improving as technology advances, and potential biomarkers are constantly emerging, but sophisticated and integrated approaches to prevention of BSI are warranted.
Collapse
Affiliation(s)
- Julie P Greeves
- Army Health and Performance Research, Army HQ, Andover, United Kingdom; Norwich Medical School, University of East Anglia, United Kingdom; Division of Surgery and Interventional Science, UCL, United Kingdom.
| | - Belinda Beck
- School of Health Sciences and Social Work, Griffith University, Australia; The Bone Clinic, Australia.
| | - Bradley C Nindl
- School of Health and Rehabilitation Sciences, University of Pittsburgh, United States.
| | - Thomas J O'Leary
- Army Health and Performance Research, Army HQ, Andover, United Kingdom; Division of Surgery and Interventional Science, UCL, United Kingdom.
| |
Collapse
|
20
|
Diaz-Canestro C, Chen J, Liu Y, Han H, Wang Y, Honoré E, Lee CH, Lam KSL, Tse MA, Xu A. A machine-learning algorithm integrating baseline serum proteomic signatures predicts exercise responsiveness in overweight males with prediabetes. Cell Rep Med 2023; 4:100944. [PMID: 36787735 PMCID: PMC9975321 DOI: 10.1016/j.xcrm.2023.100944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/11/2022] [Accepted: 01/20/2023] [Indexed: 02/15/2023]
Abstract
The molecular transducers conferring the benefits of chronic exercise in diabetes prevention remain to be comprehensively investigated. Herein, serum proteomic profiling of 688 inflammatory and metabolic biomarkers in 36 medication-naive overweight and obese men with prediabetes reveals hundreds of exercise-responsive proteins modulated by 12-week high-intensity interval exercise training, including regulators of metabolism, cardiovascular system, inflammation, and apoptosis. Strong associations are found between proteins involved in gastro-intestinal mucosal immunity and metabolic outcomes. Exercise-induced changes in trefoil factor 2 (TFF2) are associated with changes in insulin resistance and fasting insulin, whereas baseline levels of the pancreatic secretory granule membrane major glycoprotein GP2 are related to changes in fasting glucose and glucose tolerance. A hybrid set of 23 proteins including TFF2 are differentially altered in exercise responders and non-responders. Furthermore, a machine-learning algorithm integrating baseline proteomic signatures accurately predicts individualized metabolic responsiveness to exercise training.
Collapse
Affiliation(s)
- Candela Diaz-Canestro
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jiarui Chen
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Hao Han
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Eric Honoré
- Université Côte d'Azur, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Institut de Pharmacologie Moléculaire et Cellulaire, Labex ICST, Valbonne, France
| | - Chi-Ho Lee
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Karen S L Lam
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Michael Andrew Tse
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Centre for Sports and Exercise, The University of Hong Kong, Hong Kong, China.
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
21
|
Wong L, McMahon LP. Crosstalk between bone and muscle in chronic kidney disease. Front Endocrinol (Lausanne) 2023; 14:1146868. [PMID: 37033253 PMCID: PMC10076741 DOI: 10.3389/fendo.2023.1146868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
With increasing life expectancy, the related disorders of bone loss, metabolic dysregulation and sarcopenia have become major health threats to the elderly. Each of these conditions is prevalent in patients with chronic kidney disease (CKD), particularly in more advanced stages. Our current understanding of the bone-muscle interaction is beyond mechanical coupling, where bone and muscle have been identified as interrelated secretory organs, and regulation of both bone and muscle metabolism occurs through osteokines and myokines via autocrine, paracrine and endocrine systems. This review appraises the current knowledge regarding biochemical crosstalk between bone and muscle, and considers recent progress related to the role of osteokines and myokines in CKD, including modulatory effects of physical exercise and potential therapeutic targets to improve musculoskeletal health in CKD patients.
Collapse
Affiliation(s)
- Limy Wong
- Department of Renal Medicine, Monash University Eastern Health Clinical School, Box Hill, VIC, Australia
- Department of Renal Medicine, Eastern Health, Box Hill, VIC, Australia
- *Correspondence: Limy Wong,
| | - Lawrence P. McMahon
- Department of Renal Medicine, Monash University Eastern Health Clinical School, Box Hill, VIC, Australia
- Department of Renal Medicine, Eastern Health, Box Hill, VIC, Australia
| |
Collapse
|
22
|
Takada S, Fumoto Y, Kinugawa S. Ergogenic effects of caffeine are mediated by myokines. Front Sports Act Living 2022; 4:969623. [PMID: 36570495 PMCID: PMC9774489 DOI: 10.3389/fspor.2022.969623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/31/2022] [Indexed: 12/13/2022] Open
Abstract
Exercise has long been known to effectively improve and enhance skeletal muscle function and performance. The favorable effects of exercise on remote organs other than skeletal muscle are well known, but the underlying mechanism has remained elusive. Recent studies have indicated that skeletal muscle not only enables body movement, but also contributes to body homeostasis and the systemic stress response via the expression and/or secretion of cytokines (so-called myokines). Not only the induction of muscle contraction itself, but also changes in intracellular calcium concentration ([Ca2+]i) have been suggested to be involved in myokine production and secretion. Caffeine is widely known as a Ca2+ ionophore, which improves skeletal muscle function and exercise performance (i.e., an "ergogenic aid"). Interestingly, some studies reported that caffeine or an increase in [Ca2+]i enhances the expression and/or secretion of myokines. In this review, we discuss the association between caffeine as an ergogenic aid and myokine regulation.
Collapse
Affiliation(s)
- Shingo Takada
- Department of Lifelong Sport, School of Sports Education, Hokusho University, Ebetsu, Japan,Department of Molecular Biology, Hokkaido University Graduate School of Medicine, Sapporo, Japan,*Correspondence: Shingo Takada ;
| | - Yoshizuki Fumoto
- Department of Molecular Biology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan,Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan,Shintaro Kinugawa
| |
Collapse
|
23
|
Barros D, Marques EA, Magalhães J, Carvalho J. Energy metabolism and frailty: The potential role of exercise-induced myokines - A narrative review. Ageing Res Rev 2022; 82:101780. [PMID: 36334911 DOI: 10.1016/j.arr.2022.101780] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022]
Abstract
Frailty is a complex condition that emerges from dysregulation in multiple physiological systems. Increasing evidence suggests the potential role of age-related energy dysregulation as a key driver of frailty. Exercise is considered the most efficacious intervention to prevent and even ameliorate frailty as it up-tunes and improves the function of several related systems. However, the mechanisms and molecules responsible for these intersystem benefits are not fully understood. The skeletal muscle is considered a secretory organ with endocrine functions that can produce and secrete exercise-related molecules such as myokines. These molecules are cytokines and other peptides released by muscle fibers in response to acute and/or chronic exercise. The available evidence supports that several myokines can elicit autocrine, paracrine, or endocrine effects, partly mediating inter-organ crosstalk and also having a critical role in improving cardiovascular, metabolic, immune, and neurological health. This review describes the current evidence about the potential link between energy metabolism dysregulation and frailty and provides a theoretical framework for the potential role of myokines (via exercise) in counteracting frailty. It also summarizes the physiological role of selected myokines and their response to different acute and chronic exercise protocols in older adults.
Collapse
Affiliation(s)
- Duarte Barros
- The Research Centre in Physical Activity, Health and Leisure, CIAFEL, University of Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal.
| | - Elisa A Marques
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University of Maia (ISMAI), Portugal; School of Sport and Exercise Sciences, Loughborough University, Loughborough, UK
| | - José Magalhães
- The Research Centre in Physical Activity, Health and Leisure, CIAFEL, University of Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Joana Carvalho
- The Research Centre in Physical Activity, Health and Leisure, CIAFEL, University of Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| |
Collapse
|
24
|
Ahsan M, Garneau L, Aguer C. The bidirectional relationship between AMPK pathway activation and myokine secretion in skeletal muscle: How it affects energy metabolism. Front Physiol 2022; 13:1040809. [PMID: 36479347 PMCID: PMC9721351 DOI: 10.3389/fphys.2022.1040809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2023] Open
Abstract
Myokines are peptides and proteins secreted by skeletal muscle cells, into the interstitium, or in the blood. Their regulation may be dependent or independent of muscle contraction to induce a variety of metabolic effects. Numerous myokines have been implicated in influencing energy metabolism via AMP-activated protein kinase (AMPK) signalling. As AMPK is centrally involved in glucose and lipid metabolism, it is important to understand how myokines influence its signalling, and vice versa. Such insight will better elucidate the mechanism of metabolic regulation during exercise and at rest. This review encompasses the latest research conducted on the relationship between AMPK signalling and myokines within skeletal muscles via autocrine or paracrine signalling.
Collapse
Affiliation(s)
- Mahdi Ahsan
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Léa Garneau
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Institut du Savoir Montfort –Recherche, Ottawa, ON, Canada
| | - Céline Aguer
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- Institut du Savoir Montfort –Recherche, Ottawa, ON, Canada
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University—Campus Outaouais, Gatineau, QC, Canada
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
25
|
Gomes MM, da Silva MMR, de Araújo IM, de Paula FJA. Bone, fat, and muscle interactions in health and disease. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2022; 66:611-620. [PMID: 36382750 PMCID: PMC10118823 DOI: 10.20945/2359-3997000000550] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/14/2022] [Indexed: 11/18/2022]
Abstract
Energy metabolism is a point of integration among the various organs and tissues of the human body, not only in terms of consumption of energy substrates but also because it concentrates a wide interconnected network controlled by endocrine factors. Thus, not only do tissues consume substrates, but they also participate in modulating energy metabolism. Soft mesenchymal tissues, in particular, play a key role in this process. The recognition that high energy consumption is involved in bone remodeling has been accompanied by evidence showing that osteoblasts and osteocytes produce factors that influence, for example, insulin sensitivity and appetite. Additionally, there are significant interactions between muscle, adipose, and bone tissues to control mutual tissue trophism. Not by chance, trophic and functional changes in these tissues go hand in hand from the beginning of an individual's development until aging. Likewise, metabolic and nutritional diseases deeply affect the musculoskeletal system and adipose tissue. The present narrative review highlights the importance of the interaction of the mesenchymal tissues for bone development and maintenance and the impact on bone from diseases marked by functional and trophic disorders of adipose and muscle tissues.
Collapse
Affiliation(s)
- Mayra Macena Gomes
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, SP, Brasil
| | | | - Iana Mizumukai de Araújo
- Departamento de Medicina Interna, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo, SP, Brasil
| | | |
Collapse
|
26
|
Koseki H, Osaki M, Honda Y, Sunagawa S, Imai C, Shida T, Matsumura U, Sakamoto J, Tomonaga I, Yokoo S, Mizukami S, Okita M. Progression of microstructural deterioration in load-bearing immobilization osteopenia. PLoS One 2022; 17:e0275439. [PMID: 36331919 PMCID: PMC9635731 DOI: 10.1371/journal.pone.0275439] [Citation(s) in RCA: 3] [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: 06/20/2022] [Accepted: 09/16/2022] [Indexed: 11/06/2022] Open
Abstract
Purpose Immobilization osteopenia is a major healthcare problem in clinical and social medicine. However, the mechanisms underlying this bone pathology caused by immobilization under load-bearing conditions are not yet fully understood. This study aimed to evaluate sequential changes to the three-dimensional microstructure of bone in load-bearing immobilization osteopenia using a fixed-limb rat model. Materials and method Eight-week-old specific-pathogen-free male Wistar rats were divided into an immobilized group and a control group (n = 60 each). Hind limbs in the immobilized group were fixed using orthopedic casts with fixation periods of 1, 2, 4, 8, and 12 weeks. Feeding and weight-bearing were freely permitted. Length of the right femur was measured after each fixation period and bone microstructure was analyzed by micro-computed tomography. The architectural parameters of cortical and cancellous bone were analyzed statistically. Results Femoral length was significantly shorter in the immobilized group than in the control group after 2 weeks. Total area and marrow area were significantly lower in the immobilized group than in the control group from 1 to 12 weeks. Cortical bone area, cortical thickness, and polar moment of inertia decreased significantly after 2 weeks. Some cancellous bone parameters showed osteoporotic changes at 2 weeks after immobilization and the gap with the control group widened as the fixation period extended (P < 0.05). Conclusion The present results indicate that load-bearing immobilization triggers early deterioration of microstructure in both cortical and cancellous bone after 2 weeks.
Collapse
Affiliation(s)
- Hironobu Koseki
- Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- * E-mail:
| | - Makoto Osaki
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuichiro Honda
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shinya Sunagawa
- Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Rehabilitation, Wajinkai Hospital, Nagasaki, Japan
| | - Chieko Imai
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takayuki Shida
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Umi Matsumura
- Department of Health Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Junya Sakamoto
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Iku Tomonaga
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Seiichi Yokoo
- Department of Nursing, Fukuoka International University of Health and Welfare, Fukuoka, Japan
| | - Satoshi Mizukami
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Minoru Okita
- Department of Physical Therapy Science, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
27
|
Zamboni M, Mazzali G, Brunelli A, Saatchi T, Urbani S, Giani A, Rossi AP, Zoico E, Fantin F. The Role of Crosstalk between Adipose Cells and Myocytes in the Pathogenesis of Sarcopenic Obesity in the Elderly. Cells 2022; 11:3361. [PMID: 36359757 PMCID: PMC9655977 DOI: 10.3390/cells11213361] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/08/2022] [Accepted: 10/14/2022] [Indexed: 11/15/2023] Open
Abstract
As a result of aging, body composition changes, with a decline in muscle mass and an increase in adipose tissue (AT), which reallocates from subcutaneous to visceral depots and stores ectopically in the liver, heart and muscles. Furthermore, with aging, muscle and AT, both of which have recognized endocrine activity, become dysfunctional and contribute, in the case of positive energy balance, to the development of sarcopenic obesity (SO). SO is defined as the co-existence of excess adiposity and low muscle mass and function, and its prevalence increases with age. SO is strongly associated with greater morbidity and mortality. The pathogenesis of SO is complex and multifactorial. This review focuses mainly on the role of crosstalk between age-related dysfunctional adipose and muscle cells as one of the mechanisms leading to SO. A better understanding of this mechanisms may be useful for development of prevention strategies and treatments aimed at reducing the occurrence of SO.
Collapse
Affiliation(s)
- Mauro Zamboni
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Gloria Mazzali
- Geriatrics Division, Department of Medicine, University of Verona, 37126 Verona, Italy
| | - Anna Brunelli
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Tanaz Saatchi
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Silvia Urbani
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Anna Giani
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Andrea P. Rossi
- Geriatrics Division, Department of Medicine, AULSS2, Ospedale Ca’Foncello, 31100 Treviso, Italy
| | - Elena Zoico
- Geriatrics Division, Department of Medicine, University of Verona, 37126 Verona, Italy
| | - Francesco Fantin
- Geriatrics Division, Department of Medicine, University of Verona, 37126 Verona, Italy
| |
Collapse
|
28
|
Gao T, Liu F, Ban B, Hou Y, Li G, Jiang M, Yang Q, Zhang M. Association between the ratio of serum creatinine to cystatin C and bone mineral density in Chinese older adults patients with type 2 diabetes mellitus. Front Nutr 2022; 9:1035853. [PMID: 36337638 PMCID: PMC9634484 DOI: 10.3389/fnut.2022.1035853] [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: 09/03/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background The ratio of creatinine to cystatin C (Cre/CysC), a marker of muscle function and muscle mass, can be used to predict sarcopenia in different populations. Since sarcopenia is closely associated with osteoporosis, this study investigated the association between Cre/CysC and bone mineral density (BMD) in patients with type 2 diabetes mellitus (T2DM). Method This cross-sectional study included 391 Chinese patients with T2DM. General information, biochemical indicators, and the BMD of lumbar spine (LS), femoral neck (FN), and total hip (TH) were measured. Results Pearson correlation analysis showed that Cre/CysC was significantly positively correlated with the BMD of LS (r = 0.170, p = 0.001), FN (r = 0.178, p < 0.001), and TH (r = 0.205, p < 0.001). The results of stepwise linear regression suggested that Cre/CysC was the only biochemical predictor of the BMD at three sites (LS: β = 0.137, p = 0.01; FN: β = 0.097, p = 0.038; TH: β = 0.145, p = 0.002). Conclusion In older patients with T2DM, high Cre/CysC value is independently positively associated with BMD and hence, Cre/CysC may serve as a valuable marker of osteoporosis.
Collapse
Affiliation(s)
- Ting Gao
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Fupeng Liu
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Bo Ban
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Yue Hou
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Guangxin Li
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Mingming Jiang
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong, China
| | - Qing Yang
- Department of Nutrition, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Mei Zhang
- Department of Endocrinology, Affiliated Hospital of Jining Medical University, Jining, Shandong, China
- *Correspondence: Mei Zhang,
| |
Collapse
|
29
|
Orsatti CL, Orsatti FL, Bezerra TG, Quevedo A, Nahas EAP. Interleukin-15 are associated with insulin resistance in postmenopausal women with metabolic syndrome. Gynecol Endocrinol 2022; 38:765-770. [PMID: 35921852 DOI: 10.1080/09513590.2022.2105832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
Abstract
OBJECTIVE To determine if higher levels of circulating interleukin (IL)-15 are positively associated with improvement in insulin resistance in postmenopausal women (PW) with metabolic syndrome (MS). METHODS According to the median value of IL-15 at baseline, PW older than or equal to 45 years were divided into two groups: higher (n = 43) and lower (n = 42) IL-15. There was a 9-month follow-up period with clinical assessments at baseline and at 9 months (criteria of metabolic syndrome, body fat, and insulin resistance). Insulin resistance (IR) was calculated according to the Homeostasis Model Assessment-estimated insulin resistance (HOMA-IR). For IL-1β, IL-6, IL-10, IL-13, IL-33, IL-15, and TNF-α was determined using immunoassay Magnetic Bead Panel. RESULTS There was an interaction between the time and group only for insulin (p = .008) and HOMA-IR (p = .024). After adjusting for confounding variables (clinical and ILs), the HOMA-IR (p = .006) and insulin (p = .003) were lower in the higher-IL-15 group [HOMA-IR: 2.2 (95% CI: 1.9-2.5) and insulin: 9.1 µIU/mL (95% CI: 7.9-10.3)] when compared to the lower-IL-15 group [HOMA-IR: 3.1 (95% CI: 2.6-3.6) and insulin: 12.9 (95% CI: 11.1-14.9)] after 9 months of follow-up. CONCLUSION Higher levels of circulating IL-15 are positively associated with improvements in IR in PW with MS.
Collapse
Affiliation(s)
| | - Fabio Lera Orsatti
- Applied Physiology, Nutrition and Exercise Research Group - PhyNEr, Institute of Health Sciences, Federal University of Triangulo Mineiro - UFTM, Uberaba, Minas Gerais, Brazil
| | | | - Ana Quevedo
- Department Health Science, Oeste Paulista University - UNOESTE, Jaú, SP, Brazil
| | - Eliana Aguiar Petri Nahas
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| |
Collapse
|
30
|
Renzini A, D’Onghia M, Coletti D, Moresi V. Histone Deacetylases as Modulators of the Crosstalk Between Skeletal Muscle and Other Organs. Front Physiol 2022; 13:706003. [PMID: 35250605 PMCID: PMC8895239 DOI: 10.3389/fphys.2022.706003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle plays a major role in controlling body mass and metabolism: it is the most abundant tissue of the body and a major source of humoral factors; in addition, it is primarily responsible for glucose uptake and storage, as well as for protein metabolism. Muscle acts as a metabolic hub, in a crosstalk with other organs and tissues, such as the liver, the brain, and fat tissue. Cytokines, adipokines, and myokines are pivotal mediators of such crosstalk. Many of these circulating factors modulate histone deacetylase (HDAC) expression and/or activity. HDACs form a numerous family of enzymes, divided into four classes based on their homology to their orthologs in yeast. Eleven family members are considered classic HDACs, with a highly conserved deacetylase domain, and fall into Classes I, II, and IV, while class III members are named Sirtuins and are structurally and mechanistically distinct from the members of the other classes. HDACs are key regulators of skeletal muscle metabolism, both in physiological conditions and following metabolic stress, participating in the highly dynamic adaptative responses of the muscle to external stimuli. In turn, HDAC expression and activity are closely regulated by the metabolic demands of the skeletal muscle. For instance, NAD+ levels link Class III (Sirtuin) enzymatic activity to the energy status of the cell, and starvation or exercise affect Class II HDAC stability and intracellular localization. SUMOylation or phosphorylation of Class II HDACs are modulated by circulating factors, thus establishing a bidirectional link between HDAC activity and endocrine, paracrine, and autocrine factors. Indeed, besides being targets of adipo-myokines, HDACs affect the synthesis of myokines by skeletal muscle, altering the composition of the humoral milieu and ultimately contributing to the muscle functioning as an endocrine organ. In this review, we discuss recent findings on the interplay between HDACs and circulating factors, in relation to skeletal muscle metabolism and its adaptative response to energy demand. We believe that enhancing knowledge on the specific functions of HDACs may have clinical implications leading to the use of improved HDAC inhibitors for the treatment of metabolic syndromes or aging.
Collapse
Affiliation(s)
- Alessandra Renzini
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Marco D’Onghia
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Dario Coletti
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
- Biological Adaptation and Ageing, Institut de Biologie Paris-Seine, Sorbonne Université, Paris, France
| | - Viviana Moresi
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
- Institute of Nanotechnology (Nanotec), National Research Council, Rome, Italy
| |
Collapse
|
31
|
Alizadeh Pahlavani H. Exercise Therapy for People With Sarcopenic Obesity: Myokines and Adipokines as Effective Actors. Front Endocrinol (Lausanne) 2022; 13:811751. [PMID: 35250869 PMCID: PMC8892203 DOI: 10.3389/fendo.2022.811751] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
Sarcopenic obesity is defined as a multifactorial disease in aging with decreased body muscle, decreased muscle strength, decreased independence, increased fat mass, due to decreased physical activity, changes in adipokines and myokines, and decreased satellite cells. People with sarcopenic obesity cause harmful changes in myokines and adipokines. These changes are due to a decrease interleukin-10 (IL-10), interleukin-15 (IL-15), insulin-like growth factor hormone (IGF-1), irisin, leukemia inhibitory factor (LIF), fibroblast growth factor-21 (FGF-21), adiponectin, and apelin. While factors such as myostatin, leptin, interleukin-6 (IL-6), interleukin-8 (IL-8), and resistin increase. The consequences of these changes are an increase in inflammatory factors, increased degradation of muscle proteins, increased fat mass, and decreased muscle tissue, which exacerbates sarcopenia obesity. In contrast, exercise, especially strength training, reverses this process, which includes increasing muscle protein synthesis, increasing myogenesis, increasing mitochondrial biogenesis, increasing brown fat, reducing white fat, reducing inflammatory factors, and reducing muscle atrophy. Since some people with chronic diseases are not able to do high-intensity strength training, exercises with blood flow restriction (BFR) are newly recommended. Numerous studies have shown that low-intensity BFR training produces the same increase in hypertrophy and muscle strength such as high-intensity strength training. Therefore, it seems that exercise interventions with BFR can be an effective way to prevent the exacerbation of sarcopenia obesity. However, due to limited studies on adipokines and exercises with BFR in people with sarcopenic obesity, more research is needed.
Collapse
|
32
|
Sabaratnam R, Wojtaszewski JFP, Højlund K. Factors mediating exercise-induced organ crosstalk. Acta Physiol (Oxf) 2022; 234:e13766. [PMID: 34981891 DOI: 10.1111/apha.13766] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 10/11/2021] [Accepted: 01/01/2022] [Indexed: 12/21/2022]
Abstract
Exercise activates a plethora of metabolic and signalling pathways in skeletal muscle and other organs causing numerous systemic beneficial metabolic effects. Thus, regular exercise may ameliorate and prevent the development of several chronic metabolic diseases. Skeletal muscle is recognized as an important endocrine organ regulating systemic adaptations to exercise. Skeletal muscle may mediate crosstalk with other organs through the release of exercise-induced cytokines, peptides and proteins, termed myokines, into the circulation. Importantly, other tissues such as the liver and adipose tissue may also release cytokines and peptides in response to exercise. Hence, exercise-released molecules are collectively called exerkines. Moreover, extracellular vesicles (EVs), in the form of exosomes or microvesicles, may carry some of the signals involved in tissue crosstalk. This review focuses on the role of factors potentially mediating crosstalk between muscle and other tissues in response to exercise.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Steno Diabetes Center Odense Odense University Hospital Odense C Denmark
- Section of Molecular Diabetes & Metabolism, Department of Clinical Research & Department of Molecular Medicine University of Southern Denmark Odense C Denmark
| | - Jørgen F. P. Wojtaszewski
- Section of Molecular Physiology Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense Odense University Hospital Odense C Denmark
- Section of Molecular Diabetes & Metabolism, Department of Clinical Research & Department of Molecular Medicine University of Southern Denmark Odense C Denmark
| |
Collapse
|
33
|
Yoshida S, Fujimoto T, Takahashi T, Sugimoto K, Akasaka H, Tanaka M, Huang Y, Yasunobe Y, Xie K, Ohnishi Y, Minami T, Takami Y, Yamamoto K, Rakugi H. IL-15RA regulates IL-15 localization and protein expression in skeletal muscle cells. Exp Physiol 2022; 107:222-232. [PMID: 35100657 DOI: 10.1113/ep090205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/24/2022] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? How are the dynamics of IL-15 and its receptors altered during the differentiation of myoblasts into myotubes, and how is IL-15 regulated? What is the main finding and its importance? ABSTRACT Interleukin-15 (IL-15) is a myokine in the Interleukin-2 (IL-2) family that is generated in the skeletal muscle during exercise. The functional effect of IL-15 involves muscle regeneration and metabolic regulation in skeletal muscle. Reports have indicated that the mechanism of Interleukin-15 receptor subunit alpha (IL-15RA) regulates IL-15 localization in immune cells. However, the dynamic of IL-15 and its receptors, which regulate the IL-15 pathway in skeletal muscle differentiation, have not yet been clarified. This study investigated the mechanism of IL-15 regulation using a mouse skeletal muscle cell line, C2C12 cells. We found that the mRNA expression of IL-15, Interleukin 2 Receptor Subunit Beta (IL-2RB) (CD122), and Interleukin 2 Receptor Subunit Gamma (IL-2RG) (CD132) increased, but that IL-15RA exhibits different kinetics as differentiation progresses. We also found that IL-15, mainly localized in the cytosol, preassembled with IL-15RA in the cytosol and fused to the plasma membrane. Moreover, IL-15RA increased IL-15 protein levels. Our findings suggest that genes comprising the IL-15 signaling complex are enhanced with the differentiation of myotubes and that IL-15RA regulates the protein kinetics of IL-15 signaling in skeletal muscle. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shino Yoshida
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Taku Fujimoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.,Institute for Biogenesis Research, Department of Anatomy Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96813, USA
| | - Toshimasa Takahashi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Ken Sugimoto
- Department of General and Geriatric Medicine, Kawasaki Medical University, Okayama, 700-8505, Japan
| | - Hiroshi Akasaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Minoru Tanaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.,Department of Rehabilitation Science, Graduate School of Health Sciences, Kobe University, Kobe, Hyogo, 654-0142, Japan.,Department of Rehabilitation Science, Osaka Health Science University, Osaka, 530-0043, Japan
| | - Yibin Huang
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yukiko Yasunobe
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Keyu Xie
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yuri Ohnishi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Tomohiro Minami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yoichi Takami
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
34
|
Bilski J, Pierzchalski P, Szczepanik M, Bonior J, Zoladz JA. Multifactorial Mechanism of Sarcopenia and Sarcopenic Obesity. Role of Physical Exercise, Microbiota and Myokines. Cells 2022; 11:cells11010160. [PMID: 35011721 PMCID: PMC8750433 DOI: 10.3390/cells11010160] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/27/2021] [Accepted: 12/31/2021] [Indexed: 02/07/2023] Open
Abstract
Obesity and ageing place a tremendous strain on the global healthcare system. Age-related sarcopenia is characterized by decreased muscular strength, decreased muscle quantity, quality, and decreased functional performance. Sarcopenic obesity (SO) is a condition that combines sarcopenia and obesity and has a substantial influence on the older adults’ health. Because of the complicated pathophysiology, there are disagreements and challenges in identifying and diagnosing SO. Recently, it has become clear that dysbiosis may play a role in the onset and progression of sarcopenia and SO. Skeletal muscle secretes myokines during contraction, which play an important role in controlling muscle growth, function, and metabolic balance. Myokine dysfunction can cause and aggravate obesity, sarcopenia, and SO. The only ways to prevent and slow the progression of sarcopenia, particularly sarcopenic obesity, are physical activity and correct nutritional support. While exercise cannot completely prevent sarcopenia and age-related loss in muscular function, it can certainly delay development and slow down the rate of sarcopenia. The purpose of this review was to discuss potential pathways to muscle deterioration in obese individuals. We also want to present the current understanding of the role of various factors, including microbiota and myokines, in the process of sarcopenia and SO.
Collapse
Affiliation(s)
- Jan Bilski
- Department of Biomechanics and Kinesiology, Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-008 Krakow, Poland
- Correspondence: ; Tel.: +48-12-421-93-51
| | - Piotr Pierzchalski
- Department of Medical Physiology, Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-126 Krakow, Poland; (P.P.); (J.B.)
| | - Marian Szczepanik
- Department of Medical Biology, Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-034 Krakow, Poland;
| | - Joanna Bonior
- Department of Medical Physiology, Chair of Biomedical Sciences, Faculty of Health Sciences, Institute of Physiotherapy, Jagiellonian University Medical College, 31-126 Krakow, Poland; (P.P.); (J.B.)
| | - Jerzy A. Zoladz
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, 31-066 Krakow, Poland;
| |
Collapse
|
35
|
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
|
36
|
Kim JS, Galvão DA, Newton RU, Gray E, Taaffe DR. Exercise-induced myokines and their effect on prostate cancer. Nat Rev Urol 2021; 18:519-542. [PMID: 34158658 DOI: 10.1038/s41585-021-00476-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
Exercise is recognized by clinicians in the field of clinical oncology for its potential role in reducing the risk of certain cancers and in reducing the risk of disease recurrence and progression; yet, the underlying mechanisms behind this reduction in risk are not fully understood. Studies applying post-exercise blood serum directly to various types of cancer cell lines provide insight that exercise might have a role in inhibiting cancer growth via altered soluble and cell-free blood contents. Myokines, which are cytokines produced by muscle and secreted into the bloodstream, might offer multiple benefits to cellular metabolism (such as a reduction in insulin resistance, improved glucose uptake and reduced adiposity), and blood myokine levels can be altered with exercise. Alterations in the levels of myokines such as IL-6, IL-15, IL-10, irisin, secreted protein acidic risk in cysteine (SPARC), myostatin, oncostatin M and decorin might exert a direct inhibitory effect on cancer growth via inhibiting proliferation, promoting apoptosis, inducing cell-cycle arrest and inhibiting the epithermal transition to mesenchymal cells. The association of insulin resistance, hyperinsulinaemia and hyperlipidaemia with obesity can create a tumour-favourable environment; exercise-induced myokines can manipulate this environment by regulating adipose tissue and adipocytes. Exercise-induced myokines also have a critical role in increasing cytotoxicity and the infiltration of immune cells into the tumour.
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
| | - 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.
| | - 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
| | - Elin Gray
- 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
| |
Collapse
|
37
|
Zumbaugh MD, Yen CN, Bodmer JS, Shi H, Gerrard DE. Skeletal Muscle O-GlcNAc Transferase Action on Global Metabolism Is Partially Mediated Through Interleukin-15. Front Physiol 2021; 12:682052. [PMID: 34326778 PMCID: PMC8313823 DOI: 10.3389/fphys.2021.682052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/24/2021] [Indexed: 01/11/2023] Open
Abstract
Besides its roles in locomotion and thermogenesis, skeletal muscle plays a significant role in global glucose metabolism and insulin sensitivity through complex nutrient sensing networks. Our previous work showed that the muscle-specific ablation of O-GlcNAc transferase (OGT) led to a lean phenotype through enhanced interleukin-15 (IL-15) expression. We also showed OGT epigenetically modified and repressed the Il15 promoter. However, whether there is a causal relationship between OGT ablation-induced IL-15 secretion and the lean phenotype remains unknown. To address this question, we generated muscle specific OGT and interleukin-15 receptor alpha subunit (IL-15rα) double knockout mice (mDKO). Deletion of IL-15rα in skeletal muscle impaired IL-15 secretion. When fed with a high-fat diet, mDKO mice were no longer protected against HFD-induced obesity compared to wild-type mice. After 22 weeks of HFD feeding, mDKO mice had an intermediate body weight and glucose sensitivity compared to wild-type and OGT knockout mice. Taken together, these data suggest that OGT action is partially mediated by muscle IL-15 production and provides some clarity into how disrupting the O-GlcNAc nutrient signaling pathway leads to a lean phenotype. Further, our work suggests that interfering with the OGT-IL15 nutrient sensing axis may provide a new avenue for combating obesity and metabolic disorders.
Collapse
Affiliation(s)
- Morgan D Zumbaugh
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Con-Ning Yen
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jocelyn S Bodmer
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Hao Shi
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - David E Gerrard
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
38
|
Messad F, Louveau I, Renaudeau D, Gilbert H, Gondret F. Analysis of merged whole blood transcriptomic datasets to identify circulating molecular biomarkers of feed efficiency in growing pigs. BMC Genomics 2021; 22:501. [PMID: 34217223 PMCID: PMC8254903 DOI: 10.1186/s12864-021-07843-4] [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: 11/24/2020] [Accepted: 06/24/2021] [Indexed: 11/10/2022] Open
Abstract
Background Improving feed efficiency (FE) is an important goal due to its economic and environmental significance for farm animal production. The FE phenotype is complex and based on the measurements of the individual feed consumption and average daily gain during a test period, which is costly and time-consuming. The identification of reliable predictors of FE is a strategy to reduce phenotyping efforts. Results Gene expression data of the whole blood from three independent experiments were combined and analyzed by machine learning algorithms to propose molecular biomarkers of FE traits in growing pigs. These datasets included Large White pigs from two lines divergently selected for residual feed intake (RFI), a measure of net FE, and in which individual feed conversion ratio (FCR) and blood microarray data were available. Merging the three datasets allowed considering FCR values (Mean = 2.85; Min = 1.92; Max = 5.00) for a total of n = 148 pigs, with a large range of body weight (15 to 115 kg) and different test period duration (2 to 9 weeks). Random forest (RF) and gradient tree boosting (GTB) were applied on the whole blood transcripts (26,687 annotated molecular probes) to identify the most important variables for binary classification on RFI groups and a quantitative prediction of FCR, respectively. The dataset was split into learning (n = 74) and validation sets (n = 74). With iterative steps for variable selection, about three hundred’s (328 to 391) molecular probes participating in various biological pathways, were identified as important predictors of RFI or FCR. With the GTB algorithm, simpler models were proposed combining 34 expressed unique genes to classify pigs into RFI groups (100% of success), and 25 expressed unique genes to predict FCR values (R2 = 0.80, RMSE = 8%). The accuracy performance of RF models was slightly lower in classification and markedly lower in regression. Conclusion From small subsets of genes expressed in the whole blood, it is possible to predict the binary class and the individual value of feed efficiency. These predictive models offer good perspectives to identify animals with higher feed efficiency in precision farming applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07843-4.
Collapse
Affiliation(s)
- Farouk Messad
- PEGASE, INRAE, Institut Agro, 35590, Saint-Gilles, France
| | | | | | - Hélène Gilbert
- GenPhySE, INRAE, INP-ENVT, 31326, Castanet Tolosan, France
| | | |
Collapse
|
39
|
Kornsuthisopon C, Manokawinchoke J, Sonpoung O, Osathanon T, Damrongsri D. Interleukin 15 participates in Jagged1-induced mineralization in human dental pulp cells. Arch Oral Biol 2021; 128:105163. [PMID: 34058721 DOI: 10.1016/j.archoralbio.2021.105163] [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: 01/26/2021] [Revised: 04/25/2021] [Accepted: 05/19/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Crosstalk between Notch and other cell signaling molecules has been implicated to regulate the osteogenic differentiation. Understanding the interaction between Notch and IL15 is essential to reveal molecular mechanism. Thus, the objective of the present study was to investigate whether IL15 participates in the Notch signaling-induced mineral deposition in human dental pulp cells (hDPs). METHODS hDPs were explanted from dental pulp tissues. To activate Notch signaling, the cells were seeded on Jagged1-immobilized surfaces. The mRNA expression was evaluated using real-time polymerase chain reaction. hDPs were treated with 5-50 ng/mL IL15. Cell viability and proliferation were determined using an MTT assay. Mineral deposition was examined using alizarin red s and Von Kossa staining. In some experiments, the cells were pretreated with a JAK inhibitor prior to stimulation. RESULTS Jagged1 induced IL15 and IL15RA expression in hDPs. IL15 treatment significantly increased mineral deposition at 14 d and upregulated ALP, OCN, OSX, ANKH, and ENPP1 mRNA expression. IL15-induced mineralization was attenuated by JAK inhibitor pretreatment. Further, JAK inhibitor pretreatment inhibited the effect of Jagged1 on hDP mineral deposition. CONCLUSION IL15 promoted the osteogenic differentiation in hDPs. Moreover, IL15 participated in the Jagged1-induced mineralization in hDPs.
Collapse
Affiliation(s)
- Chatvadee Kornsuthisopon
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Jeeranan Manokawinchoke
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Opor Sonpoung
- Oral Biology Research Center, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Damrong Damrongsri
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
40
|
Cao Z, Zhao K, Jose I, Hoogenraad NJ, Osellame LD. Biomarkers for Cancer Cachexia: A Mini Review. Int J Mol Sci 2021; 22:4501. [PMID: 33925872 PMCID: PMC8123431 DOI: 10.3390/ijms22094501] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
Cancer cachexia is a common condition in many cancer patients, particularly those with advanced disease. Cancer cachexia patients are generally less tolerant to chemotherapies and radiotherapies, largely limiting their treatment options. While the search for treatments of this condition are ongoing, standards for the efficacy of treatments have yet to be developed. Current diagnostic criteria for cancer cachexia are primarily based on loss of body mass and muscle function. However, these criteria are rather limiting, and in time, when weight loss is noticeable, it may be too late for treatment. Consequently, biomarkers for cancer cachexia would be valuable adjuncts to current diagnostic criteria, and for assessing potential treatments. Using high throughput methods such as "omics approaches", a plethora of potential biomarkers have been identified. This article reviews and summarizes current studies of biomarkers for cancer cachexia.
Collapse
Affiliation(s)
- Zhipeng Cao
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
| | - Kening Zhao
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Irvin Jose
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
| | - Nick J. Hoogenraad
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| | - Laura D. Osellame
- Department of Biochemistry and Genetics, La Trobe University, Bundoora, VIC 3086, Australia; (K.Z.); (I.J.); (N.J.H.)
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, School of Cancer Medicine, La Trobe University, Melbourne, VIC 3084, Australia
| |
Collapse
|
41
|
Atakan MM, Koşar ŞN, Güzel Y, Tin HT, Yan X. The Role of Exercise, Diet, and Cytokines in Preventing Obesity and Improving Adipose Tissue. Nutrients 2021; 13:nu13051459. [PMID: 33922998 PMCID: PMC8145589 DOI: 10.3390/nu13051459] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity continues to rise worldwide despite evidence-based public health recommendations. The promise to adopt a healthy lifestyle is increasingly important for tackling this global epidemic. Calorie restriction or regular exercise or a combination of the two is accepted as an effective strategy in preventing or treating obesity. Furthermore, the benefits conferred by regular exercise to overcome obesity are attributed not only to reduced adiposity or reduced levels of circulating lipids but also to the proteins, peptides, enzymes, and metabolites that are released from contracting skeletal muscle or other organs. The secretion of these molecules called cytokines in response to exercise induces browning of white adipose tissue by increasing the expression of brown adipocyte-specific genes within the white adipose tissue, suggesting that exercise-induced cytokines may play a significant role in preventing obesity. In this review, we present research-based evidence supporting the effects of exercise and various diet interventions on preventing obesity and adipose tissue health. We also discuss the interplay between adipose tissue and the cytokines secreted from skeletal muscle and other organs that are known to affect adipose tissue and metabolism.
Collapse
Affiliation(s)
- Muhammed Mustafa Atakan
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey; (M.M.A.); (Ş.N.K.); (Y.G.)
| | - Şükran Nazan Koşar
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey; (M.M.A.); (Ş.N.K.); (Y.G.)
| | - Yasemin Güzel
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey; (M.M.A.); (Ş.N.K.); (Y.G.)
| | - Hiu Tung Tin
- Institute for Health and Sport (iHeS), Victoria University, P.O. Box 14428, Melbourne 8001, Australia;
| | - Xu Yan
- Institute for Health and Sport (iHeS), Victoria University, P.O. Box 14428, Melbourne 8001, Australia;
- Sarcopenia Research Program, Australia Institute for Musculoskeletal Sciences (AIMSS), Melbourne 3021, Australia
- Correspondence: ; Tel.: +61-3-9919-4024; Fax: +61-3-9919-5615
| |
Collapse
|
42
|
Leal DV, Ferreira A, Watson EL, Wilund KR, Viana JL. Muscle-Bone Crosstalk in Chronic Kidney Disease: The Potential Modulatory Effects of Exercise. Calcif Tissue Int 2021; 108:461-475. [PMID: 33388899 DOI: 10.1007/s00223-020-00782-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) is a prevalent worldwide public burden that increasingly compromises overall health as the disease progresses. Two of the most negatively affected tissues are bone and skeletal muscle, with CKD negatively impacting their structure, function and activity, impairing the quality of life of these patients and contributing to morbidity and mortality. Whereas skeletal health in this population has conventionally been associated with bone and mineral disorders, sarcopenia has been observed to impact skeletal muscle health in CKD. Indeed, bone and muscle tissues are linked anatomically and physiologically, and together regulate functional and metabolic mechanisms. With the initial crosstalk between the skeleton and muscle proposed to explain bone formation through muscle contraction, it is now understood that this communication occurs through the interaction of myokines and osteokines, with the skeletal muscle secretome playing a pivotal role in the regulation of bone activity. Regular exercise has been reported to be beneficial to overall health. Also, the positive regulatory effect that exercise has been proposed to have on bone and muscle anatomical, functional, and metabolic activity has led to the proposal of regular physical exercise as a therapeutic strategy for muscle and bone-related disorders. The detection of bone- and muscle-derived cytokine secretion following physical exercise has strengthened the idea of a cross communication between these organs. Hence, this review presents an overview of the impact of CKD in bone and skeletal muscle, and narrates how these tissues intrinsically communicate with each other, with focus on the potential effect of exercise in the modulation of this intercommunication.
Collapse
Affiliation(s)
- Diogo V Leal
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University Institute of Maia, ISMAI, Maia, Portugal
| | - Aníbal Ferreira
- Department of Nephrology, Curry Cabral Hospital, Hospital Centre of Central Lisbon, Lisbon, Portugal
- Nova Medical School, Lisbon, Portugal
| | - Emma L Watson
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Kenneth R Wilund
- Department of Kinesiology and Community Health, University of Illinois At Urbana-Champaign, Champaign, IL, USA
| | - João L Viana
- Research Center in Sports Sciences, Health Sciences and Human Development, CIDESD, University Institute of Maia, ISMAI, Maia, Portugal.
| |
Collapse
|
43
|
Tang L, An S, Zhang Z, Fan X, Guo J, Sun L, Ta D. MSTN is a key mediator for low-intensity pulsed ultrasound preventing bone loss in hindlimb-suspended rats. Bone 2021; 143:115610. [PMID: 32829040 DOI: 10.1016/j.bone.2020.115610] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
Low-intensity pulsed ultrasound (LIPUS) has been used to accelerate bone fracture healing. However, the issue whether LIPUS is effective in preventing osteoporosis has not been clarified, and if so, what possible mechanisms might be responsible. Myostatin (MSTN) is a negative regulator of muscle growth, and its absence will trigger a positive response to bone. In this study, we examined the effects of LIPUS on bone micro-structure, mechanical properties and damage healing of hindlimb-suspended rats, and investigated whether the inhibition of MSTN plays a role in this process. The rats were randomly divided into four groups: Normal control group (NC), Hind limb suspension group (HLS), Hind limb suspension and 80 mW/cm2 LIPUS irradiation group (HLS+ 80 mW/cm2), Hind limb suspension and 30 mW/cm2 LIPUS irradiation group (HLS+ 30 mW/cm2). The HLS+ 80 mW/cm2 rats were treated with LIPUS (1 MHz, 80 mW/cm2) and the HLS+ 30 mW/cm2 rats were treated with LIPUS (1 MHz, 30 mW/cm2) on the femur for 20 min/day for 28 days. MC3T3-E1 cells were respectively cultured with the serum of wild type mouse and MSTN knockout mouse at 1% concentration for 7 days. After 28 days, LIPUS effectively prevented the destruction of bone microstructure and the decline of mechanical properties, and promoted bone defect healing in the tail-suspended rats. In addition, LIPUS effectively reduced the MSTN content in the quadriceps and serum of the tail-suspended rats, inhibited its receptor and downstream signaling molecules and activated the Wnt signaling pathway in femurs. Growth of MC-3T3-E1 cell cultured with the serum of MSTN knockout mice was superior to that with wild mice serum on day 7. These results indicate that MSTN is a key mediator in LIPUS preventing bone loss caused by hindlimb-suspension.
Collapse
Affiliation(s)
- Liang Tang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Shasha An
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Zhihao Zhang
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Xiushan Fan
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China
| | - Jianzhong Guo
- Shaanxi Key Laboratory of Ultrasonics, Shaanxi Normal University, Xi'an 710119, China
| | - Lijun Sun
- Institute of Sports Biology, Shaanxi Normal University, Xi'an 710119, China.
| | - Dean Ta
- Department of Electronic Engineering, Fudan University, Shanghai 200433, China; Human Phenome Institute, Fudan University, Shanghai 201203, China; Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention (MICCAI) of Shanghai, Shanghai 200032, China.
| |
Collapse
|
44
|
Effect of Various Exercise Regimens on Selected Exercise-Induced Cytokines in Healthy People. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18031261. [PMID: 33572495 PMCID: PMC7908590 DOI: 10.3390/ijerph18031261] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 12/21/2022]
Abstract
Different forms of physical activity—endurance, resistance or dynamic power—stimulate cytokine release from various tissues to the bloodstream. Receptors for exercise-induced cytokines are present in muscle tissue, adipose tissue, liver, brain, bones, cardiovascular system, immune system, pancreas, and skin. They have autocrine, paracrine and endocrine activities. Many of them regulate the myocyte growth and differentiation necessary for muscle hypertrophy and myogenesis. They also modify energy homeostasis, lipid, carbohydrate, and protein metabolism, regulate inflammation and exchange information (crosstalk) between remote organs. So far, interleukin 6 and irisin have been the best studied exercise-induced cytokines. However, many more can be grouped into myokines, hepatokines and adipomyokines. This review focuses on the less known exercise-induced cytokines such as myostatin, follistatin, decorin, brain-derived neurotrophic factor, fibroblast growth factor 21 and interleukin 15, and their relation to various forms of exercise, i.e., acute vs. chronic, regular training in healthy people.
Collapse
|
45
|
Pang BPS, Chan WS, Chan CB. Mitochondria Homeostasis and Oxidant/Antioxidant Balance in Skeletal Muscle-Do Myokines Play a Role? Antioxidants (Basel) 2021; 10:antiox10020179. [PMID: 33513795 PMCID: PMC7911667 DOI: 10.3390/antiox10020179] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are the cellular powerhouses that generate adenosine triphosphate (ATP) to substantiate various biochemical activities. Instead of being a static intracellular structure, they are dynamic organelles that perform constant structural and functional remodeling in response to different metabolic stresses. In situations that require a high ATP supply, new mitochondria are assembled (mitochondrial biogenesis) or formed by fusing the existing mitochondria (mitochondrial fusion) to maximize the oxidative capacity. On the other hand, nutrient overload may produce detrimental metabolites such as reactive oxidative species (ROS) that wreck the organelle, leading to the split of damaged mitochondria (mitofission) for clearance (mitophagy). These vital processes are tightly regulated by a sophisticated quality control system involving energy sensing, intracellular membrane interaction, autophagy, and proteasomal degradation to optimize the number of healthy mitochondria. The effective mitochondrial surveillance is particularly important to skeletal muscle fitness because of its large tissue mass as well as its high metabolic activities for supporting the intensive myofiber contractility. Indeed, the failure of the mitochondrial quality control system in skeletal muscle is associated with diseases such as insulin resistance, aging, and muscle wasting. While the mitochondrial dynamics in cells are believed to be intrinsically controlled by the energy content and nutrient availability, other upstream regulators such as hormonal signals from distal organs or factors generated by the muscle itself may also play a critical role. It is now clear that skeletal muscle actively participates in systemic energy homeostasis via producing hundreds of myokines. Acting either as autocrine/paracrine or circulating hormones to crosstalk with other organs, these secretory myokines regulate a large number of physiological activities including insulin sensitivity, fuel utilization, cell differentiation, and appetite behavior. In this article, we will review the mechanism of myokines in mitochondrial quality control and ROS balance, and discuss their translational potential.
Collapse
|
46
|
Yamada T, Sugiyama G, Mori Y. Masticatory muscle function affects the pathological conditions of dentofacial deformities. JAPANESE DENTAL SCIENCE REVIEW 2020; 56:56-61. [PMID: 31956379 PMCID: PMC6957801 DOI: 10.1016/j.jdsr.2019.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022] Open
Abstract
The causes of dentofacial deformities include various known syndromes, genetics, environmental and neuromuscular factors, trauma, and tumors. Above all, the functional effects of muscles are important, and deformation of the mandible is often associated with a mechanical imbalance of the masticatory muscles. With the vertical position of the face, weakness of the sling of the masseter muscle and medial pterygoid muscle causes dilatation of the mandibular angle. In patients with a deep bite, excessive function of the masticatory muscles is reported. Myosin heavy chain (MyHC) properties also affect jawbone morphology. In short-face patients, the proportion of type II fibers, which are fast muscles, is high. The proportions of muscle fiber types are genetically determined but can be altered by postnatal environmental factors. Orthognathic surgery may results in the transition of MyHC to type II (fast) fibers, but excessive stretching enhances the release of inflammatory mediators and causes a shift toward a greater proportion of slow muscle fibers. This feature can be related to postoperative relapse. Bones and muscles are in close crosstalk, and it may be possible to use biochemical approaches as well as biomechanical considerations for the treatment of jaw deformities.
Collapse
Affiliation(s)
- Tomohiro Yamada
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Japan
| | - Goro Sugiyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Japan
| | - Yoshihide Mori
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Japan
| |
Collapse
|
47
|
Dysregulated Autophagy Mediates Sarcopenic Obesity and Its Complications via AMPK and PGC1α Signaling Pathways: Potential Involvement of Gut Dysbiosis as a Pathological Link. Int J Mol Sci 2020; 21:ijms21186887. [PMID: 32961822 PMCID: PMC7555990 DOI: 10.3390/ijms21186887] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022] Open
Abstract
Sarcopenic obesity (SOB), which is closely related to being elderly as a feature of aging, is recently gaining attention because it is associated with many other age-related diseases that present as altered intercellular communication, dysregulated nutrient sensing, and mitochondrial dysfunction. Along with insulin resistance and inflammation as the core pathogenesis of SOB, autophagy has recently gained attention as a significant mechanism of muscle aging in SOB. Known as important cellular metabolic regulators, the AMP-activated protein kinase (AMPK) and the peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α) signaling pathways play an important role in autophagy, inflammation, and insulin resistance, as well as mutual communication between skeletal muscle, adipose tissue, and the liver. Furthermore, AMPK and PGC-1α signaling pathways are implicated in the gut microbiome-muscle axis. In this review, we describe the pathological link between SOB and its associated complications such as metabolic, cardiovascular, and liver disease, falls and fractures, osteoarthritis, pulmonary disease, and mental health via dysregulated autophagy controlled by AMPK and/or PGC-1α signaling pathways. Here, we propose potential treatments for SOB by modulating autophagy activity and gut dysbiosis based on plausible pathological links.
Collapse
|
48
|
Chen W, Wang L, You W, Shan T. Myokines mediate the cross talk between skeletal muscle and other organs. J Cell Physiol 2020; 236:2393-2412. [PMID: 32885426 DOI: 10.1002/jcp.30033] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Myokines are muscle-derived cytokines and chemokines that act extensively on organs and exert beneficial metabolic functions in the whole-body through specific signal networks. Myokines as mediators provide the conceptual basis for a whole new paradigm useful for understanding how skeletal muscle communicates with other organs. In this review, we summarize and discuss classes of myokines and their physiological functions in mediating the regulatory roles of skeletal muscle on other organs and the regulation of the whole-body energy metabolism. We review the mechanisms involved in the interaction between skeletal muscle and nonmuscle organs through myokines. Moreover, we clarify the connection between exercise, myokines and disease development, which may contribute to the understanding of a potential mechanism by which physical inactivity affects the process of metabolic diseases via myokines. Based on the current findings, myokines are important factors that mediate the effect of skeletal muscle on other organ functions and whole-body metabolism.
Collapse
Affiliation(s)
- Wentao Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| |
Collapse
|
49
|
β-hydroxy-β-methylbutyrate supplementation benefits the effects of resistance training on body fat reduction via increased irisin expression in white adipose tissue. Biol Sport 2020; 38:113-121. [PMID: 33795920 PMCID: PMC7996383 DOI: 10.5114/biolsport.2020.97671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/01/2020] [Accepted: 07/21/2020] [Indexed: 12/25/2022] Open
Abstract
The effects of resistance training (RT) associated with calcium β-hydroxyβ-methylbutyrate (CaHMB) supplementation on the body composition and gene expression of cytokines related to skeletal muscle hypertrophy and adipose tissue metabolism were studied in rats. Male Wistar rats were divided into four groups of 12 animals: sedentary control (SC); sedentary supplemented (SS); resistance training control (RTC) and resistance training supplemented (RTS). Rats from RTC and RTS groups were submitted to an RT programme and those from SS and RTS groups received 1 mL of CaHMB (320 mg kg-1 day-1) by gavage, for 8 weeks. We evaluated: body composition; plasma lipid profile; the gene expression of interleukin (IL)-6, IL-10, IL-15 and fibronectin type III domain-containing protein 5 (FNDC-5) in skeletal muscle, and IL-6, mitochondrial uncoupling protein 1 (UCP-1) in white adipose tissue (WAT); and the concentration of irisin in WAT. Compared to RTC alone, the combination of CaHMB with RT (RTS) further reduced abdominal circumference (5.3%), Lee index (2.4%), fat percentage (24.4%), plasma VLDL cholesterol (16.8%) and triglycerides (17%) and increased the gene expression of FNDC-5 (78.9%) and IL-6 (47.4%) in skeletal muscle and irisin concentration (26.9%) in WAT. Neither RT nor CaHMB affected the protein percentage or the gene expression of IL-6 and UCP-1 in WAT and IL-10, IL-15 in skeletal muscle. In conclusion, CaHMB supplementation increased the beneficial effects of RT on body fat reduction and was associated with muscular genic expression of IL-6 and FNDC-5 and irisin concentration in WAT, despite the lack of change in protein mass and maximal strength.
Collapse
|
50
|
Effect of treatment with conditioned media derived from C2C12 myotube on adipogenesis and lipolysis in 3T3-L1 adipocytes. PLoS One 2020; 15:e0237095. [PMID: 32756599 PMCID: PMC7406041 DOI: 10.1371/journal.pone.0237095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/20/2020] [Indexed: 01/04/2023] Open
Abstract
Regular exercise is an effective strategy that is used to prevent and treat obesity as well as type 2 diabetes. Exercise-induced myokine secretion is considered a mechanism that coordinates communication between muscles and other organs. In order to examine the possibility of novel communications from muscle to adipose tissue mediated by myokines, we treated 3T3-L1 adipocytes with C2C12 myotube electrical pulse stimulation-conditioned media (EPS-CM), using a C2C12 myotube contraction system stimulated by an electrical pulse. Continuous treatment with myotube EPS-CM promoted adipogenesis of 3T3-L1 pre-adipocytes via the upregulation of the peroxisome proliferator-activated receptor-gamma (PPARγ) 2 and PPARγ-regulated gene expression. Furthermore, our results revealed that myotube EPS-CM induces lipolysis and secretion of adiponectin in mature adipocytes. EPS-CM obtained from a C2C12 myoblast culture did not induce such changes in these genes, suggesting that contraction-induced myokine(s) secretion occurs particularly in differentiated myotubes. Thus, contraction-induced secretion of myokine(s) promotes adipogenesis and lipid metabolism in 3T3-L1 adipocytes. These findings suggest the possibility that skeletal muscle communicates to adipose tissues during exercise, probably by the intermediary of unidentified myokines.
Collapse
|