1
|
Hashemolhosseini S, Gessler L. Crosstalk among canonical Wnt and Hippo pathway members in skeletal muscle and at the neuromuscular junction. Neural Regen Res 2025; 20:2464-2479. [PMID: 39248171 PMCID: PMC11801303 DOI: 10.4103/nrr.nrr-d-24-00417] [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: 04/15/2024] [Revised: 07/04/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
Skeletal muscles are essential for locomotion, posture, and metabolic regulation. To understand physiological processes, exercise adaptation, and muscle-related disorders, it is critical to understand the molecular pathways that underlie skeletal muscle function. The process of muscle contraction, orchestrated by a complex interplay of molecular events, is at the core of skeletal muscle function. Muscle contraction is initiated by an action potential and neuromuscular transmission requiring a neuromuscular junction. Within muscle fibers, calcium ions play a critical role in mediating the interaction between actin and myosin filaments that generate force. Regulation of calcium release from the sarcoplasmic reticulum plays a key role in excitation-contraction coupling. The development and growth of skeletal muscle are regulated by a network of molecular pathways collectively known as myogenesis. Myogenic regulators coordinate the differentiation of myoblasts into mature muscle fibers. Signaling pathways regulate muscle protein synthesis and hypertrophy in response to mechanical stimuli and nutrient availability. Several muscle-related diseases, including congenital myasthenic disorders, sarcopenia, muscular dystrophies, and metabolic myopathies, are underpinned by dysregulated molecular pathways in skeletal muscle. Therapeutic interventions aimed at preserving muscle mass and function, enhancing regeneration, and improving metabolic health hold promise by targeting specific molecular pathways. Other molecular signaling pathways in skeletal muscle include the canonical Wnt signaling pathway, a critical regulator of myogenesis, muscle regeneration, and metabolic function, and the Hippo signaling pathway. In recent years, more details have been uncovered about the role of these two pathways during myogenesis and in developing and adult skeletal muscle fibers, and at the neuromuscular junction. In fact, research in the last few years now suggests that these two signaling pathways are interconnected and that they jointly control physiological and pathophysiological processes in muscle fibers. In this review, we will summarize and discuss the data on these two pathways, focusing on their concerted action next to their contribution to skeletal muscle biology. However, an in-depth discussion of the non-canonical Wnt pathway, the fibro/adipogenic precursors, or the mechanosensory aspects of these pathways is not the focus of this review.
Collapse
Affiliation(s)
- Said Hashemolhosseini
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lea Gessler
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Welc SS, Brotto M, White KE, Bonewald LF. Aging: A struggle for beneficial to overcome negative factors made by muscle and bone. Mech Ageing Dev 2025; 224:112039. [PMID: 39952614 PMCID: PMC11893237 DOI: 10.1016/j.mad.2025.112039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/15/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Musculoskeletal health is strongly influenced by regulatory interactions of bone and muscle. Recent discoveries have identified a number of key mechanisms through which soluble factors released during exercise by bone exert positive effects on muscle and by muscle on bone. Although exercise can delay the negative effects of aging, these beneficial effects are diminished with aging. The limited response of aged muscle and bone tissue to exercise are accompanied by a failure in bone and muscle communication. Here, we propose that exercise induced beneficial factors must battle changes in circulating endocrine and inflammatory factors that occur with aging. Furthermore, sedentary behavior results in the release of negative factors impacting the ability of bone and muscle to respond to physical activity especially with aging. In this review we report on exercise responsive factors and evidence of modification occurring with aging.
Collapse
Affiliation(s)
- Steven S Welc
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas-Arlington, Arlington, TX 76019, USA.
| | - Kenneth E White
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Department of Molecular and Medical Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Lynda F Bonewald
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
3
|
Hoseini R, Hoseini Z, Kamangar A. Myogenic differentiation markers in muscle tissue after aerobic training. Heliyon 2025; 11:e41888. [PMID: 39897925 PMCID: PMC11787638 DOI: 10.1016/j.heliyon.2025.e41888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 11/21/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Aerobic training induces a myriad of adaptations in muscle tissue, encompassing alterations in muscle fiber type composition, hypertrophy, and metabolic capacity. Understanding the potential role of myogenic differentiation markers (MDFs), such as Pax7, MyoD, Myogenin, and myosin heavy chain (MHC) isoforms, in mediating these adaptations is of paramount importance. The review delves into the intricate molecular mechanisms underlying the regulation of MDFs following aerobic training, elucidating the role of key signaling pathways including the MAPK/ERK, PI3K/Akt, and AMPK pathways, among others. These pathways play pivotal roles in orchestrating the expression and activity of MDFs, ultimately influencing muscle adaptation and regeneration. The comprehension of MDFs in the context of aerobic training is far-reaching, offering the potential for targeted interventions to optimize muscle adaptation and regeneration. This review identifies the need for further research to unveil the precise molecular mechanisms of the activation and interaction of myogenic differentiation markers with other signaling pathways, as well as to explore their potential as therapeutic targets for muscle-related conditions. This review article also provides a thorough analysis of MDFs in muscle tissue after aerobic training, highlighting their potential clinical implications and outlining future research directions in this area.
Collapse
Affiliation(s)
- Rastegar Hoseini
- Assistant Professor of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Zahra Hoseini
- PhD of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Ayob Kamangar
- PhD Student of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
4
|
Feng S, Zhou H, Lin X, Zhu S, Chen H, Zhou H, Wang R, Wang P, Shao X, Wang J. Exercise promotes skeletal muscle growth in adolescents via modulating Mettl3-mediated m6A methylation of MyoD in muscle satellite cells. Cell Mol Biol Lett 2024; 29:150. [PMID: 39633280 PMCID: PMC11616192 DOI: 10.1186/s11658-024-00670-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Exercise exerts positive impacts on skeletal muscle health and homeostasis. Emerging evidence suggests that m6A methylation is involved in various physiological processes. However, the impact of exercise on adolescent skeletal muscle growth and the underlying epigenetic mechanisms remain poorly understood. METHODS The lower-limb skeletal muscles were harvested from exercise and control groups to compare the skeletal muscle growth in adolescents. mRNA sequencing was conducted to explore the mechanisms underlying enhanced skeletal muscle growth following exercise. The effects and mechanisms of Mettl3-mediated m6A methylation on adolescent skeletal muscle growth were investigated using muscle satellite cell (MuSC)-specific Mettl3 knockout (KO) mice. The potential function of MyoD for skeletal muscle growth in adolescents was explored by phenotypes after overexpression and evaluation of in vivo myogenesis. Additionally, the effects of the methyl donor betaine on adolescent skeletal muscle growth were investigated in vitro and in vivo. RESULTS Exercise could promote skeletal muscle growth in adolescents. Sequencing data analysis and confirmation assays uncovered that exercise significantly increased Mettl3-mediated m6A methylation and elevated the expression levels of activation marker MyoD in MuSCs. Establishment of MuSC-specific Mettl3 KO mice further demonstrated that Mettl3-mediated m6A methylation in MyoD contributed to skeletal muscle growth during adolescence. Mettl3-mediated m6A methylation regulated MyoD mRNA stability at the posttranscriptional level in MuSCs, with a functional site at 234 bp A. Increased expression of MyoD could contribute to myogenesis of adolescent MuSCs. Furthermore, the methyl donor betaine could enhance MyoD expression, contributing to MuSCs activation and skeletal muscle growth in adolescents by boosting m6A methylation levels. CONCLUSIONS Exercise promoted skeletal muscle growth in adolescents through facilitating MyoD mRNA stability of MuSCs in a Mettl3-mediated m6A-dependent manner. The methyl donor betaine could be a potential alternative to exercise for promoting adolescent skeletal muscle growth by directly augmenting the global levels of m6A methylation. These findings may provide a theoretical foundation for encouraging daily fitness exercise and ensuring healthy growth in adolescents.
Collapse
Affiliation(s)
- Shujing Feng
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Hao Zhou
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingzuan Lin
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
| | - Siyuan Zhu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Huifang Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Han Zhou
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Peng Wang
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiexiang Shao
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jianhua Wang
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
5
|
Feng L, Li B, Yong SS, Wu X, Tian Z. Exercise and nutrition benefit skeletal muscle: From influence factor and intervention strategy to molecular mechanism. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:302-314. [PMID: 39309454 PMCID: PMC11411340 DOI: 10.1016/j.smhs.2024.02.004] [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: 12/05/2023] [Revised: 01/24/2024] [Accepted: 02/18/2024] [Indexed: 09/25/2024] Open
Abstract
Sarcopenia is a progressive systemic skeletal muscle disease induced by various physiological and pathological factors, including aging, malnutrition, denervation, and cardiovascular diseases, manifesting as the decline of skeletal muscle mass and function. Both exercise and nutrition produce beneficial effects on skeletal muscle growth and are viewed as feasible strategies to prevent sarcopenia. Mechanisms involve regulating blood flow, oxidative stress, inflammation, apoptosis, protein synthesis and degradation, and satellite cell activation through exerkines and gut microbiomes. In this review, we summarized and discussed the latest progress and future development of the above mechanisms for providing a theoretical basis and ideas for the prevention and treatment of sarcopenia.
Collapse
Affiliation(s)
- Lili Feng
- College of Education, Physical Education Department, Zhejiang University, Hangzhou, 310058, China
| | - Bowen Li
- College of Education, Physical Education Department, Zhejiang University, Hangzhou, 310058, China
| | - Su Sean Yong
- College of Education, Physical Education Department, Zhejiang University, Hangzhou, 310058, China
| | - Xiaonan Wu
- The Information and Communication College, National University of Defense Technology, Xi'an, 710106, China
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, 710119, China
| |
Collapse
|
6
|
Espino-Gonzalez E, Dalbram E, Mounier R, Gondin J, Farup J, Jessen N, Treebak JT. Impaired skeletal muscle regeneration in diabetes: From cellular and molecular mechanisms to novel treatments. Cell Metab 2024; 36:1204-1236. [PMID: 38490209 DOI: 10.1016/j.cmet.2024.02.014] [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: 09/07/2023] [Revised: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Diabetes represents a major public health concern with a considerable impact on human life and healthcare expenditures. It is now well established that diabetes is characterized by a severe skeletal muscle pathology that limits functional capacity and quality of life. Increasing evidence indicates that diabetes is also one of the most prevalent disorders characterized by impaired skeletal muscle regeneration, yet underlying mechanisms and therapeutic treatments remain poorly established. In this review, we describe the cellular and molecular alterations currently known to occur during skeletal muscle regeneration in people with diabetes and animal models of diabetes, including its associated comorbidities, e.g., obesity, hyperinsulinemia, and insulin resistance. We describe the role of myogenic and non-myogenic cell types on muscle regeneration in conditions with or without diabetes. Therapies for skeletal muscle regeneration and gaps in our knowledge are also discussed, while proposing future directions for the field.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
7
|
Li P, Han X, Li J, Wang Y, Cao Y, Wu W, Liu X. Aerobic exercise training engages the canonical wnt pathway to improve pulmonary function and inflammation in COPD. BMC Pulm Med 2024; 24:236. [PMID: 38745304 PMCID: PMC11095004 DOI: 10.1186/s12890-024-03048-z] [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: 12/22/2023] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND We studied whether the exercise improves cigarette smoke (CS) induced chronic obstructive pulmonary disease (COPD) in mice through inhibition of inflammation mediated by Wnt/β-catenin-peroxisome proliferator-activated receptor (PPAR) γ signaling. METHODS Firstly, we observed the effect of exercise on pulmonary inflammation, lung function, and Wnt/β-catenin-PPARγ. A total of 30 male C57BL/6J mice were divided into the control group (CG), smoke group (SG), low-intensity exercise group (LEG), moderate-intensity exercise group (MEG), and high-intensity exercise group (HEG). All the groups, except for CG, underwent whole-body progressive exposure to CS for 25 weeks. Then, we assessed the maximal exercise capacity of mice from the LEG, MEG, and HEG, and performed an 8-week treadmill exercise intervention. Then, we used LiCl (Wnt/β-catenin agonist) and XAV939 (Wnt/β-catenin antagonist) to investigate whether Wnt/β-catenin-PPARγ pathway played a role in the improvement of COPD via exercise. Male C57BL/6J mice were randomly divided into six groups (n = 6 per group): CG, SG, LiCl group, LiCl and exercise group, XAV939 group, and XAV939 and exercise group. Mice except those in the CG were exposed to CS, and those in the exercise groups were subjected to moderate-intensity exercise training. All the mice were subjected to lung function test, lung histological assessment, and analysis of inflammatory markers in the bronchoalveolar lavage fluid, as well as detection of Wnt1, β-catenin and PPARγ proteins in the lung tissue. RESULTS Exercise of various intensities alleviated lung structural changes, pulmonary function and inflammation in COPD, with moderate-intensity exercise exhibiting significant and comprehensive effects on the alleviation of pulmonary inflammation and improvement of lung function. Low-, moderate-, and high-intensity exercise decreased β-catenin levels and increased those of PPARγ significantly, and only moderate-intensity exercise reduced the level of Wnt1 protein. Moderate-intensity exercise relieved the inflammation aggravated by Wnt agonist. Wnt antagonist combined with moderate-intensity exercise increased the levels of PPARγ, which may explain the highest improvement of pulmonary function observed in this group. CONCLUSIONS Exercise effectively decreases COPD pulmonary inflammation and improves pulmonary function. The beneficial role of exercise may be exerted through Wnt/β-catenin-PPARγ pathway.
Collapse
Affiliation(s)
- Peijun Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Xiaoyu Han
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, P.R. China
| | - Jian Li
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, P.R. China
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, P.R. China
| | - Yingqi Wang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Yuanyuan Cao
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, P.R. China
| | - Weibing Wu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, P.R. China.
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, 201203, P.R. China.
| |
Collapse
|
8
|
Stoikos J, Kurgan N, Kottaras S, Fajardo VA, Gittings W, Klentrou P. Effects of sclerostin injection on soleus and extensor digitorum longus muscle tissue in male mice. Can J Physiol Pharmacol 2024; 102:293-304. [PMID: 37976473 DOI: 10.1139/cjpp-2023-0268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Sclerostin, a potent inhibitor of the Wnt signaling pathway, plays a critical role in bone homeostasis. Evidence suggests that sclerostin may also be involved in crosstalk between other tissues, including muscle. This pilot study attempted to examine the effects of sclerostin on soleus and extensor digitorum longus (EDL) muscle tissue from male mice that were given continuous recombinant sclerostin injections for 4 weeks. A total of 48 10-week-old male C57BL/6J mice were assigned to be sedentary or perform 1 h treadmill running per day for 4 weeks and administered subcutaneous injections of either saline or recombinant sclerostin 5 days/week. Sclerostin injection led to a reduction in the soleus myosin heavy chain (MHC) I, MHC I/IIA, MHC IIA/X, and MHC IIB cross-sectional area (p < 0.05) with no exercise effects on these reductions. In contrast, there were no effects of sclerostin injections or exercise on the fast-twitch EDL muscle in terms of size, MHC protein, or markers of Wnt signaling. These findings provide preliminary evidence of sclerostin's endocrine role in muscle via decreases in myofiber cross-sectional area, which seems to be independent of fiber type but muscle type-specific. More studies, however, are needed to confirm these preliminary results.
Collapse
Affiliation(s)
- Joshua Stoikos
- Centre for Bone and Muscle Health, Department of Kinesiology, Brock University, ON, Canada
| | - Nigel Kurgan
- Centre for Bone and Muscle Health, Department of Kinesiology, Brock University, ON, Canada
| | - Steven Kottaras
- Centre for Bone and Muscle Health, Department of Kinesiology, Brock University, ON, Canada
| | - Val A Fajardo
- Centre for Bone and Muscle Health, Department of Kinesiology, Brock University, ON, Canada
| | - William Gittings
- Centre for Bone and Muscle Health, Department of Kinesiology, Brock University, ON, Canada
| | - Panagiota Klentrou
- Centre for Bone and Muscle Health, Department of Kinesiology, Brock University, ON, Canada
| |
Collapse
|
9
|
Sui H, Dou J, Shi B, Cheng X. The reciprocity of skeletal muscle and bone: an evolving view from mechanical coupling, secretory crosstalk to stem cell exchange. Front Physiol 2024; 15:1349253. [PMID: 38505709 PMCID: PMC10949226 DOI: 10.3389/fphys.2024.1349253] [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: 12/04/2023] [Accepted: 02/19/2024] [Indexed: 03/21/2024] Open
Abstract
Introduction: Muscle and bone constitute the two main parts of the musculoskeletal system and generate an intricately coordinated motion system. The crosstalk between muscle and bone has been under investigation, leading to revolutionary perspectives in recent years. Method and results: In this review, the evolving concept of muscle-bone interaction from mechanical coupling, secretory crosstalk to stem cell exchange was explained in sequence. The theory of mechanical coupling stems from the observation that the development and maintenance of bone mass are largely dependent on muscle-derived mechanical loads, which was later proved by Wolff's law, Utah paradigm and Mechanostat hypothesis. Then bone and muscle are gradually recognized as endocrine organs, which can secrete various cytokines to modulate the tissue homeostasis and remodeling to each other. The latest view presented muscle-bone interaction in a more direct way: the resident mesenchymal stromal cell in the skeletal muscle, i.e., fibro-adipogenic progenitors (FAPs), could migrate to the bone injury site and contribute to bone regeneration. Emerging evidence even reveals the ectopic source of FAPs from tissue outside the musculoskeletal system, highlighting its dynamic property. Conclusion: FAPs have been established as the critical cell connecting muscle and bone, which provides a new modality to study inter-tissue communication. A comprehensive and integrated perspective of muscle and bone will facilitate in-depth research in the musculoskeletal system and promote novel therapeutic avenues in treating musculoskeletal disorders.
Collapse
Affiliation(s)
| | | | | | - Xu Cheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Cisterna B, Malatesta M. Molecular and Structural Alterations of Skeletal Muscle Tissue Nuclei during Aging. Int J Mol Sci 2024; 25:1833. [PMID: 38339110 PMCID: PMC10855217 DOI: 10.3390/ijms25031833] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Aging is accompanied by a progressive loss of skeletal muscle mass and strength. The mechanisms underlying this phenomenon are certainly multifactorial and still remain to be fully elucidated. Changes in the cell nucleus structure and function have been considered among the possible contributing causes. This review offers an overview of the current knowledge on skeletal muscle nuclei in aging, focusing on the impairment of nuclear pathways potentially involved in age-related muscle decline. In skeletal muscle two types of cells are present: fiber cells, constituting the contractile muscle mass and containing hundreds of myonuclei, and the satellite cells, i.e., the myogenic mononuclear stem cells occurring at the periphery of the fibers and responsible for muscle growth and repair. Research conducted on different experimental models and with different methodological approaches demonstrated that both the myonuclei and satellite cell nuclei of aged skeletal muscles undergo several structural and molecular alterations, affecting chromatin organization, gene expression, and transcriptional and post-transcriptional activities. These alterations play a key role in the impairment of muscle fiber homeostasis and regeneration, thus contributing to the age-related decrease in skeletal muscle mass and function.
Collapse
Affiliation(s)
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy;
| |
Collapse
|
11
|
Ochi E, Barrington A, Wehling‐Henricks M, Avila M, Kuro‐o M, Tidball JG. Klotho regulates the myogenic response of muscle to mechanical loading and exercise. Exp Physiol 2023; 108:1531-1547. [PMID: 37864311 PMCID: PMC10841225 DOI: 10.1113/ep091263] [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: 04/14/2023] [Accepted: 08/16/2023] [Indexed: 10/22/2023]
Abstract
NEW FINDINGS What is the central question of this study? Does the hormone Klotho affect the myogenic response of muscle cells to mechanical loading or exercise? What is the main finding and its importance? Klotho prevents direct, mechanical activation of genes that regulate muscle differentiation, including genes that encode the myogenic regulatory factor myogenin and proteins in the canonical Wnt signalling pathway. Similarly, elevated levels of klotho expression in vivo prevent the exercise-induced increase in myogenin-expressing cells and reduce exercise-induced activation of the Wnt pathway. These findings demonstrate a new mechanism through which the responses of muscle to the mechanical environment are regulated. ABSTRACT Muscle growth is influenced by changes in the mechanical environment that affect the expression of genes that regulate myogenesis. We tested whether the hormone Klotho could influence the response of muscle to mechanical loading. Applying mechanical loads to myoblasts in vitro increased RNA encoding transcription factors that are expressed in activated myoblasts (Myod) and in myogenic cells that have initiated terminal differentiation (Myog). However, application of Klotho to myoblasts prevented the loading-induced activation of Myog without affecting loading-induced activation of Myod. This indicates that elevated Klotho inhibits mechanically-induced differentiation of myogenic cells. Elevated Klotho also reduced the transcription of genes encoding proteins involved in the canonical Wnt pathway or their target genes (Wnt9a, Wnt10a, Ccnd1). Because the canonical Wnt pathway promotes differentiation of myogenic cells, these findings indicate that Klotho inhibits the differentiation of myogenic cells experiencing mechanical loading. We then tested whether these effects of Klotho occurred in muscles of mice experiencing high-intensity interval training (HIIT) by comparing wild-type mice and klotho transgenic mice. The expression of a klotho transgene combined with HIIT synergized to tremendously elevate numbers of Pax7+ satellite cells and activated MyoD+ cells. However, transgene expression prevented the increase in myogenin+ cells caused by HIIT in wild-type mice. Furthermore, transgene expression diminished the HIIT-induced activation of the canonical Wnt pathway in Pax7+ satellite cells. Collectively, these findings show that Klotho inhibits loading- or exercise-induced activation of muscle differentiation and indicate a new mechanism through which the responses of muscle to the mechanical environment are regulated.
Collapse
Affiliation(s)
- Eisuke Ochi
- Faculty of Bioscience and Applied ChemistryHosei UniversityTokyoJapan
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Alice Barrington
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| | | | - Marcus Avila
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| | - Makoto Kuro‐o
- Division of Anti‐Aging MedicineCenter for Molecular MedicineJichi Medical UniversityTochigiJapan
| | - James G. Tidball
- Department of Integrative Biology and PhysiologyUniversity of CaliforniaLos AngelesCAUSA
- Molecular, Cellular & Integrative Physiology ProgramUniversity of CaliforniaLos AngelesCAUSA
- Department of BioengineeringUniversity of CaliforniaLos AngelesCAUSA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLAUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
12
|
Rahman NIA, Lam CL, Sulaiman N, Abdullah NAH, Nordin F, Ariffin SHZ, Yazid MD. PAX7, a Key for Myogenesis Modulation in Muscular Dystrophies through Multiple Signaling Pathways: A Systematic Review. Int J Mol Sci 2023; 24:13051. [PMID: 37685856 PMCID: PMC10487808 DOI: 10.3390/ijms241713051] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 09/10/2023] Open
Abstract
Muscular dystrophy is a heterogenous group of hereditary muscle disorders caused by mutations in the genes responsible for muscle development, and is generally defined by a disastrous progression of muscle wasting and massive loss in muscle regeneration. Pax7 is closely associated with myogenesis, which is governed by various signaling pathways throughout a lifetime and is frequently used as an indicator in muscle research. In this review, an extensive literature search adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines was performed to identify research that examined signaling pathways in living models, while quantifying Pax7 expression in myogenesis. A total of 247 articles were retrieved from the Web of Science (WoS), PubMed and Scopus databases and were thoroughly examined and evaluated, resulting in 19 articles which met the inclusion criteria. Admittedly, we were only able to discuss the quantification of Pax7 carried out in research affecting various type of genes and signaling pathways, rather than the expression of Pax7 itself, due to the massive differences in approach, factor molecules and signaling pathways analyzed across the research. However, we highlighted the thorough evidence for the alteration of the muscle stem cell precursor Pax7 in multiple signaling pathways described in different living models, with an emphasis on the novel approach that could be taken in manipulating Pax7 expression itself in dystrophic muscle, towards the discovery of an effective treatment for muscular dystrophy. Therefore, we believe that this could be applied to the potential gap in muscle research that could be filled by tuning the well-established marker expression to improve dystrophic muscle.
Collapse
Affiliation(s)
- Nor Idayu A. Rahman
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Chung Liang Lam
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Nadiah Sulaiman
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Nur Atiqah Haizum Abdullah
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Fazlina Nordin
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| | - Shahrul Hisham Zainal Ariffin
- Centre of Biotechnology & Functional Food, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.A.R.)
| |
Collapse
|
13
|
Wu J, Ding P, Wu H, Yang P, Guo H, Tian Y, Meng L, Zhao Q. Sarcopenia: Molecular regulatory network for loss of muscle mass and function. Front Nutr 2023; 10:1037200. [PMID: 36819699 PMCID: PMC9932270 DOI: 10.3389/fnut.2023.1037200] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Skeletal muscle is the foundation of human function and plays a key role in producing exercise, bone protection, and energy metabolism. Sarcopenia is a systemic disease, which is characterized by degenerative changes in skeletal muscle mass, strength, and function. Therefore, sarcopenia often causes weakness, prolonged hospitalization, falls and other adverse consequences that reduce the quality of life, and even lead to death. In recent years, sarcopenia has become the focus of in-depth research. Researchers have suggested some molecular mechanisms for sarcopenia according to different muscle physiology. These mechanisms cover neuromuscular junction lesion, imbalance of protein synthesis and breakdown, satellite cells dysfunction, etc. We summarize the latest research progress on the molecular mechanism of sarcopenia in this review in order to provide new ideas for future researchers to find valuable therapeutic targets and develop relevant prevention strategies.
Collapse
Affiliation(s)
- Jiaxiang Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Ping’an Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Haotian Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Peigang Yang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Honghai Guo
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Yuan Tian
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China
| | - Lingjiao Meng
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China,Research Center of the Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Lingjiao Meng,
| | - Qun Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China,Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China,*Correspondence: Qun Zhao,
| |
Collapse
|
14
|
Fujimaki S, Ono Y. Murine Models of Tenotomy-Induced Mechanical Overloading and Tail-Suspension-Induced Mechanical Unloading. Methods Mol Biol 2023; 2640:207-215. [PMID: 36995597 DOI: 10.1007/978-1-0716-3036-5_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Skeletal muscle is a highly plastic tissue that can alter its mass and strength in response to mechanical stimulation, such as overloading and unloading, which lead to muscle hypertrophy and atrophy, respectively. Mechanical loading in the muscle influences muscle stem cell dynamics, including activation, proliferation, and differentiation. Although experimental models of mechanical overloading and unloading have been widely used for the investigation of the molecular mechanisms regulating muscle plasticity and stem cell function, few studies have described the methods in detail. Here, we describe the appropriate procedures for tenotomy-induced mechanical overloading and tail-suspension-induced mechanical unloading, which are the most common and simple methods to induce muscle hypertrophy and atrophy in mouse models.
Collapse
Affiliation(s)
- Shin Fujimaki
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
15
|
Lee B, Kim SK, Shin YJ, Son YH, Yang JW, Lee SM, Yang YR, Lee KP, Kwon KS. Genome-wide analysis of a cellular exercise model based on electrical pulse stimulation. Sci Rep 2022; 12:21251. [PMID: 36481702 PMCID: PMC9731977 DOI: 10.1038/s41598-022-25758-2] [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: 04/14/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle communicates with other organs via myokines, which are secreted by muscle during exercise and exert various effects. Despite much investigation of the exercise, the underlying molecular mechanisms are still not fully understood. Here, we applied an in vitro exercise model in which cultured C2C12 myotubes were subjected to electrical pulse stimulation (EPS), which mimics contracting muscle. Based on the significantly up- and down-regulated genes in EPS, we constructed an in silico model to predict exercise responses at the transcriptional level. The in silico model revealed similarities in the transcriptomes of the EPS and exercised animals. Comparative analysis of the EPS data and exercised mouse muscle identified putative biomarkers in exercise signaling pathways and enabled to discover novel exercise-induced myokines. Biochemical analysis of selected exercise signature genes in muscle from exercised mice showed that EPS mimics in vivo exercise, at least in part, at the transcriptional level. Consequently, we provide a novel myokine, Amphiregulin (AREG), up-regulated both in vitro and in vivo, that would be a potential target for exercise mimetics.
Collapse
Affiliation(s)
- Bora Lee
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Seon Kyu Kim
- grid.249967.70000 0004 0636 3099Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea ,grid.412786.e0000 0004 1791 8264Department of Bioinformatics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea
| | - Yeo Jin Shin
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Young Hoon Son
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Jae Won Yang
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea ,grid.412786.e0000 0004 1791 8264Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea
| | - Seung-Min Lee
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Yong Ryul Yang
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea
| | - Kwang-Pyo Lee
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea ,grid.412786.e0000 0004 1791 8264Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea ,Aventi Inc., Daejeon, 34141 Republic of Korea
| | - Ki-Sun Kwon
- grid.249967.70000 0004 0636 3099Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141 Republic of Korea ,grid.412786.e0000 0004 1791 8264Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113 Republic of Korea ,Aventi Inc., Daejeon, 34141 Republic of Korea
| |
Collapse
|
16
|
Li-Li F, Bo-Wen L, Yue X, Zhen-Jun T, Meng-Xin C. Aerobic exercise and resistance exercise alleviate skeletal muscle atrophy through IGF-1/IGF-1R-PI3K/Akt pathway in mice with myocardial infarction. Am J Physiol Cell Physiol 2021; 322:C164-C176. [PMID: 34852207 DOI: 10.1152/ajpcell.00344.2021] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVES Myocardial infarction (MI)-induced heart failure (HF) is commonly accompanied with profound effects on skeletal muscle. With the process of MI-induced HF, perturbations in skeletal muscle contribute to muscle atrophy. Exercise is viewed as a feasible strategy to prevent muscle atrophy. The aims of this study were to investigate whether exercise could alleviate MI-induced skeletal muscle atrophy via insulin-like growth factor 1 (IGF-1) pathway in mice. MATERIALS AND METHODS Male C57/BL6 mice were used to establish the MI model and divided into three groups: sedentary MI group, MI with aerobic exercise group and MI with resistance exercise group, sham-operated group was used as control. Exercise-trained animals were subjected to four-weeks of aerobic exercise (AE) or resistance exercise (RE). Cardiac function, muscle weight, myofiber size, levels of IGF-1 signaling and proteins related to myogenesis, protein synthesis and degradation and cell apoptosis in gastrocnemius muscle were detected. And H2O2-treated C2C12 cells were intervened with recombinant human IGF-1, IGF-1R inhibitor NVP-AEW541 and PI3K inhibitor LY294002 to explore the mechanism. Results:Exercises up-regulated the IGF-1/IGF-1R-phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling, increased the expressions of Pax7, myogenic regulatory factors (MRFs) and protein synthesis, reduced protein degradation and cell apoptosis in MI-mice. In vitro, IGF-1 up-regulated the levels of Pax7 and MRFs, mTOR and P70S6K, reduced MuRF1, MAFbx and inhibited cell apoptosis via IGF-1R-PI3K/Akt pathway. CONCLUSION AE and RE, safely and effectively, alleviate skeletal muscle atrophy by regulating the levels of myogenesis, protein degradation and cells apoptosis in mice with MI via activating IGF-1/IGF-1R-PI3K/Akt pathway.
Collapse
Affiliation(s)
- Feng Li-Li
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Li Bo-Wen
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, China.,College of Education, Physical Education Department, Zhejiang University, China
| | - Xi Yue
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Tian Zhen-Jun
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Cai Meng-Xin
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
17
|
Fujimaki T, Ando T, Hata T, Takayama Y, Ohba T, Ichikawa J, Takiyama Y, Tatsuno R, Koyama K, Haro H. Exogenous parathyroid hormone attenuates ovariectomy-induced skeletal muscle weakness in vivo. Bone 2021; 151:116029. [PMID: 34111645 DOI: 10.1016/j.bone.2021.116029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/01/2021] [Accepted: 06/01/2021] [Indexed: 10/21/2022]
Abstract
Osteoporosis commonly affects the elderly and is associated with significant morbidity and mortality. Loss of bone mineral density induces muscle atrophy and increases fracture risk. However, muscle lipid content and droplet size are increased by aging and mobility impairments, inversely correlated with muscle function, and a cause of reduced motor function. Teriparatide, the synthetic form of human parathyroid hormone (PTH) 1-34, has been widely used to treat osteoporosis. Although PTH positively affects muscle differentiation in vitro, the precise function and mechanisms of muscle mass and power preservation are still poorly understood, especially in vivo. In this study, we investigated the effect of PTH on skeletal muscle atrophy and dysfunction using an ovariectomized murine model. Eight-week-old female C57BL/6J mice were ovariectomized or sham-operated. Within each surgical group, the mice were divided into PTH injection or control subgroups. Motor function was evaluated based on grip strength, treadmill running, and lactic acid concentration. PTH receptor was expressed in skeletal muscle cells and myoblasts. PTH inhibited ovariectomy-induced bone loss but not uterine atrophy or increased body weight; PTH not only abolished ovariectomy-induced reduction in grip strength and maximum running speed, but also significantly reduced the ovariectomy-induced increase in lactic acid concentration (compared with that observed in the vehicle control). PTH also abrogated the ovariectomy-induced reduction in the oxidative capacity of muscle fibers, their cross-sectional area, and intramyocellular lipid content, and induced cell proliferation, cell migration, and muscle differentiation, while reducing lipid secretion by C2C12 myoblasts via the Wnt/β-catenin pathway. PTH significantly ameliorated muscle weakness and attenuated exercise-induced lactate levels in ovariectomized mice. Our in vitro study demonstrated that PTH/Wnt signaling regulated the proliferation, migration, and differentiation of myoblasts and also reduced lipid secretion in myoblasts. Thus, PTH could regulate several aspects of muscle function and physiology, and may represent a novel therapeutic strategy for patients with osteoporosis.
Collapse
Affiliation(s)
- Taro Fujimaki
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Takashi Ando
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan.
| | - Takanori Hata
- Department of Neurology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yoshihiro Takayama
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Tetsuro Ohba
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Jiro Ichikawa
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yoshihisa Takiyama
- Department of Neurology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Rikito Tatsuno
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Katsuhiro Koyama
- Graduate School Department of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Hirotaka Haro
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
18
|
Zhu X, Guo W. Meta-Analyses of Multiple Gene Expression Profiles to Screen Hub Genes Related to Osteoarthritis. Public Health Genomics 2021; 24:267-279. [PMID: 34340232 DOI: 10.1159/000517308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/15/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND This study aimed to screen and validate the crucial genes involved in osteoarthritis (OA) and explore its potential molecular mechanisms. METHODS Four expression profile datasets related to OA were downloaded from the Gene Expression Omnibus (GEO). The differentially expressed genes (DEGs) from 4 microarray patterns were identified by the meta-analysis method. The weighted gene co-expression network analysis (WGCNA) method was used to investigate stable modules most related to OA. In addition, a protein-protein interaction (PPI) network was built to explore hub genes in OA. Moreover, OA-related genes and pathways were retrieved from Comparative Toxicogenomics Database (CTD). RESULTS A total of 1,136 DEGs were identified from 4 datasets. Based on these DEGs, WGCNA further explored 370 genes included in the 3 OA-related stable modules. A total of 10 hub genes were identified in the PPI network, including AKT1, CDC42, HLA-DQA2, TUBB, TWISTNB, GSK3B, FZD2, KLC1, GUSB, and RHOG. Besides, 5 pathways including "Lysosome," "Pathways in cancer," "Wnt signaling pathway," "ECM-receptor interaction" and "Focal adhesion" in CTD and enrichment analysis and 5 OA-related hub genes (including GSK3B, CDC42, AKT1, FZD2, and GUSB) were identified. CONCLUSION In this study, the meta-analysis was used to screen the central genes associated with OA in a variety of gene expression profiles. Three OA-related modules (green, turquoise, and yellow) containing 370 genes were identified through WGCNA. It was discovered through the gene-pathway network that GSK3B, CDC42, AKT1, FZD2, and GUSB may be key genes related to the progress of OA and may become promising therapeutic targets.
Collapse
Affiliation(s)
- Xianyang Zhu
- Department of Orthopedics, Taizhou People's Hospital, Taizhou, China
| | - Wen Guo
- Department of Orthopedics, Taizhou People's Hospital, Taizhou, China
| |
Collapse
|
19
|
Gasperetti A, James CA, Cerrone M, Delmar M, Calkins H, Duru F. Arrhythmogenic right ventricular cardiomyopathy and sports activity: from molecular pathways in diseased hearts to new insights into the athletic heart mimicry. Eur Heart J 2021; 42:1231-1243. [PMID: 33200174 DOI: 10.1093/eurheartj/ehaa821] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/12/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited disease associated with a high risk of sudden cardiac death. Among other factors, physical exercise has been clearly identified as a strong determinant of phenotypic expression of the disease, arrhythmia risk, and disease progression. Because of this, current guidelines advise that individuals with ARVC should not participate in competitive or frequent high-intensity endurance exercise. Exercise-induced electrical and morphological para-physiological remodelling (the so-called 'athlete's heart') may mimic several of the classic features of ARVC. Therefore, the current International Task Force Criteria for disease diagnosis may not perform as well in athletes. Clear adjudication between the two conditions is often a real challenge, with false positives, that may lead to unnecessary treatments, and false negatives, which may leave patients unprotected, both of which are equally inacceptable. This review aims to summarize the molecular interactions caused by physical activity in inducing cardiac structural alterations, and the impact of sports on arrhythmia occurrence and other clinical consequences in patients with ARVC, and help the physicians in setting the two conditions apart.
Collapse
Affiliation(s)
- Alessio Gasperetti
- Division of Cardiology, University Heart Center Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland
| | - Cynthia A James
- Division of Cardiology, Johns Hopkins Hospital, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Marina Cerrone
- Leon H Charney Division of Cardiology, New York University School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| | - Mario Delmar
- Leon H Charney Division of Cardiology, New York University School of Medicine, 550 1st Avenue, New York, NY 10016, USA
| | - Hugh Calkins
- Division of Cardiology, Johns Hopkins Hospital, 1800 Orleans St, Baltimore, MD 21287, USA
| | - Firat Duru
- Division of Cardiology, University Heart Center Zurich, Rämistrasse 100, CH-8091 Zurich, Switzerland.,Center for Integrative Human Physiology, University of Zurich, Rämistrasse 71, Zurich 8006, Switzerland
| |
Collapse
|
20
|
Heidari S, Azizbeigi K, Bahmanpour K. MicroRNA-145, Wnt3a, and Dab2 Genes Expression Changes of the Cardiomyocytes in Hypercholesterolemic Rats Exposed to the Aerobic Training. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:288-296. [PMID: 35875335 PMCID: PMC9273158 DOI: 10.22088/ijmcm.bums.10.4.288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/10/2022] [Indexed: 11/16/2022]
Abstract
The current study aimed to investigate the effect of a 12-week endurance training (ET) on microRNA-145 (miR-145) changes and Wnt3a and Dab2 cardiomyocytes genes expression of hypercholesterolemic Wistar male rats. Thirty-two male Wistar rats (191.2±19 g, 6-8 weeks age) were randomly assigned into the aerobic exercise-normal nutrition (ANN; n=8), hypercholesterolemic (HCL; n=8), aerobic exercise- hypercholesterolemic (ACL; n=8), and normal nutrition (NN; n=8). Hypercholesterolemia was created by adding 1% cholesterol to the food of the HCL and ACL rats. ET was done five sessions per week on nonconsecutive days for 12 weeks. Twenty-four hours after the last training session, the rats were killed, and the cardiomyocytes were removed. The expression of miR-145, Wnt3a, and Dab2 genes in cardiomyocytes was assessed by real time PCR method. The expression of miR-145 significantly increased in the ANN group in comparison with other groups (P = 0.001). Also, Dab2 gene expression significantly decreased in the ANN group in comparison with ACL (P = 0.001) and HCL (P = 0.001) groups. The results also showed that the Wnt3a in the ANN group was significantly different from NN (p=0.001), ACL, and HCL (p=0.001) groups. It can be concluded that aerobic training and cholesterol-rich foods play an essential regulatory role in the expression of miR-145, Dab2, and Wnt3a genes. However, cholesterol-rich foods appear to play a more significant regulatory role than aerobic exercise training.
Collapse
Affiliation(s)
- Soran Heidari
- Department of Physical Education, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.
| | - Kamal Azizbeigi
- Department of Physical Education, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.,Corresponding author: Department of Physical Education, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.
| | - Kaveh Bahmanpour
- Nursing Department, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran.
| |
Collapse
|
21
|
Tidball JG, Flores I, Welc SS, Wehling-Henricks M, Ochi E. Aging of the immune system and impaired muscle regeneration: A failure of immunomodulation of adult myogenesis. Exp Gerontol 2020; 145:111200. [PMID: 33359378 DOI: 10.1016/j.exger.2020.111200] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/17/2020] [Accepted: 12/08/2020] [Indexed: 12/16/2022]
Abstract
Skeletal muscle regeneration that follows acute injury is strongly influenced by interactions with immune cells that invade and proliferate in the damaged tissue. Discoveries over the past 20 years have identified many of the key mechanisms through which myeloid cells, especially macrophages, regulate muscle regeneration. In addition, lymphoid cells that include CD8+ T-cells and regulatory T-cells also significantly affect the course of muscle regeneration. During aging, the regenerative capacity of skeletal muscle declines, which can contribute to progressive loss of muscle mass and function. Those age-related reductions in muscle regeneration are accompanied by systemic, age-related changes in the immune system, that affect many of the myeloid and lymphoid cell populations that can influence muscle regeneration. In this review, we present recent discoveries that indicate that aging of the immune system contributes to the diminished regenerative capacity of aging muscle. Intrinsic, age-related changes in immune cells modify their expression of factors that affect the function of a population of muscle stem cells, called satellite cells, that are necessary for normal muscle regeneration. For example, age-related reductions in the expression of growth differentiation factor-3 (GDF3) or CXCL10 by macrophages negatively affect adult myogenesis, by disrupting regulatory interactions between macrophages and satellite cells. Those changes contribute to a reduction in the numbers and myogenic capacity of satellite cells in old muscle, which reduces their ability to restore damaged muscle. In addition, aging produces changes in the expression of molecules that regulate the inflammatory response to injured muscle, which also contributes to age-related defects in muscle regeneration. For example, age-related increases in the production of osteopontin by macrophages disrupts the normal inflammatory response to muscle injury, resulting in regenerative defects. These nascent findings represent the beginning of a newly-developing field of investigation into mechanisms through which aging of the immune system affects muscle regeneration.
Collapse
Affiliation(s)
- James G Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, United States of America; Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, United States of America; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, United States of America.
| | - Ivan Flores
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, United States of America
| | - Steven S Welc
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, United States of America
| | - Eisuke Ochi
- Hosei University, Faculty of Bioscience and Applied Chemistry, 3-7-2, Kajino, Koganei, Tokyo 184-8584, Japan
| |
Collapse
|
22
|
Yuan H, Ruan Y, Tan Y, Reed-Maldonado AB, Chen Y, Zhao D, Wang Z, Zhou F, Peng D, Banie L, Wang G, Liu J, Lin G, Qi LS, Lue TF. Regenerating Urethral Striated Muscle by CRISPRi/dCas9-KRAB-Mediated Myostatin Silencing for Obesity-Associated Stress Urinary Incontinence. CRISPR J 2020; 3:562-572. [PMID: 33346712 PMCID: PMC7757699 DOI: 10.1089/crispr.2020.0077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Overweight females are prone to obesity-associated stress urinary incontinence (OA-SUI), and there are no definitive medical therapies for this common urologic condition. This study was designed to test the hypothesis that regenerative therapy to restore urethral striated muscle (stM) and pelvic floor muscles might represent a valuable therapeutic approach. For the in vitro experiment, single-guide RNAs targeting myostatin (MSTN) were used for CRISPRi/dCas9-Kruppel associated box (KRAB)-mediated gene silencing. For the in vivo experiment, a total of 14 female lean ZUC-Leprfa 186 and 14 fatty ZUC-Leprfa 185 rats were used as control and CRISPRi-MSTN treated groups, respectively. The results indicated that lentivirus-mediated expression of MSTN CRISPRi/dCas9-KRAB caused sustained downregulation of MSTN in rat L6 myoblast cells and significantly enhanced myogenesis in vitro. In vivo, the urethral sphincter injection of lentiviral-MSTN sgRNA and lentiviral-dCas9-KRAB significantly increased the leak point pressure, the thickness of the stM layer, the ratio of stM to smooth muscle, and the number of neuromuscular junctions. Downregulation of MSTN with CRISPRi/dCas9-KRAB-mediated gene silencing significantly enhanced myogenesis in vitro and in vivo. It also improved urethral continence in the OA-SUI rat model.
Collapse
Affiliation(s)
- Huixing Yuan
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
- Department of Urology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, PR China; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Yajun Ruan
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
- Department of Urology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, PR China; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Yan Tan
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Amanda B. Reed-Maldonado
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
- Department of Urology, Tripler Army Medical Center, 1 Jarrett White Road, Honolulu, Hawaii, USA; and Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Yinwei Chen
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Dehua Zhao
- Department of Bioengineering, Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Zhao Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Feng Zhou
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Dongyi Peng
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, PR China; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Lei S. Qi
- Department of Bioengineering, Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| | - Tom F. Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California, USA; Department of Chemical and Systems Biology, ChEM-H, Stanford University, Stanford, California, USA
| |
Collapse
|
23
|
Bachman JF, Blanc RS, Paris ND, Kallenbach JG, Johnston CJ, Hernady E, Williams JP, Chakkalakal JV. Radiation-Induced Damage to Prepubertal Pax7+ Skeletal Muscle Stem Cells Drives Lifelong Deficits in Myofiber Size and Nuclear Number. iScience 2020; 23:101760. [PMID: 33241204 PMCID: PMC7674517 DOI: 10.1016/j.isci.2020.101760] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/08/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022] Open
Abstract
During prepubertal development, muscle stem cells (satellite cells, SCs) actively contribute to myofiber growth. Because some SCs are active during this time, they may be particularly susceptible to damage. Using a Small Animal Radiation Research Platform (SARRP), we investigated the effects of local fractionated radiation treatment on prepubertal SCs. Immediately after this regimen, there was a reduction in SC number. Although surviving SCs had deficiencies in function, some myogenic potential remained. Indeed, some muscle regenerative capacity persisted immediately after irradiation. Lastly, we assessed the long-term consequences of radiation-induced SC loss during prepuberty. We observed a reduction of myofiber size and corresponding loss of nuclei in both fast- and slow-contracting muscles 14 months post-irradiation. Notably, prepubertal SC depletion mimicked these lifelong deficits. This work highlights the susceptibility of prepubertal SCs to radiation exposure. We also reveal the importance of prepubertal SC contribution to the lifelong maintenance of skeletal muscle. Increased sensitivity of satellite cells to irradiation during prepubertal growth Prepubertal irradiation leads to lifelong deficits in skeletal muscle regenerative capacity Lifelong reduction in myofiber size and nuclear number is a consequence of prepubertal irradiation Satellite cell ablation mimics the lifelong effects of prepubertal irradiation on myofiber size and nuclear number
Collapse
Affiliation(s)
- John F Bachman
- Department of Pathology and Laboratory Medicine, Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, USA.,Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Roméo S Blanc
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Nicole D Paris
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacob G Kallenbach
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Carl J Johnston
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Eric Hernady
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacqueline P Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Joe V Chakkalakal
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.,Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, NY, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
24
|
Riasat K, Bardell D, Goljanek-Whysall K, Clegg PD, Peffers MJ. Epigenetic mechanisms in Tendon Ageing. Br Med Bull 2020; 135:90-107. [PMID: 32827252 PMCID: PMC7585832 DOI: 10.1093/bmb/ldaa023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Tendon is a composite material with a well-ordered hierarchical structure exhibiting viscoelastic properties designed to transfer force. It is recognized that the incidence of tendon injury increases with age, suggesting a deterioration in homeostatic mechanisms or reparative processes. This review summarizes epigenetic mechanisms identified in ageing healthy tendon. SOURCES OF DATA We searched multiple databases to produce a systematic review on the role of epigenetic mechanisms in tendon ageing. AREAS OF AGREEMENT Epigenetic mechanisms are important in predisposing ageing tendon to injury. AREAS OF CONTROVERSY The relative importance of epigenetic mechanisms are unknown in terms of promoting healthy ageing. It is also unknown whether these changes represent protective mechanisms to function or predispose to pathology. GROWING POINT Epigenetic markers in ageing tendon, which are under-researched including genome-wide chromatin accessibility, should be investigated. AREAS TIMELY FOR DEVELOPING RESEARCH Metanalysis through integration of multiple datasets and platforms will enable a holistic understanding of the epigenome in ageing and its relevance to disease.
Collapse
Affiliation(s)
- Kiran Riasat
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - David Bardell
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK.,Institute of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, Wirral CH64 7TE, UK
| | - Katarzyna Goljanek-Whysall
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Peter D Clegg
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Mandy J Peffers
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| |
Collapse
|
25
|
Sameri S, Samadi P, Dehghan R, Salem E, Fayazi N, Amini R. Stem Cell Aging in Lifespan and Disease: A State-of-the-Art Review. Curr Stem Cell Res Ther 2020; 15:362-378. [DOI: 10.2174/1574888x15666200213105155] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/09/2019] [Accepted: 12/31/2019] [Indexed: 12/11/2022]
Abstract
Aging is considered as inevitable changes at different levels of genome, cell, and organism.
From the accumulation of DNA damages to imperfect protein homeostasis, altered cellular communication
and exhaustion of stem cells, aging is a major risk factor for many prevalent diseases, such as
cancer, cardiovascular disease, pulmonary disease, diabetes, and neurological disorders. The cells are
dynamic systems, which, through a cycle of processes such as replication, growth, and death, could
replenish the bodies’ organs and tissues, keeping an entire organism in optimal working order. In many
different tissues, adult stem cells are behind these processes, replenishing dying cells to maintain normal
tissue function and regenerating injured tissues. Therefore, adult stem cells play a vital role in preventing
the aging of organs and tissues, and can delay aging. However, during aging, these cells also
undergo some detrimental changes such as alterations in the microenvironment, a decline in the regenerative
capacity, and loss of function. This review aimed to discuss age-related changes of stem cells in
different tissues and cells, including skin, muscles, brain, heart, hair follicles, liver, and lung.
Collapse
Affiliation(s)
- Saba Sameri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Pouria Samadi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Razieh Dehghan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Elham Salem
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nashmin Fayazi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
26
|
Brett JO, Arjona M, Ikeda M, Quarta M, de Morrée A, Egner IM, Perandini LA, Ishak HD, Goshayeshi A, Benjamin DI, Both P, Rodríguez-Mateo C, Betley MJ, Wyss-Coray T, Rando TA. Exercise rejuvenates quiescent skeletal muscle stem cells in old mice through restoration of Cyclin D1. Nat Metab 2020; 2:307-317. [PMID: 32601609 PMCID: PMC7323974 DOI: 10.1038/s42255-020-0190-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/12/2020] [Indexed: 01/01/2023]
Abstract
Aging impairs tissue repair. This is pronounced in skeletal muscle, whose regeneration by muscle stem cells (MuSCs) is robust in young adult animals but inefficient in older organisms. Despite this functional decline, old MuSCs are amenable to rejuvenation through strategies that improve the systemic milieu, such as heterochronic parabiosis. One such strategy, exercise, has long been appreciated for its benefits on healthspan, but its effects on aged stem cell function in the context of tissue regeneration are incompletely understood. Here we show that exercise in the form of voluntary wheel running accelerates muscle repair in old animals and improves old MuSC function. Through transcriptional profiling and genetic studies, we discovered that the restoration of old MuSC activation ability hinges on restoration of Cyclin D1, whose expression declines with age in MuSCs. Pharmacologic studies revealed that Cyclin D1 maintains MuSC activation capacity by repressing TGFβ signaling. Taken together, these studies demonstrate that voluntary exercise is a practicable intervention for old MuSC rejuvenation. Furthermore, this work highlights the distinct role of Cyclin D1 in stem cell quiescence.
Collapse
Affiliation(s)
- Jamie O Brett
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Marina Arjona
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Mika Ikeda
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Marco Quarta
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
- Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Antoine de Morrée
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Ingrid M Egner
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Luiz A Perandini
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Heather D Ishak
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Armon Goshayeshi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel I Benjamin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Pieter Both
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
- Stem Cell Biology and Regenerative Medicine Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Cristina Rodríguez-Mateo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael J Betley
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Neurosciences Interdepartmental Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
- Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Paul F. Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.
- Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA.
- Neurology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
27
|
Chen X, Xu H, Hou J, Wang H, Zheng Y, Li H, Cai H, Han X, Dai J. Epithelial cell senescence induces pulmonary fibrosis through Nanog-mediated fibroblast activation. Aging (Albany NY) 2019; 12:242-259. [PMID: 31891567 PMCID: PMC6977687 DOI: 10.18632/aging.102613] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive lung disease tightly correlated with aging. The pathological features of IPF include epithelial cell senescence and abundant foci of highly activated pulmonary fibroblasts. However, the underlying mechanism between epithelial cell senescence and pulmonary fibroblast activation remain to be elucidated. In our study, we demonstrated that Nanog, as a pluripotency gene, played an essential role in the activation of pulmonary fibroblasts. In the progression of IPF, senescent epithelial cells could contribute to the activation of pulmonary fibroblasts via increasing the expression of senescence-associated secretory phenotype (SASP). In addition, we found activated pulmonary fibroblasts exhibited aberrant activation of Wnt/β-catenin signalling and elevated expression of Nanog. Further study revealed that the activation of Wnt/β-catenin signalling was responsible for senescent epithelial cell-induced Nanog phenotype in pulmonary fibroblasts. β-catenin was observed to bind to the promoter of Nanog during the activation of pulmonary fibroblasts. Targeted inhibition of epithelial cell senescence or Nanog could effectively suppress the activation of pulmonary fibroblasts and impair the development of pulmonary fibrosis, indicating a potential for the exploration of novel anti-fibrotic strategies.
Collapse
Affiliation(s)
- Xiang Chen
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.,Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Hongyang Xu
- Department of Critical Care Medicine, The Affiliated WuXi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Jiwei Hou
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Hui Wang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yi Zheng
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hui Li
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hourong Cai
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210093, China
| | - Jinghong Dai
- Department of Pulmonary and Critical Care Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
28
|
Son YH, Lee SM, Lee SH, Yoon JH, Kang JS, Yang YR, Kwon KS. Comparative molecular analysis of endurance exercise in vivo with electrically stimulated in vitro myotube contraction. J Appl Physiol (1985) 2019; 127:1742-1753. [DOI: 10.1152/japplphysiol.00091.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Exercise has positive effects on health and improves a variety of disease conditions. An in vitro model of exercise has been developed to better understand its molecular mechanisms. While various conditions have been used to mimic in vivo exercise, no specific conditions have matched a specific type of in vivo exercise. Here, we screened various electrical pulse stimulation (EPS) conditions and compared the molecular events under each condition in myotube culture with that obtained under voluntary wheel running (VWR), a mild endurance exercise, in mice. Both EPS and VWR upregulated the mRNA levels of genes involved in the slow-type twitch ( Myh7 and Myh2) and myogenesis ( Myod and Myog) and increased the protein expression of peroxisome proliferator-activated receptor-γ coactivator-1α, which is involved in mitochondrial biogenesis. These changes were accompanied by activation of p38 and AMPK. However, neither condition induced the expression of muscle-specific E3 ligases such as MAFbx and MuRF1. Both EPS and VWR consistently induced antioxidant genes such as Sod3 and Gpx4 but did not cause similar changes in the expression levels of the calcium channel/pump-related genes Ryr and Serca. Furthermore, both EPS and VWR reduced glycogen levels but not lactate levels as assessed in post-EPS culture medium and post-VWR serum, respectively. Thus we identified an in vitro EPS condition that effectively mimics VWR in mice, which can facilitate further studies of the detailed molecular mechanisms of endurance exercise in the absence of interference from multiple tissues and organs. NEW & NOTEWORTHY This study establishes an optimal condition for electrical pulse stimulation (EPS) in myotubes that shows a similar molecular signature as voluntary wheel running. The specific EPS condition 1) upregulates the mRNA of slow-twitch muscle components and myogenic transcription factors, 2) induces antioxidant genes without any muscle damage, and 3) promotes peroxisome proliferator-activated receptor-γ coactivator-1α and its upstream regulators involved in mitochondrial biogenesis.
Collapse
Affiliation(s)
- Young Hoon Son
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Seung-Min Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Seol Hee Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jong Hyeon Yoon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jae Sook Kang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Yong Ryoul Yang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
29
|
Li L, Cheng X, Chen L, Li J, Luo W, Li C. Long Noncoding Ribonucleic Acid MSTRG.59589 Promotes Porcine Skeletal Muscle Satellite Cells Differentiation by Enhancing the Function of PALLD. Front Genet 2019; 10:1220. [PMID: 31850071 PMCID: PMC6887656 DOI: 10.3389/fgene.2019.01220] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/05/2019] [Indexed: 01/08/2023] Open
Abstract
Skeletal muscle satellite cells are a class of undifferentiated mononuclear myogenic stem cells distributed between the myofibroblast and membrane basement. Since their development determines the development of skeletal muscles, knowledge of their proliferation, differentiation, and fate is vital for understanding skeletal muscle development. Increasing evidence have shown that long noncoding RNA (lncRNA) plays an important role in regulating the development process of satellite cells. Based on the results of our previous studies, we screened lncRNA MSTRG.59589, which is highly expressed in skeletal muscle tissue. In the present study, knockdown of MSTRG.59589 significantly inhibited satellite cell differentiation at various time points, whereas overexpression of MSTRG.59589 demonstrated opposite effects. An MSTRG.59589 knockdown cell model was constructed for transcriptome sequencing, and RNA sequencing analysis screened out a large number of differentially expressed genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses of these differentially expressed genes revealed that they are mainly enriched in actin cytoskeleton, muscle contraction, and other pathways related to muscle development. Mechanistic analyses showed that MSTRG.59589 could promote the differentiation process of skeletal muscle satellite cells by positively regulating the expression level of the target gene PALLD. This experiment lays a theoretical foundation for deeper studies on the mechanism of MSTRG.59589 in the differentiation of porcine skeletal muscle satellite cells.
Collapse
Affiliation(s)
- Long Li
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Xiaofang Cheng
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Ling Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Jingxuan Li
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Wenzhe Luo
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| | - Changchun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education and Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
30
|
Cui K, Kang N, Banie L, Zhou T, Liu T, Wang B, Ruan Y, Peng D, Wang HS, Wang T, Wang G, Reed-Maldonado AB, Chen Z, Lin G, Lue TF. Microenergy acoustic pulses induced myogenesis of urethral striated muscle stem/progenitor cells. Transl Androl Urol 2019; 8:489-500. [PMID: 32133280 DOI: 10.21037/tau.2019.08.18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Stress urinary incontinence (SUI) is a common disorder with high prevalence in women across their life span, but there are no non-surgical curative options for the condition. Stem cell-based therapy, especially endogenous stem cell therapy may be a potential treatment method for SUI. The aims of this study are to identify, isolate, and assay the function of urethral striated muscle derived stem/progenitor cells (uMDSCs) and to assess uMDSC response to microenergy acoustic pulses (MAP). Methods Urethral striated muscle was identified utilizing 3D imaging of solvent organs (3DISCO) and immunofluorescence (IF). uMDSCs were isolated and purified from Zucker Lean (ZL) (ZUC-LEAN) (ZUC-Leprfa 186) rats, with magnetic-activated cell sorting (MACS) and pre-plating methods. The stemness and differentiation potential of the uMDSCs were measured by cell proliferation, EdU, flow cytometry, IF, and Western blot. Results Comparison of the cell proliferation assays between MACS and pre-plating reveals the advantage of MACS over pre-plating. In addition, the study reveals that uMDSCs form myotubes when treated with MAP. Conclusions The uMDSCs within female rat urethral striated muscle could be a therapeutic target of MAP in managing SUI.
Collapse
Affiliation(s)
- Kai Cui
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.,Department of Urology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ning Kang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Tie Zhou
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.,Department of Urology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai 200433, China
| | - Tianshu Liu
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Bohan Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Yajun Ruan
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Dongyi Peng
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Hsun Shuan Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Tianyu Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Amanda B Reed-Maldonado
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Zhong Chen
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.,Department of Urology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
31
|
Liu D, Li S, Cui Y, Tong H, Li S, Yan Y. Podocan affects C2C12 myogenic differentiation by enhancing Wnt/β-catenin signaling. J Cell Physiol 2019; 234:11130-11139. [PMID: 30652305 DOI: 10.1002/jcp.27763] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022]
Abstract
Podocan, a small leucine-rich repeat protein, is a negative regulator of cell proliferation. In this study, we demonstrated that podocan is involved in the differentiation of C2C12 murine myoblasts. Podocan expression increased with the progression of C2C12 differentiation. As expect, siRNA-mediated podocan knockdown inhibited C2C12 differentiation, as indicated by inhibition of MYOG, MYH2, and desmin expression, as well as reductions in the differentiation and fusion indices. Overexpression of podocan using dCas9 technology promoted C2C12 cell differentiation. In addition, supplementation of culture medium with podocan influenced C2C12 differentiation. Podocan knockdown reduced Wnt/β-catenin signaling, characterized by a reduction in the nuclear translocation of β-catenin, whereas podocan overexpression had the opposite effect. Furthermore, treatment with XAV939, an inhibitor of Wnt/β-catenin, reduced the podocan-mediated promotion of C2C12 differentiation. Induction of muscle injury in mice by bupivacaine administration suggested that podocan may play a role in muscle regeneration. In summary, our results suggest that podocan is required for normal C2C12 differentiation and that its role in myogenesis is mediated by the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Dan Liu
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shuang Li
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yafeng Cui
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Huili Tong
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shufeng Li
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yunqin Yan
- The Laboratory of Cell and Development, Northeast Agricultural University, Harbin, Heilongjiang, China
| |
Collapse
|
32
|
Bonewald L. Use it or lose it to age: A review of bone and muscle communication. Bone 2019; 120:212-218. [PMID: 30408611 PMCID: PMC6360108 DOI: 10.1016/j.bone.2018.11.002] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/03/2018] [Accepted: 11/03/2018] [Indexed: 12/21/2022]
Abstract
Until recently, it was assumed that the only interaction between muscle and bone is mechanical, that the muscle acts as a pulley and the bone as a lever to move the organism. A relatively new concept is that muscle, especially contracted muscle, acts as a secretory organ, regulating metabolism. An even newer concept is that bone, especially the osteocytes in bone, act as endocrine cells targeting other organs such as kidney and more recently, muscle. These two new concepts logically led to the third concept: that muscle and bone communicate via soluble factors. Crosstalk occurs through muscle factors such as myostatin, irisin, and a muscle metabolite, β-aminoisobutyric acid, BAIBA, and through bone factors such as osteocalcin, transforming growth factor beta, TGFβ, Prostaglandin E2, PGE2 and Wnts. Some of these factors have positive and some negative effects on the opposing tissue. One feature both bone and muscle have in common is that their tissues are mechanically loaded and many of their secreted factors are regulated by load. This mechanical loading, also known as exercise, has beneficial effects on many systems leading to the hypothesis that muscle and bone factors can be responsible for the beneficial effects of exercise. Many of the characteristics of aging and diseases associated with aging such as sarcopenia and osteoporosis and neurological conditions such as Alzheimer's disease and dementia, are delayed by exercise. This beneficial effect has been ascribed to increased blood flow increasing oxygen and nutrients, but could also be due to the secretome of the musculoskeletal system as outlined in this review.
Collapse
|
33
|
Fariyike B, Singleton Q, Hunter M, Hill WD, Isales CM, Hamrick MW, Fulzele S. Role of MicroRNA-141 in the Aging Musculoskeletal System: A Current Overview. Mech Ageing Dev 2019; 178:9-15. [PMID: 30528652 PMCID: PMC6998035 DOI: 10.1016/j.mad.2018.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/31/2018] [Accepted: 12/05/2018] [Indexed: 02/07/2023]
Abstract
MicroRNA's are small non-coding RNAs that regulate the expression of genes by targeting the 3' UTR's of mRNA. Studies reveal that miRNAs play a pivotal role in normal musculoskeletal function such as mesenchymal stem cell differentiation, survivability and apoptosis, osteogenesis, and chondrogenesis. Changes in normal miRNA expression have been linked to a number of pathological disease processes. Additionally, with aging, it is noted that there is dysregulation in the normal function of stem cell differentiation, bone formation/degradation, chondrocyte function, and muscle degeneration. Due to the change in expression of miRNA in degenerative musculoskeletal pathology, it is believed that these molecules may be at least partially responsible for cellular dysfunction. A number of miRNAs have already been identified to play a role in osteoarthritis, osteoporosis and sarcopenia. One miRNA that has become of interest recently is miRNA 141. The purpose of this article is to review the current literature available on miRNA 141 and how it could play a role in osteoporosis, osteoarthritis and musculoskeletal pathology overall.
Collapse
Affiliation(s)
- Babatunde Fariyike
- Department of Orthopedics, Augusta University, Augusta, GA, United States
| | - Quante Singleton
- Department of Orthopedics, Augusta University, Augusta, GA, United States
| | - Monte Hunter
- Department of Orthopedics, Augusta University, Augusta, GA, United States
| | - William D Hill
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States; Charlie Norwood VA Medical Center, Augusta, GA, United States
| | - Carlos M Isales
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Department of Medicine, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Mark W Hamrick
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Department of Cell biology and Anatomy, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States
| | - Sadanand Fulzele
- Department of Orthopedics, Augusta University, Augusta, GA, United States; Institute of Regenerative and Reparative medicine, Augusta University, Augusta, GA, United States.
| |
Collapse
|
34
|
Hu S, Yang L, Wu C, Liu TY. Regulation of Wnt signaling by physical exercise in the cell biological processes of the locomotor system. Physiol Int 2019; 106:1-20. [PMID: 30917670 DOI: 10.1556/2060.106.2019.07] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the past decade, researches on Wnt signaling in cell biology have made remarkable progress regarding our understanding of embryonic development, bone formation, muscle injury and repair, neurogenesis, and tumorigenesis. The study also showed that physical activity can reverse age-dependent decline in skeletal muscle, preventing osteoporosis, regenerative neurogenesis, hippocampal function, cognitive ability, and neuromuscular junction formation, and the age-dependent recession is highly correlated with Wnt signaling pathways. However, how the biological processes in cell and physical activity during/following exercise affect the Wnt signaling path of the locomotor system is largely unknown. In this study, we first briefly introduce the important features of the cellular biological processes of exercise in the locomotor system. Then, we discuss Wnt signaling and review the very few studies that have examined Wnt signaling pathways in cellular biological processes of the locomotor system during physical exercise.
Collapse
Affiliation(s)
- S Hu
- 1 College of Physical Education and Sports Science, HengYang Normal University , Hengyang, Hunan, China
| | - L Yang
- 2 Department of Neuroscience and Regenerative Medicine, Augusta University , Augusta, GA, USA
| | - C Wu
- 3 Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University , Guangzhou, China
| | - Tc-Y Liu
- 3 Laboratory of Laser Sports Medicine, College of Physical Education and Sports Science, South China Normal University , Guangzhou, China
| |
Collapse
|
35
|
Lee DE, Bareja A, Bartlett DB, White JP. Autophagy as a Therapeutic Target to Enhance Aged Muscle Regeneration. Cells 2019; 8:cells8020183. [PMID: 30791569 PMCID: PMC6406986 DOI: 10.3390/cells8020183] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/30/2019] [Accepted: 02/14/2019] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle has remarkable regenerative capacity, relying on precise coordination between resident muscle stem cells (satellite cells) and the immune system. The age-related decline in skeletal muscle regenerative capacity contributes to the onset of sarcopenia, prolonged hospitalization, and loss of autonomy. Although several age-sensitive pathways have been identified, further investigation is needed to define targets of cellular dysfunction. Autophagy, a process of cellular catabolism, is emerging as a key regulator of muscle regeneration affecting stem cell, immune cell, and myofiber function. Muscle stem cell senescence is associated with a suppression of autophagy during key phases of the regenerative program. Macrophages, a key immune cell involved in muscle repair, also rely on autophagy to aid in tissue repair. This review will focus on the role of autophagy in various aspects of the regenerative program, including adult skeletal muscle stem cells, monocytes/macrophages, and corresponding age-associated dysfunction. Furthermore, we will highlight rejuvenation strategies that alter autophagy to improve muscle regenerative function.
Collapse
Affiliation(s)
- David E Lee
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA.
| | - Akshay Bareja
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA.
| | - David B Bartlett
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA.
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Durham, NC 27701, USA.
- Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27701, USA.
| | - James P White
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27701, USA.
- Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, Durham, NC 27701, USA.
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC 27701, USA.
| |
Collapse
|
36
|
Nagahisa H, Miyata H. Influence of hypoxic stimulation on angiogenesis and satellite cells in mouse skeletal muscle. PLoS One 2018; 13:e0207040. [PMID: 30408093 PMCID: PMC6224099 DOI: 10.1371/journal.pone.0207040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/23/2018] [Indexed: 01/22/2023] Open
Abstract
We clarified in our previous study that hypoxic training promotes angiogenesis in skeletal muscle, but the mechanism of angiogenesis in skeletal muscle remains unknown. In this study, we investigated the influence of differences in hypoxia exposure on angiogenesis in skeletal muscles at differing ages and metabolic characteristics at which the production of reactive oxygen species and nitric oxide may differ. Ten-week-old (young) and 20-month-old (old) mice were separated into control (N), continuous hypoxia (H), and intermittent hypoxia (IH) groups. The H group was exposed to 16% O2 hypoxia for 5 days and the IH group was exposed to 16% O2 hypoxia at one-hour intervals during the light period for 5 days. After completion of hypoxia exposure, the soleus and gastrocnemius muscles were immediately excised, and mRNA expression of angiogenesis- and satellite cell-related genes was investigated using real-time RT-PCR. In addition, muscle fiber type composition, muscle fiber area, number of satellite cells, and capillary density were measured immunohistochemically. In the young soleus muscle, the muscle fiber area was decreased in the H group, and mRNA expression of satellite cell activation-related MyoD, MHCe, and BDNF was significantly increased. On the other hand, in the old soleus muscle, nNOS and VEGF-A mRNA expression, and the capillary density were significantly increased in the H group. In the superficial portion of the gastrocnemius, mRNA expression of FGF2, an angiogenic factor secreted by satellite cells, was significantly increased in the young IH group. In addition, a positive correlation between VEGF-A mRNA expression and nNOS mRNA expression in the soleus muscle and eNOS mRNA expression in the superficial portion of the gastrocnemius was noted. These data demonstrated that age, hypoxia exposure method and muscle metabolic characteristics are related, which results in significant differences in angiogenesis.
Collapse
Affiliation(s)
- Hiroshi Nagahisa
- Biological Sciences, Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi, Japan
| | - Hirofumi Miyata
- Biological Sciences, Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi, Japan
- * E-mail:
| |
Collapse
|
37
|
Mumford PW, Romero MA, Mao X, Mobley CB, Kephart WC, Haun CT, Roberson PA, Young KC, Martin JS, Yarrow JF, Beck DT, Roberts MD. Cross talk between androgen and Wnt signaling potentially contributes to age-related skeletal muscle atrophy in rats. J Appl Physiol (1985) 2018; 125:486-494. [PMID: 29722624 DOI: 10.1152/japplphysiol.00768.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We sought to determine whether age-related gastrocnemius muscle mass loss was associated with parallel decrements in androgen receptor (AR) or select Wnt signaling markers. To test this hypothesis, serum-free and total testosterone (TEST) and gastrocnemius AR and Wnt signaling markers were analyzed in male Fischer 344 rats that were 3, 6, 12, 18, and 24 mo (mo) old ( n = 9 per group). Free and total TEST was greatest in 6 mo rats, and AR protein and Wnt5 protein levels linearly declined with aging. There were associations between Wnt5 protein levels and relative gastrocnemius mass ( r = 0.395, P = 0.007) as well as AR and Wnt5 protein levels (r = 0.670, P < 0.001). We next tested the hypothesis that Wnt5 affects muscle fiber size by treating C2C12-derived myotubes with lower (75 ng/ml) and higher (150 ng/ml) concentrations of recombinant Wnt5a protein. Both treatments increased myotube size ( P < 0.05) suggesting this ligand may affect muscle fiber size in vivo. We next tested if Wnt5a protein levels were androgen-modulated by examining 10-mo-old male Fischer 344 rats ( n = 10-11 per group) that were orchiectomized and treated with testosterone-enanthate (TEST-E); trenbolone enanthate (TREN), a nonaromatizable synthetic testosterone analogue; or a vehicle (ORX only) for 4 wk. Interestingly, TEST-E and TREN treatments increased Wnt5a protein in the androgen-sensitive levator ani/bulbocavernosus muscle compared with ORX only ( P < 0.05). To summarize, aromatizable and nonaromatizable androgens increase Wnt5a protein expression in skeletal muscle, age-related decrements in muscle AR may contribute Wnt5a protein decrements, and our in vitro data imply this mechanism may contribute to age-related muscle loss. NEW & NOTEWORTHY Results from this study demonstrate androgen and Wnt5 protein expression decrease with aging, and this may be a mechanism involved with age-related muscle loss.
Collapse
Affiliation(s)
| | | | - Xuansong Mao
- School of Kinesiology, Auburn University , Auburn, Alabama
| | | | | | - Cody T Haun
- School of Kinesiology, Auburn University , Auburn, Alabama
| | | | - Kaelin C Young
- School of Kinesiology, Auburn University , Auburn, Alabama
- Edward Via College of Osteopathic Medicine-Auburn Campus , Auburn, Alabama
| | - Jeffrey S Martin
- School of Kinesiology, Auburn University , Auburn, Alabama
- Edward Via College of Osteopathic Medicine-Auburn Campus , Auburn, Alabama
| | - Joshua F Yarrow
- North Florida/South Georgia Veterans Health System, Malcom Randall Veterans Affairs Medical Center , Gainesville, Florida
- Division of Endocrinology, Diabetes, and Metabolism, University of Florida College of Medicine , Gainesville, Florida
| | - Darren T Beck
- School of Kinesiology, Auburn University , Auburn, Alabama
- Edward Via College of Osteopathic Medicine-Auburn Campus , Auburn, Alabama
| | - Michael D Roberts
- School of Kinesiology, Auburn University , Auburn, Alabama
- Edward Via College of Osteopathic Medicine-Auburn Campus , Auburn, Alabama
| |
Collapse
|
38
|
Vanderplanck C, Tassin A, Ansseau E, Charron S, Wauters A, Lancelot C, Vancutsem K, Laoudj-Chenivesse D, Belayew A, Coppée F. Overexpression of the double homeodomain protein DUX4c interferes with myofibrillogenesis and induces clustering of myonuclei. Skelet Muscle 2018; 8:2. [PMID: 29329560 PMCID: PMC5767009 DOI: 10.1186/s13395-017-0148-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Facioscapulohumeral muscular dystrophy (FSHD) is associated with DNA hypomethylation at the 4q35 D4Z4 repeat array. Both the causal gene DUX4 and its homolog DUX4c are induced. DUX4c is immunodetected in every myonucleus of proliferative cells, while DUX4 is present in only 1/1000 of myonuclei where it initiates a gene deregulation cascade. FSHD primary myoblasts differentiate into either atrophic or disorganized myotubes. DUX4 expression induces atrophic myotubes and associated FSHD markers. Although DUX4 silencing normalizes the FSHD atrophic myotube phenotype, this is not the case for the disorganized phenotype. DUX4c overexpression increases the proliferation rate of human TE671 rhabdomyosarcoma cells and inhibits their differentiation, suggesting a normal role during muscle differentiation. METHODS By gain- and loss-of-function experiments in primary human muscle cells, we studied the DUX4c impact on proliferation, differentiation, myotube morphology, and FSHD markers. RESULTS In primary myoblasts, DUX4c overexpression increased the staining intensity of KI67 (a proliferation marker) in adjacent cells and delayed differentiation. In differentiating cells, DUX4c overexpression led to the expression of some FSHD markers including β-catenin and to the formation of disorganized myotubes presenting large clusters of nuclei and cytoskeletal defects. These were more severe when DUX4c was expressed before the cytoskeleton reorganized and myofibrils assembled. In addition, endogenous DUX4c was detected at a higher level in FSHD myotubes presenting abnormal clusters of nuclei and cytoskeletal disorganization. We found that the disorganized FSHD myotube phenotype could be rescued by silencing of DUX4c, not DUX4. CONCLUSION Excess DUX4c could disturb cytoskeletal organization and nuclear distribution in FSHD myotubes. We suggest that DUX4c up-regulation could contribute to DUX4 toxicity in the muscle fibers by favoring the clustering of myonuclei and therefore facilitating DUX4 diffusion among them. Defining DUX4c functions in the healthy skeletal muscle should help to design new targeted FSHD therapy by DUX4 or DUX4c inhibition without suppressing DUX4c normal function.
Collapse
Affiliation(s)
- Céline Vanderplanck
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000 Mons, Belgium
| | - Alexandra Tassin
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000 Mons, Belgium
| | - Eugénie Ansseau
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000 Mons, Belgium
| | - Sébastien Charron
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000 Mons, Belgium
| | - Armelle Wauters
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000 Mons, Belgium
| | - Céline Lancelot
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000 Mons, Belgium
| | - Kelly Vancutsem
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000 Mons, Belgium
| | | | - Alexandra Belayew
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000 Mons, Belgium
| | - Frédérique Coppée
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, 6, Avenue du Champs de Mars, B-7000 Mons, Belgium
| |
Collapse
|
39
|
[Molecular mechanism of sarcopenia]. Nihon Ronen Igakkai Zasshi 2018; 55:13-24. [PMID: 29503355 DOI: 10.3143/geriatrics.55.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
|
40
|
Huh MS, Young KG, Yan K, Price-O’Dea T, Picketts DJ. Recovery from impaired muscle growth arises from prolonged postnatal accretion of myonuclei in Atrx mutant mice. PLoS One 2017; 12:e0186989. [PMID: 29095838 PMCID: PMC5667798 DOI: 10.1371/journal.pone.0186989] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 10/11/2017] [Indexed: 01/13/2023] Open
Abstract
Reduced muscle mass due to pathological development can occur through several mechanisms, including the loss or reduced proliferation of muscle stem cells. Muscle-specific ablation of the α-thalassemia mental retardation syndrome mutant protein, Atrx, in transgenic mice results in animals with a severely reduced muscle mass at three weeks of age; yet this muscle mass reduction resolves by adult age. Here, we explore the cellular mechanism underlying this effect. Analysis of Atrx mutant mice included testing for grip strength and rotorod performance. Muscle fiber length, fiber volume and numbers of myofiber-associated nuclei were determined from individual EDL or soleus myofibers isolated at three, five, or eight weeks. Myofibers from three week old Atrx mutant mice are smaller with fewer myofiber-associated nuclei and reduced volume compared to control animals, despite similar fiber numbers. Nonetheless, the grip strength of Atrx mutant mice was comparable to control mice when adjusted for body weight. Myofiber volume remained smaller at five weeks, becoming comparable to controls by 8 weeks of age. Concomitantly, increased numbers of myofiber-associated nuclei and Ki67+ myoblasts indicated that the recovery of muscle mass likely arises from the prolonged accretion of new myonuclei. This suggests that under disease conditions the muscle satellite stem cell niche can remain in a prolonged active state, allowing for the addition of a minimum number of myonuclei required to achieve a normal muscle size.
Collapse
Affiliation(s)
- Michael S. Huh
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON Canada
| | - Kevin G. Young
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON Canada
| | - Keqin Yan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON Canada
| | - Tina Price-O’Dea
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON Canada
| | - David J. Picketts
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON Canada
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, ON Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON Canada
- * E-mail:
| |
Collapse
|
41
|
Diabetes-Induced Dysfunction of Mitochondria and Stem Cells in Skeletal Muscle and the Nervous System. Int J Mol Sci 2017; 18:ijms18102147. [PMID: 29036909 PMCID: PMC5666829 DOI: 10.3390/ijms18102147] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/11/2017] [Indexed: 12/21/2022] Open
Abstract
Diabetes mellitus is one of the most common metabolic diseases spread all over the world, which results in hyperglycemia caused by the breakdown of insulin secretion or insulin action or both. Diabetes has been reported to disrupt the functions and dynamics of mitochondria, which play a fundamental role in regulating metabolic pathways and are crucial to maintain appropriate energy balance. Similar to mitochondria, the functions and the abilities of stem cells are attenuated under diabetic condition in several tissues. In recent years, several studies have suggested that the regulation of mitochondria functions and dynamics is critical for the precise differentiation of stem cells. Importantly, physical exercise is very useful for preventing the diabetic alteration by improving the functions of both mitochondria and stem cells. In the present review, we provide an overview of the diabetic alterations of mitochondria and stem cells and the preventive effects of physical exercise on diabetes, focused on skeletal muscle and the nervous system. We propose physical exercise as a countermeasure for the dysfunction of mitochondria and stem cells in several target tissues under diabetes complication and to improve the physiological function of patients with diabetes, resulting in their quality of life being maintained.
Collapse
|
42
|
Wang B, Zhou J, Banie L, Reed-Maldonado AB, Ning H, Lu Z, Ruan Y, Zhou T, Wang HS, Oh BS, Wang G, Qi SL, Lin G, Lue TF. Low-intensity extracorporeal shock wave therapy promotes myogenesis through PERK/ATF4 pathway. Neurourol Urodyn 2017; 37:699-707. [PMID: 28763567 DOI: 10.1002/nau.23380] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 07/10/2017] [Indexed: 12/27/2022]
Abstract
AIM Stress urinary incontinence (SUI) is a significant health problem for women. Treatments employing muscle derived stem cells (MDSCs) may be a promising approach to this prevalent, bothersome condition, but these treatments are invasive and require collection of cells from one site for injection into another. It is also unknown whether or not these cells establish themselves and function as muscle cells in the target tissues. Alternatively, low-intensity extracorporeal shock wave therapy (Li-ESWT) is non-invasive and has shown positive outcomes in the treatment of multiple musculoskeletal disorders, but the biological effects responsible for clinical success are not yet well understood. The aim of this study is to explore the possibility of employing Li-ESWT for activation of MDSCs in situ and to further elucidate the underlying biological effects and mechanisms of action in urethral muscle. METHODS Urethral muscle derived stem cells (uMDSCs) were harvest from Zucker Lean (ZUC-LEAN) (ZUC-Leprfa 186) rats and characterized with flow cytometry. Li-ESWT (0.02 mJ/mm2 , 3 Hz, 200 pulses) and GSK2656157, an inhibitor of PERK pathway, were applied to L6 rat myoblast cells. To assess for myotube formation, we used immunofluorescence staining and western blot analysis in uMDSCs and L6 cells. RESULTS The results indicate that uMDSCs could form myotubes. Myotube formation was significantly increased by the Li-ESWT as was the expression of muscle heavy chain (MHC) and myogenic factor 5 (Myf5) in L6 cells in vitro. Li-ESWT activated protein kinase RNA-like ER kinase (PERK) pathway by increasing the phosphorylation levels of PERK and eukaryotic initiation factor 2a (eIF2α) and by increasing activating transcription factor 4 (ATF4). In addition, GSK2656157, an inhibitor of PERK, effectively inhibited the myotube formation in L6 rat myoblast cells. Furthermore, GSK2656157 also attenuated myotube formation induced by Li-ESWT. CONCLUSION In conclusion, this experiment reveals that rat uMDSCs can be isolated successfully and can form myotubes in vitro. PERK/ATF4 pathway was involved in myotube formation, and L6 rat myoblast cells were activated by Li-ESWT to form myotubes. These findings suggest that PERK/ATF4 pathway is activated by Li-ESWT. This study elucidates one of the biochemical pathways responsible for the clinical improvements seen after Li-ESWT. It is possible that this information will help to establish Li-ESWT as an acceptable treatment modality and may help to further refine the use of Li-ESWT in the clinical practice of medicine.
Collapse
Affiliation(s)
- Bohan Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California.,Department of Urology, The Second Hospital, Zhejiang University, Hangzhou, China
| | - Jun Zhou
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Amanda B Reed-Maldonado
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Hongxiu Ning
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Zhihua Lu
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Yajun Ruan
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Tie Zhou
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Hsun Shuan Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Byung Seok Oh
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Stanley Lei Qi
- Department of Bioengineering, Stanford University, Stanford, California
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| |
Collapse
|
43
|
Yu SJ, Kim HJ, Lee ES, Park CG, Cho SJ, Jeon SH. β-Catenin Accumulation Is Associated With Increased Expression of Nanog Protein and Predicts Maintenance of MSC Self-Renewal. Cell Transplant 2017; 26:365-377. [PMID: 27684957 PMCID: PMC5657765 DOI: 10.3727/096368916x693040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 12/07/2016] [Indexed: 01/01/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are self-renewing cells with the ability to differentiate into organized, functional network of cells. Recent studies have revealed that activation of the Wnt/β-catenin pathway by a glycogen synthase kinase (GSK)-3-specific pharmacological inhibitor, Bio, results in the maintenance of self-renewal in both mouse and human ES cells. The molecular mechanism behind the maintenance of hMSCs by these factors, however, is not fully understood. We found that rEGF enhances the level of β-catenin, a component of the Wnt/β-catenin signaling pathway. Furthermore, it was found that β-catenin upregulates Nanog. EGF activates the β-catenin pathway via the Ras protein and also increased the Nanog protein and gene expression levels 2 h after rEGF treatment. These results suggest that adding EGF can enhance β-catenin and Nanog expression in MSCs and facilitate EGF-mediated maintenance of MSC self-renewal. EGF was shown to augment MSC proliferation while preserving early progenitors within MSC population and thus did not induce differentiation. Thus, EGF not only can be used to expand MSC in vitro but also be utilized to autologous transplantation of MSCs in vivo.
Collapse
Affiliation(s)
- Sang-Jin Yu
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
- FOS Clinic, SM Tower (3rd Floor), 334 Gangnam-Daero, Gangnam-Gu, Seoul 135-936, Republic of Korea
| | - Hyun-Je Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Eui Seok Lee
- Department of Oral and Maxillofacial Surgery, College of Medicine, Korea University, Guro Hospital, Seoul, Republic of Korea
| | - Chung-Gyu Park
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute for Endemic Disease, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Translational Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Su Jin Cho
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Republic of Korea
| | - Soung-Hoo Jeon
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute for Endemic Disease, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Translational Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
44
|
Furrer R, Handschin C. Optimized Engagement of Macrophages and Satellite Cells in the Repair and Regeneration of Exercised Muscle. RESEARCH AND PERSPECTIVES IN ENDOCRINE INTERACTIONS 2017. [DOI: 10.1007/978-3-319-72790-5_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Abreu P, Mendes SVD, Ceccatto VM, Hirabara SM. Satellite cell activation induced by aerobic muscle adaptation in response to endurance exercise in humans and rodents. Life Sci 2016; 170:33-40. [PMID: 27888112 DOI: 10.1016/j.lfs.2016.11.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/31/2016] [Accepted: 11/21/2016] [Indexed: 11/15/2022]
Abstract
Although the requirement of satellite cells activation and expansion following injury, mechanical load or growth stimulus provoked by resistance exercise has been well established, their function in response to aerobic exercise adaptation remains unclear. A clear relationship between satellite cell expansion in fiber-type specific myosin heavy chain and aerobic performance has been related, independent of myonuclear accretion or muscle growth. However, the trigger for this activation process is not fully understood yet and it seems to be a multi-faceted and well-orchestrated process. Emerging in vitro studies suggest a role for metabolic pathways and oxygen availability for satellite cell activation, modulating the self-renewal potential and cell fate control. The goal of this review is to describe and discuss the current knowledge about the satellite cell activation and expansion in response to aerobic exercise adaptation in human and rodent models. Additionally, findings about the in vitro metabolic control, which seems be involved in the satellite cell activation and cell fate control, are presented and discussed.
Collapse
Affiliation(s)
- Phablo Abreu
- Institute of Biomedical Sciences, University of Sao Paulo, SP, Brazil; Institute of Biomedical Sciences, State University of Ceará, CE, Brazil.
| | | | | | - Sandro Massao Hirabara
- Institute of Biomedical Sciences, University of Sao Paulo, SP, Brazil; Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, Sao Paulo, SP, Brazil
| |
Collapse
|
46
|
Nakanishi R, Hirayama Y, Tanaka M, Maeshige N, Kondo H, Ishihara A, Roy RR, Fujino H. Nucleoprotein supplementation enhances the recovery of rat soleus mass with reloading after hindlimb unloading-induced atrophy via myonuclei accretion and increased protein synthesis. Nutr Res 2016; 36:1335-1344. [PMID: 27866827 DOI: 10.1016/j.nutres.2016.10.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 10/06/2016] [Accepted: 10/20/2016] [Indexed: 01/08/2023]
Abstract
Hindlimb unloading results in muscle atrophy and a period of reloading has been shown to partially recover the lost muscle mass. Two of the mechanisms involved in this recovery of muscle mass are the activation of protein synthesis pathways and an increase in myonuclei number. The additional myonuclei are provided by satellite cells that are activated by the mechanical stress associated with the reloading of the muscles and eventually incorporated into the muscle fibers. Amino acid supplementation with exercise also can increase skeletal muscle mass through enhancement of protein synthesis and nucleotide supplements can promote cell cycle activity. Therefore, we hypothesized that nucleoprotein supplementation, a combination of amino acids and nucleotides, would enhance the recovery of muscle mass to a greater extent than reloading alone after a period of unloading. Adult rats were assigned to 4 groups: control, hindlimb unloaded (HU; 14 days), reloaded (5 days) after hindlimb unloading (HUR), and reloaded after hindlimb unloading with nucleoprotein supplementation (HUR + NP). Compared with the HUR group, the HUR + NP group had larger soleus muscles and fiber cross-sectional areas, higher levels of phosphorylated rpS6, and higher numbers of myonuclei and myogenin-positive cells. These results suggest that nucleoprotein supplementation has a synergistic effect with reloading in recovering skeletal muscle properties after a period of unloading via rpS6 activation and satellite cell differentiation and incorporation into the muscle fibers. Therefore, this supplement may be an effective therapeutic regimen to include in rehabilitative strategies for a variety of muscle wasting conditions such as aging, cancer cachexia, muscular dystrophy, bed rest, and cast immobilization.
Collapse
Affiliation(s)
- Ryosuke Nakanishi
- Rehabilitation Science, Graduate School of Health Sciences, Kobe University, 7-10-2 Tomogaoka, Kobe 654-0142, Japan
| | - Yusuke Hirayama
- Rehabilitation Science, Graduate School of Health Sciences, Kobe University, 7-10-2 Tomogaoka, Kobe 654-0142, Japan
| | - Minoru Tanaka
- Rehabilitation Science, Graduate School of Health Sciences, Kobe University, 7-10-2 Tomogaoka, Kobe 654-0142, Japan; Department of Physical Therapy, Osaka Yukioka College of Health Science, 1-1-41 Soujiji, Ibaraki 567-0801, Japan
| | - Noriaki Maeshige
- Rehabilitation Science, Graduate School of Health Sciences, Kobe University, 7-10-2 Tomogaoka, Kobe 654-0142, Japan
| | - Hiroyo Kondo
- Department of Food Science and Nutrition, Nagoya Women's University, 3-40 Shiojicho, Nagoya 467-8611, Japan
| | - Akihiko Ishihara
- Laboratory of Cell Biology and Life Science, Graduate School of Human and Environmental Studies, Kyoto University, Yoshida-nihonmatsucho, Kyoto 606-8501, Japan
| | - Roland R Roy
- Brain Research Institute and Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095-7239, USA
| | - Hidemi Fujino
- Rehabilitation Science, Graduate School of Health Sciences, Kobe University, 7-10-2 Tomogaoka, Kobe 654-0142, Japan.
| |
Collapse
|
47
|
Segalés J, Perdiguero E, Muñoz-Cánoves P. Regulation of Muscle Stem Cell Functions: A Focus on the p38 MAPK Signaling Pathway. Front Cell Dev Biol 2016; 4:91. [PMID: 27626031 PMCID: PMC5003838 DOI: 10.3389/fcell.2016.00091] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/17/2016] [Indexed: 12/17/2022] Open
Abstract
Formation of skeletal muscle fibers (myogenesis) during development and after tissue injury in the adult constitutes an excellent paradigm to investigate the mechanisms whereby environmental cues control gene expression programs in muscle stem cells (satellite cells) by acting on transcriptional and epigenetic effectors. Here we will review the molecular mechanisms implicated in the transition of satellite cells throughout the distinct myogenic stages (i.e., activation from quiescence, proliferation, differentiation, and self-renewal). We will also discuss recent findings on the causes underlying satellite cell functional decline with aging. In particular, our review will focus on the epigenetic changes underlying fate decisions and on how the p38 MAPK signaling pathway integrates the environmental signals at the chromatin to build up satellite cell adaptive responses during the process of muscle regeneration, and how these responses are altered in aging. A better comprehension of the signaling pathways connecting external and intrinsic factors will illuminate the path for improving muscle regeneration in the aged.
Collapse
Affiliation(s)
- Jessica Segalés
- Cell Biology Group, Department of Experimental and Health Sciences, CIBER on Neurodegenerative diseases (CIBERNED), Pompeu Fabra University Barcelona, Spain
| | - Eusebio Perdiguero
- Cell Biology Group, Department of Experimental and Health Sciences, CIBER on Neurodegenerative diseases (CIBERNED), Pompeu Fabra University Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Cell Biology Group, Department of Experimental and Health Sciences, CIBER on Neurodegenerative diseases (CIBERNED), Pompeu Fabra UniversityBarcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA)Barcelona, Spain; Tissue Regeneration Laboratory, Centro Nacional de Investigaciones CardiovascularesMadrid, Spain
| |
Collapse
|
48
|
Sharples AP, Stewart CE, Seaborne RA. Does skeletal muscle have an 'epi'-memory? The role of epigenetics in nutritional programming, metabolic disease, aging and exercise. Aging Cell 2016; 15:603-16. [PMID: 27102569 PMCID: PMC4933662 DOI: 10.1111/acel.12486] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2016] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle mass, quality and adaptability are fundamental in promoting muscle performance, maintaining metabolic function and supporting longevity and healthspan. Skeletal muscle is programmable and can ‘remember’ early‐life metabolic stimuli affecting its function in adult life. In this review, the authors pose the question as to whether skeletal muscle has an ‘epi’‐memory? Following an initial encounter with an environmental stimulus, we discuss the underlying molecular and epigenetic mechanisms enabling skeletal muscle to adapt, should it re‐encounter the stimulus in later life. We also define skeletal muscle memory and outline the scientific literature contributing to this field. Furthermore, we review the evidence for early‐life nutrient stress and low birth weight in animals and human cohort studies, respectively, and discuss the underlying molecular mechanisms culminating in skeletal muscle dysfunction, metabolic disease and loss of skeletal muscle mass across the lifespan. We also summarize and discuss studies that isolate muscle stem cells from different environmental niches in vivo (physically active, diabetic, cachectic, aged) and how they reportedly remember this environment once isolated in vitro. Finally, we will outline the molecular and epigenetic mechanisms underlying skeletal muscle memory and review the epigenetic regulation of exercise‐induced skeletal muscle adaptation, highlighting exercise interventions as suitable models to investigate skeletal muscle memory in humans. We believe that understanding the ‘epi’‐memory of skeletal muscle will enable the next generation of targeted therapies to promote muscle growth and reduce muscle loss to enable healthy aging.
Collapse
Affiliation(s)
- Adam P. Sharples
- Stem Cells, Ageing and Molecular Physiology (SCAMP) Research Unit Exercise Metabolism and Adaptation Research Group (EMARG) Research Institute for Sport and Exercise Sciences (RISES) Liverpool John Moores University Liverpool UK
| | - Claire E. Stewart
- Stem Cells, Ageing and Molecular Physiology (SCAMP) Research Unit Exercise Metabolism and Adaptation Research Group (EMARG) Research Institute for Sport and Exercise Sciences (RISES) Liverpool John Moores University Liverpool UK
| | - Robert A. Seaborne
- Stem Cells, Ageing and Molecular Physiology (SCAMP) Research Unit Exercise Metabolism and Adaptation Research Group (EMARG) Research Institute for Sport and Exercise Sciences (RISES) Liverpool John Moores University Liverpool UK
| |
Collapse
|
49
|
Fujimaki S, Wakabayashi T, Asashima M, Takemasa T, Kuwabara T. Treadmill running induces satellite cell activation in diabetic mice. Biochem Biophys Rep 2016; 8:6-13. [PMID: 28955935 PMCID: PMC5613654 DOI: 10.1016/j.bbrep.2016.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/01/2016] [Accepted: 07/08/2016] [Indexed: 11/19/2022] Open
Abstract
Skeletal muscle-derived stem cells, termed as satellite cells, play essential roles in regeneration after muscle injury in adult skeletal muscle. Diabetes mellitus (DM), one of the most common metabolic diseases, causes impairments of satellite cell function. However, the studies of the countermeasures for the DM-induced dysfunction of satellite cells have been poor. Here, we investigated the effects of chronic running exercise on satellite cell activation in diabetic mice focused on the molecular mechanism including Notch and Wnt signaling, which are contribute to the fate determination of satellite cells. Male C57BL/6 mice 4 weeks of age were injected with streptozotocin and were randomly divided into runner group and control group. Runner group mice were performed treadmill running for 4 weeks. DM attenuated satellite cell activation and the expressions of the components of Notch and Wnt signaling. However, chronic running resulted in activation of satellite cells in diabetic mice and salvaged the inactivity of Wnt signaling but not Notch signaling. Our results suggest that chronic running induces satellite cell activation via upregulation of Wnt signaling in diabetic as well as normal mice. Diabetes attenuates satellite cell activation. Diabetes downregulates the activities of Notch and Wnt signaling. Treadmill running activates satellite cells in diabetic mice. Treadmill running can salvage diabetes-induced downregulation of Wnt signaling.
Collapse
Affiliation(s)
- Shin Fujimaki
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
- Physical Education, Health and Sport Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8574, Japan
| | - Tamami Wakabayashi
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Makoto Asashima
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Tohru Takemasa
- Physical Education, Health and Sport Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8574, Japan
- Corresponding authors.
| | - Tomoko Kuwabara
- Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
- Corresponding authors.
| |
Collapse
|
50
|
Wakabayashi T, Hidaka R, Fujimaki S, Asashima M, Kuwabara T. Diabetes Impairs Wnt3 Protein-induced Neurogenesis in Olfactory Bulbs via Glutamate Transporter 1 Inhibition. J Biol Chem 2016; 291:15196-211. [PMID: 27226528 DOI: 10.1074/jbc.m115.672857] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Indexed: 12/18/2022] Open
Abstract
Diabetes is associated with impaired cognitive function. Streptozotocin (STZ)-induced diabetic rats exhibit a loss of neurogenesis and deficits in behavioral tasks involving spatial learning and memory; thus, impaired adult hippocampal neurogenesis may contribute to diabetes-associated cognitive deficits. Recent studies have demonstrated that adult neurogenesis generally occurs in the dentate gyrus of the hippocampus, the subventricular zone, and the olfactory bulbs (OB) and is defective in patients with diabetes. We hypothesized that OB neurogenesis and associated behaviors would be affected in diabetes. In this study, we show that inhibition of Wnt3-induced neurogenesis in the OB causes several behavioral deficits in STZ-induced diabetic rats, including impaired odor discrimination, cognitive dysfunction, and increased anxiety. Notably, the sodium- and chloride-dependent GABA transporters and excitatory amino acid transporters that localize to GABAergic and glutamatergic terminals decreased in the OB of diabetic rats. Moreover, GAT1 inhibitor administration also hindered Wnt3-induced neurogenesis in vitro Collectively, these data suggest that STZ-induced diabetes adversely affects OB neurogenesis via GABA and glutamate transporter systems, leading to functional impairments in olfactory performance.
Collapse
Affiliation(s)
- Tamami Wakabayashi
- From the Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565 and
| | - Ryo Hidaka
- From the Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565 and
| | - Shin Fujimaki
- From the Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565 and Physical Education, Health and Sport Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 303-8577, Japan
| | - Makoto Asashima
- From the Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565 and
| | - Tomoko Kuwabara
- From the Stem Cell Engineering Research Group, Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 5, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565 and
| |
Collapse
|