1
|
Skiles CM, Boyd G, Gouw A, Robbins E, Minchev K, Ryder J, Ploutz-Snyder L, Trappe TA, Trappe S. Myonuclear and satellite cell content of the vastus lateralis and soleus with 70 days of simulated microgravity and the NASA SPRINT exercise program. J Appl Physiol (1985) 2025; 138:195-202. [PMID: 39656504 DOI: 10.1152/japplphysiol.00468.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
We previously observed a range of whole muscle and individual slow and fast myofiber size responses (mean: +4 to -24%) in quadriceps (vastus lateralis) and triceps surae (soleus) muscles of individuals undergoing 70 days of simulated microgravity with or without the NASA SPRINT exercise countermeasures program. The purpose of the current investigation was to further explore, in these same individuals, the content of myonuclei and satellite cells, both of which are key regulators of skeletal muscle mass. Individuals completed 6° head-down-tilt bedrest (BR, n = 9), bedrest with resistance and aerobic exercise (BRE, n = 9), or bedrest with resistance and aerobic exercise and low-dose testosterone (BRE + T, n = 8). The number of myonuclei and satellite cells associated with each slow [myosin heavy chain (MHC) I] and fast (MHC IIa) myofiber in the vastus lateralis was not changed (P > 0.05) pre- to postbedrest within the BR, BRE, or BRE + T groups. Similarly, in the soleus, the number of myonuclei associated with each slow and fast myofiber, and the number of satellite cells associated with each slow myofiber were not changed (P > 0.05) pre- to postbedrest within the BR, BRE, or BRE + T groups. It appears that even with relatively large perturbations in muscle mass over a few months of simulated microgravity, or with partially or completely effective exercise countermeasures, human skeletal muscle tightly regulates the abundance of myonuclei and satellite cells. Thus, exercise countermeasures efficacy for skeletal muscle atrophy appears to be independent of myonuclei and satellite cell abundance.NEW & NOTEWORTHY This study showed that after 70 days of simulated microgravity, human skeletal muscle does not alter the number of nuclei or satellite cells associated with slow or fast myofibers in the two muscle groups most negatively influenced by microgravity exposure [i.e., quadriceps (vastus lateralis) and triceps surae (soleus)]. Furthermore, the efficacy of exercise countermeasures for maintaining the mass of these muscles does not appear to be related to the myocellular content of nuclei or satellite cells.
Collapse
Affiliation(s)
- Chad M Skiles
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Gerard Boyd
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Aaron Gouw
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Ethan Robbins
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Kiril Minchev
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Jeffrey Ryder
- Universities Space Research Association, NASA Johnson Space Center, Houston, Texas, United States
| | - Lori Ploutz-Snyder
- Universities Space Research Association, NASA Johnson Space Center, Houston, Texas, United States
| | - Todd A Trappe
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Scott Trappe
- Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| |
Collapse
|
2
|
Zhou P, Wang W, Li J, Zheng Z, Du X, Fu L, Li X. Identification of novel transcription factors regulated by H3K27 acetylation in myogenic differentiation of porcine skeletal muscle satellite cells. FASEB J 2024; 38:e70144. [PMID: 39535314 DOI: 10.1096/fj.202401285rr] [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: 06/06/2024] [Revised: 10/14/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
H3K27 acetylation (H3K27ac) is crucial in muscle development as it regulates gene expression. Dysregulation of H3K27ac level has been linked to muscle-related diseases such as Duchenne muscular dystrophy, yet the mechanisms through which H3K27ac influences myogenic differentiation are not fully understood. Here, we utilized the SGC-CBP30 drug, a CBP/p300 bromodomain inhibitor, to reduce H3K27ac level and investigated its effect on myogenic differentiation of porcine skeletal muscle satellite cells. The results demonstrated an increased H3K27ac level during normal muscle satellite cell differentiation. We found that the addition of SGC-CBP30 resulted in a reduced level of H3K27ac based on ATAC-seq and CUT&Tag data. Our analysis revealed that a cluster characterized by reduced levels of H3K27ac and increased levels of H3K27me3 was enriched with motifs corresponding to Bach2, MafK, and Fosl2 transcription factors. Furthermore, knockdown of Bach2, MafK, and Fosl2 produced a similar suppression effect on myogenic differentiation. Taken together, our study contributes to a better understanding of how H3K27ac influences myogenic differentiation.
Collapse
Affiliation(s)
- Peng Zhou
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wenxuan Wang
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Jingjin Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Zhuqing Zheng
- Institute of Agricultural Biotechnology, Jingchu University of Technology, Jingmen, Hubei, China
| | - Xiaoyong Du
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Liangliang Fu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xinyun Li
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education and Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
3
|
Hejbøl EK, Andkjær SW, Dybdal J, Klindt M, Möller S, Lambertsen KL, Schrøder HD, Frich LH. Supraspinatus Muscle Regeneration Following Rotator Cuff Tear: A Study of the Biomarkers Pax7, MyoD, and Myogenin. Int J Mol Sci 2024; 25:11742. [PMID: 39519294 PMCID: PMC11546449 DOI: 10.3390/ijms252111742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/23/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
The success of rotator cuff tendon repair relies on both tendon healing and muscle recovery. The objective of this descriptive study was to investigate the regenerative potential of the supraspinatus muscle in rotator cuff tear conditions by quantifying the expression of Pax7, MyoD, and myogenin, basic factors that regulate myogenesis. Muscle biopsies were collected from thirty-three patients aged 34 to 73 years who underwent surgery for a rotator cuff tear affecting the supraspinatus muscle. Among these patients, twenty-seven percent were women, and the age of the lesions ranged from 2 to 72 months post-initial trauma. Biopsies were harvested from the supraspinatus muscle at the end closest to the tendon, and control biopsies were harvested from the ipsilateral deltoid muscle. The densities of immunohistochemically stained Pax7+, MyoD+, and myogenin+ nuclei/mm2 were used to estimate the myogenic potential of the muscle. Adjustments were made for patient age and lesion age. We found increased density of MyoD+ and myogenin+ cells in supraspinatus muscles compared to deltoid muscles (p < 0.001 and p = 0.003, respectively). Regression analyses that combined the density of positive nuclei with patient age showed a continuous increase in Pax7 with age but also a reduction of MyoD and myogenin in older patients. When combined with lesion age, there was a decline in the density of all myogenic markers after an initial rise. Pax7 density continued to be higher in supraspinatus compared to the deltoid muscle, but the density of MyoD and myogenin terminally dropped to a density lower than in the deltoid. Our findings suggest that the supraspinatus muscle in tear conditions showed signs of initial activation of muscle regeneration. When compared to the unaffected deltoid muscle, an apparent reduction in capacity to progress to full muscle fiber maturity was also demonstrated. This pattern of inhibited myogenesis seemed to increase with both patient age and lesion age. Our results on muscle regenerative capacity indicate that younger patients with rotator cuff tears have better chances of muscle recovery and may benefit from early surgical reconstruction.
Collapse
Affiliation(s)
- Eva Kildall Hejbøl
- The Orthopedic Research Unit, Hospital Sønderjylland, Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark; (E.K.H.)
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark
| | - Stephanie Wej Andkjær
- The Orthopedic Research Unit, Hospital Sønderjylland, Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark; (E.K.H.)
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Julie Dybdal
- The Orthopedic Research Unit, Hospital Sønderjylland, Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark; (E.K.H.)
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Marie Klindt
- The Orthopedic Research Unit, Hospital Sønderjylland, Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark; (E.K.H.)
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| | - Sören Möller
- Open Patient Data Explorative Network, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
- BRIDGE, Brain Research Inter-Disciplinary Guided Excellence, University of Southern Denmark, 5230 Odense, Denmark
| | - Henrik Daa Schrøder
- Department of Pathology, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark
| | - Lars Henrik Frich
- The Orthopedic Research Unit, Hospital Sønderjylland, Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark; (E.K.H.)
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
- Department of Orthopedics, Odense University Hospital, 5000 Odense, Denmark
| |
Collapse
|
4
|
AMPK Phosphorylation Impacts Apoptosis in Differentiating Myoblasts Isolated from Atrophied Rat Soleus Muscle. Cells 2023; 12:cells12060920. [PMID: 36980261 PMCID: PMC10047078 DOI: 10.3390/cells12060920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/25/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023] Open
Abstract
Regrowth of atrophied myofibers depends on muscle satellite cells (SCs) that exist outside the plasma membrane. Muscle atrophy appears to result in reduced number of SCs due to apoptosis. Given reduced AMP-activated protein kinase (AMPK) activity during differentiation of primary myoblasts derived from atrophic muscle, we hypothesized that there may be a potential link between AMPK and susceptibility of differentiating myoblasts to apoptosis. The aim of this study was to estimate the effect of AMPK activation (via AICAR treatment) on apoptosis in differentiating myoblasts derived from atrophied rat soleus muscle. Thirty rats were randomly assigned to the following two groups: control (C, n = 10) and 7-day hindlimb suspension (HS, n = 20). Myoblasts derived from the soleus muscles of HS rats were divided into two parts: AICAR-treated cells and non-treated cells. Apoptotic processes were evaluated by using TUNEL assay, RT-PCR and WB. In differentiating myoblasts derived from the atrophied soleus, there was a significant decrease (p < 0.05) in AMPK and ACC phosphorylation in parallel with increased number of apoptotic nuclei and a significant upregulation of pro-apoptotic markers (caspase-3, -9, BAX, p53) compared to the cells derived from control muscles. AICAR treatment of atrophic muscle-derived myoblasts during differentiation prevented reductions in AMPK and ACC phosphorylation as well as maintained the number of apoptotic nuclei and the expression of pro-apoptotic markers at the control levels. Thus, the maintenance of AMPK activity can suppress enhanced apoptosis in differentiating myoblasts derived from atrophied rat soleus muscle.
Collapse
|
5
|
Henrot P, Blervaque L, Dupin I, Zysman M, Esteves P, Gouzi F, Hayot M, Pomiès P, Berger P. Cellular interplay in skeletal muscle regeneration and wasting: insights from animal models. J Cachexia Sarcopenia Muscle 2023; 14:745-757. [PMID: 36811134 PMCID: PMC10067506 DOI: 10.1002/jcsm.13103] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 02/24/2023] Open
Abstract
Skeletal muscle wasting, whether related to physiological ageing, muscle disuse or to an underlying chronic disease, is a key determinant to quality of life and mortality. However, cellular basis responsible for increased catabolism in myocytes often remains unclear. Although myocytes represent the vast majority of skeletal muscle cellular population, they are surrounded by numerous cells with various functions. Animal models, mostly rodents, can help to decipher the mechanisms behind this highly dynamic process, by allowing access to every muscle as well as time-course studies. Satellite cells (SCs) play a crucial role in muscle regeneration, within a niche also composed of fibroblasts and vascular and immune cells. Their proliferation and differentiation is altered in several models of muscle wasting such as cancer, chronic kidney disease or chronic obstructive pulmonary disease (COPD). Fibro-adipogenic progenitor cells are also responsible for functional muscle growth and repair and are associated in disease to muscle fibrosis such as in chronic kidney disease. Other cells have recently proven to have direct myogenic potential, such as pericytes. Outside their role in angiogenesis, endothelial cells and pericytes also participate to healthy muscle homoeostasis by promoting SC pool maintenance (so-called myogenesis-angiogenesis coupling). Their role in chronic diseases muscle wasting has been less studied. Immune cells are pivotal for muscle repair after injury: Macrophages undergo a transition from the M1 to the M2 state along with the transition between the inflammatory and resolutive phase of muscle repair. T regulatory lymphocytes promote and regulate this transition and are also able to activate SC proliferation and differentiation. Neural cells such as terminal Schwann cells, motor neurons and kranocytes are notably implicated in age-related sarcopenia. Last, newly identified cells in skeletal muscle, such as telocytes or interstitial tenocytes could play a role in tissular homoeostasis. We also put a special focus on cellular alterations occurring in COPD, a chronic and highly prevalent respiratory disease mainly linked to tobacco smoke exposure, where muscle wasting is strongly associated with increased mortality, and discuss the pros and cons of animal models versus human studies in this context. Finally, we discuss resident cells metabolism and present future promising leads for research, including the use of muscle organoids.
Collapse
Affiliation(s)
- Pauline Henrot
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France.,CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Léo Blervaque
- PhyMedExp, INSERM-CNRS-Montpellier University, Montpellier, France
| | - Isabelle Dupin
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France
| | - Maéva Zysman
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France.,CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Pauline Esteves
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France
| | - Fares Gouzi
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, Montpellier, France
| | - Maurice Hayot
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, Montpellier, France
| | - Pascal Pomiès
- PhyMedExp, INSERM-CNRS-Montpellier University, Montpellier, France
| | - Patrick Berger
- Centre de Recherche Cardio-thoracique de Bordeaux, Univ-Bordeaux, Pessac, France.,Centre de Recherche Cardio-thoracique de Bordeaux, INSERM, Pessac, France.,CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| |
Collapse
|
6
|
Bouredji Z, Argaw A, Frenette J. The inflammatory response, a mixed blessing for muscle homeostasis and plasticity. Front Physiol 2022; 13:1032450. [PMID: 36505042 PMCID: PMC9726740 DOI: 10.3389/fphys.2022.1032450] [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: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 11/24/2022] Open
Abstract
Skeletal muscle makes up almost half the body weight of heathy individuals and is involved in several vital functions, including breathing, thermogenesis, metabolism, and locomotion. Skeletal muscle exhibits enormous plasticity with its capacity to adapt to stimuli such as changes in mechanical loading, nutritional interventions, or environmental factors (oxidative stress, inflammation, and endocrine changes). Satellite cells and timely recruited inflammatory cells are key actors in muscle homeostasis, injury, and repair processes. Conversely, uncontrolled recruitment of inflammatory cells or chronic inflammatory processes leads to muscle atrophy, fibrosis and, ultimately, impairment of muscle function. Muscle atrophy and loss of function are reported to occur either in physiological situations such as aging, cast immobilization, and prolonged bed rest, as well as in many pathological situations, including cancers, muscular dystrophies, and several other chronic illnesses. In this review, we highlight recent discoveries with respect to the molecular mechanisms leading to muscle atrophy caused by modified mechanical loading, aging, and diseases. We also summarize current perspectives suggesting that the inflammatory process in muscle homeostasis and repair is a double-edged sword. Lastly, we review recent therapeutic approaches for treating muscle wasting disorders, with a focus on the RANK/RANKL/OPG pathway and its involvement in muscle inflammation, protection and regeneration processes.
Collapse
Affiliation(s)
- Zineb Bouredji
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CRCHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC, Canada
| | - Anteneh Argaw
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CRCHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC, Canada
| | - Jérôme Frenette
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l’Université Laval (CRCHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC, Canada,Département de Réadaptation, Faculté de Médecine, Université Laval, Quebec City, QC, Canada,*Correspondence: Jérôme Frenette,
| |
Collapse
|
7
|
Cultured Myoblasts Derived from Rat Soleus Muscle Show Altered Regulation of Proliferation and Myogenesis during the Course of Mechanical Unloading. Int J Mol Sci 2022; 23:ijms23169150. [PMID: 36012431 PMCID: PMC9409304 DOI: 10.3390/ijms23169150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/04/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
The structure and function of soleus muscle fibers undergo substantial remodeling under real or simulated microgravity conditions. However, unloading-induced changes in the functional activity of skeletal muscle primary myoblasts remain poorly studied. The purpose of our study was to investigate how short-term and long-term mechanical unloading would affect cultured myoblasts derived from rat soleus muscle. Mechanical unloading was simulated by rat hindlimb suspension model (HS). Myoblasts were purified from rat soleus at basal conditions and after 1, 3, 7, and 14 days of HS. Myoblasts were expanded in vitro, and the myogenic nature was confirmed by their ability to differentiate as well as by immunostaining/mRNA expression of myogenic markers. The proliferation activity at different time points after HS was analyzed, and transcriptome analysis was performed. We have shown that soleus-derived myoblasts differently respond to an early and later stage of HS. At the early stage of HS, the proliferative activity of myoblasts was slightly decreased, and processes related to myogenesis activation were downregulated. At the later stage of HS, we observed a decrease in myoblast proliferative potential and spontaneous upregulation of the pro-myogenic program.
Collapse
|
8
|
Abstract
Despite the evolutionary loss of tissue regenerative potential, robust skeletal muscle repair processes are largely retained even in higher vertebrates. In mammals, the skeletal muscle regeneration program is driven by resident stem cells termed satellite cells, guided by the coordinated activity of multiple intrinsic and extrinsic factors and other cell types. A thorough understanding of muscle repair mechanisms is crucial not only for combating skeletal myopathies, but for its prospective aid in devising therapeutic strategies to endow regenerative potential on otherwise regeneration-deficient organs. In this review, we discuss skeletal muscle regeneration from an evolutionary perspective, summarize the current knowledge of cellular and molecular mechanisms, and highlight novel paradigms of muscle repair revealed by explorations of the recent decade.
Collapse
Affiliation(s)
- Sajedah M Hindi
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| |
Collapse
|
9
|
Ravalli S, Federico C, Lauretta G, Saccone S, Pricoco E, Roggio F, Di Rosa M, Maugeri G, Musumeci G. Morphological Evidence of Telocytes in Skeletal Muscle Interstitium of Exercised and Sedentary Rodents. Biomedicines 2021; 9:biomedicines9070807. [PMID: 34356871 PMCID: PMC8301487 DOI: 10.3390/biomedicines9070807] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle atrophy, resulting from states of hypokinesis or immobilization, leads to morphological, metabolic, and functional changes within the muscle tissue, a large variety of which are supported by the stromal cells populating the interstitium. Telocytes represent a recently discovered population of stromal cells, which has been increasingly identified in several human organs and appears to participate in sustaining cross-talk, promoting regenerative mechanisms and supporting differentiation of local stem cell niche. The aim of this morphologic study was to investigate the presence of Telocytes in the tibialis anterior muscle of healthy rats undergoing an endurance training protocol for either 4 weeks or 16 weeks compared to sedentary rats. Histomorphometric analysis of muscle fibers diameter revealed muscle atrophy in sedentary rats. Telocytes were identified by double-positive immunofluorescence staining for CD34/CD117 and CD34/vimentin. The results showed that Telocytes were significantly reduced in sedentary rats at 16 weeks, while rats subjected to regular exercise maintained a stable Telocytes population after 16 weeks. Understanding of the relationship between Telocytes and exercise offers new chances in the field of regenerative medicine, suggesting possible triggers for Telocytes in sarcopenia and other musculoskeletal disorders, promoting adapted physical activity and rehabilitation programmes in clinical practice.
Collapse
Affiliation(s)
- Silvia Ravalli
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (S.R.); (G.L.); (E.P.); (F.R.); (M.D.R.); (G.M.)
| | - Concetta Federico
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology, University of Catania, Via Androne 81, 95124 Catania, Italy; (C.F.); (S.S.)
| | - Giovanni Lauretta
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (S.R.); (G.L.); (E.P.); (F.R.); (M.D.R.); (G.M.)
| | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology, University of Catania, Via Androne 81, 95124 Catania, Italy; (C.F.); (S.S.)
| | - Elisabetta Pricoco
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (S.R.); (G.L.); (E.P.); (F.R.); (M.D.R.); (G.M.)
| | - Federico Roggio
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (S.R.); (G.L.); (E.P.); (F.R.); (M.D.R.); (G.M.)
- Department of Psychology, Educational Science and Human Movement, University of Palermo, Via Giovanni Pascoli 6, 90144 Palermo, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (S.R.); (G.L.); (E.P.); (F.R.); (M.D.R.); (G.M.)
| | - Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (S.R.); (G.L.); (E.P.); (F.R.); (M.D.R.); (G.M.)
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human, Histology and Movement Science Section, University of Catania, Via S. Sofia 87, 95123 Catania, Italy; (S.R.); (G.L.); (E.P.); (F.R.); (M.D.R.); (G.M.)
- Research Center on Motor Activities (CRAM), University of Catania, Via S. Sofia 97, 95123 Catania, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Correspondence:
| |
Collapse
|
10
|
Vilchinskaya NA, Shenkman BS. Myosatellite Cells under Gravitational Unloading Conditions. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Papanikolaou K, Veskoukis AS, Draganidis D, Baloyiannis I, Deli CK, Poulios A, Jamurtas AZ, Fatouros IG. Redox-dependent regulation of satellite cells following aseptic muscle trauma: Implications for sports performance and nutrition. Free Radic Biol Med 2020; 161:125-138. [PMID: 33039652 DOI: 10.1016/j.freeradbiomed.2020.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/26/2020] [Accepted: 10/01/2020] [Indexed: 12/21/2022]
Abstract
Skeletal muscle satellite cells (SCs) are indispensable for tissue regeneration, remodeling and growth. Following myotrauma, SCs are activated, and assist in tissue repair. Exercise-induced muscle damage (EIMD) is characterized by a pronounced inflammatory response and the production of reactive oxygen species (ROS). Experimental evidence suggests that SCs kinetics (the propagation from a quiescent to an activated/proliferative state) following EIMD is redox-dependent and interconnected with changes in the SCs microenvironment (niche). Animal studies have shown that following aseptic myotrauma, antioxidant and/or anti-inflammatory supplementation leads to an improved recovery and skeletal muscle regeneration through enhanced SCs kinetics, suggesting a redox-dependent molecular mechanism. Although evidence suggests that antioxidant/anti-inflammatory compounds may prevent performance deterioration and enhance recovery, there is lack of information regarding the redox-dependent regulation of SCs responses following EIMD in humans. In this review, SCs kinetics following aseptic myotrauma, as well as the intrinsic redox-sensitive molecular mechanisms responsible for SCs responses are discussed. The role of redox status on SCs function should be further investigated in the future with human clinical trials in an attempt to elucidate the molecular pathways responsible for muscle recovery and provide information for potential nutritional strategies aiming at performance recovery.
Collapse
Affiliation(s)
- Konstantinos Papanikolaou
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Aristidis S Veskoukis
- Department of Nutrition and Dietetics, University of Thessaly, Argonafton 1, 42132, Trikala, Greece; Department of Biochemistry and Biotechnology, University of Thessaly, Viopolis, Mezourlo, 41500, Larissa, Greece
| | - Dimitrios Draganidis
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Ioannis Baloyiannis
- Department of Surgery, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Chariklia K Deli
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Athanasios Poulios
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Athanasios Z Jamurtas
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece
| | - Ioannis G Fatouros
- Department of Physical Education and Sport Science, University of Thessaly, Karies, Trikala, 42132, Greece.
| |
Collapse
|
12
|
Arneson-Wissink PC, Hogan KA, Ducharme AM, Samani A, Jatoi A, Doles JD. The wasting-associated metabolite succinate disrupts myogenesis and impairs skeletal muscle regeneration. JCSM RAPID COMMUNICATIONS 2020; 3:56-69. [PMID: 32905522 PMCID: PMC7470228 DOI: 10.1002/rco2.14] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND Muscle wasting is a debilitating co-morbidity affecting most advanced cancer patients. Alongside enhanced muscle catabolism, defects in muscle repair/regeneration contribute to cancer-associated wasting. Among the factors implicated in suppression of muscle regeneration are cytokines that interfere with myogenic signal transduction pathways. Less understood is how other cancer/wasting-associated cues, such as metabolites, contribute to muscle dysfunction. This study investigates how the metabolite succinate affects myogenesis and muscle regeneration. METHODS We leveraged an established ectopic metabolite treatment (cell permeable dimethyl-succinate) strategy to evaluate the ability of intracellular succinate elevation to 1) affect myoblast homeostasis (proliferation, apoptosis), 2) disrupt protein dynamics and induce wasting-associated atrophy, and 3) modulate in vitro myogenesis. In vivo succinate supplementation experiments (2% succinate, 1% sucrose vehicle) were used to corroborate and extend in vitro observations. Metabolic profiling and functional metabolic studies were then performed to investigate the impact of succinate elevation on mitochondria function. RESULTS We found that in vitro succinate supplementation elevated intracellular succinate about 2-fold, and did not have an impact on proliferation or apoptosis of C2C12 myoblasts. Elevated succinate had minor effects on protein homeostasis (~25% decrease in protein synthesis assessed by OPP staining), and no significant effect on myotube atrophy. Succinate elevation interfered with in vitro myoblast differentiation, characterized by significant decreases in late markers of myogenesis and fewer nuclei per myosin heavy chain positive structure (assessed by immunofluorescence staining). While mice orally administered succinate did not exhibit changes in overall body composition or whole muscle weights, these mice displayed smaller muscle myofiber diameters (~6% decrease in the mean of non-linear regression curves fit to the histograms of minimum feret diameter distribution), which was exacerbated when muscle regeneration was induced with barium chloride injury. Significant decreases in the mean of non-linear regression curves fit to the histograms of minimum feret diameter distributions were observed 7 days and 28 days post injury. Elevated numbers of myogenin positive cells (3-fold increase) supportive of the differentiation defects observed in vitro were observed 28 days post injury. Metabolic profiling and functional metabolic assessment of myoblasts revealed that succinate elevation caused both widespread metabolic changes and significantly lowered maximal cellular respiration (~35% decrease). CONCLUSIONS This study broadens the repertoire of wasting-associated factors that can directly modulate muscle progenitor cell function and strengthens the hypothesis that metabolic derangements are significant contributors to impaired muscle regeneration, an important aspect of cancer-associated muscle wasting.
Collapse
Affiliation(s)
- Paige C Arneson-Wissink
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Kelly A Hogan
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Alexandra M Ducharme
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Adrienne Samani
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
| | - Aminah Jatoi
- Department of Oncology, Mayo Clinic, Rochester,
Minnesota
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo
Clinic, Rochester, Minnesota, 55905 USA
- Corresponding Author: Jason D Doles, Department of
Biochemistry and Molecular Biology, Mayo Clinic, 200 First St SW, Guggenheim
16-11A1, Rochester, MN 55905, Tel: (507) 284-9372, Fax: (507) 284-3383,
| |
Collapse
|
13
|
Stem Cell Aging in Skeletal Muscle Regeneration and Disease. Int J Mol Sci 2020; 21:ijms21051830. [PMID: 32155842 PMCID: PMC7084237 DOI: 10.3390/ijms21051830] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/04/2020] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle comprises 30-40% of the weight of a healthy human body and is required for voluntary movements in humans. Mature skeletal muscle is formed by multinuclear cells, which are called myofibers. Formation of myofibers depends on the proliferation, differentiation, and fusion of muscle progenitor cells during development and after injury. Muscle progenitor cells are derived from muscle satellite (stem) cells (MuSCs), which reside on the surface of the myofiber but beneath the basement membrane. MuSCs play a central role in postnatal maintenance, growth, repair, and regeneration of skeletal muscle. In sedentary adult muscle, MuSCs are mitotically quiescent, but are promptly activated in response to muscle injury. Physiological and chronological aging induces MuSC aging, leading to an impaired regenerative capability. Importantly, in pathological situations, repetitive muscle injury induces early impairment of MuSCs due to stem cell aging and leads to early impairment of regeneration ability. In this review, we discuss (1) the role of MuSCs in muscle regeneration, (2) stem cell aging under physiological and pathological conditions, and (3) prospects related to clinical applications of controlling MuSCs.
Collapse
|
14
|
Buratti P, Covatti C, Centenaro LA, Brancalhão RMC, Torrejais MM. Morphofunctional characteristics of skeletal muscle in rats with cerebral palsy. Int J Exp Pathol 2019; 100:49-59. [PMID: 30773727 DOI: 10.1111/iep.12304] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 10/24/2018] [Accepted: 12/27/2018] [Indexed: 01/10/2023] Open
Abstract
Knowledge of skeletal muscle adaptations is important to understand the functional deficits in cerebral palsy (CP). This study aimed to investigate the morphofunctional characteristics of skeletal muscle in a CP animal model. Initially, pregnant Wistar rats were injected intraperitoneally with saline or lipopolysaccharide over the last five days of pregnancy. The control group (n = 8) consisted of male pups born to females injected with saline. The CP group (n = 8) consisted of male pups born to females injected with lipopolysaccharide, which were submitted to perinatal anoxia [day of birth, postnatal day 0 (P0)] and sensorimotor restriction (P1-P30). The open-field test was undertaken on P29 and P45. On P48, the animals were weighed, and the plantaris muscle was collected and its weight and length were measured. Transverse sections were stained with haematoxylin-eosin, NADH-TR, Masson's trichrome and non-specific esterase reaction for analysis. and transmission electron microscopy was performed. In the CP group, reductions were observed in mobility time, number of crossings and rearing frequency, body weight, muscle weight and length, and nucleus-to-fibre and capillary-to-fibre ratios. There was a statistically significant increase in the percentage area of the muscle section occupied by collagen; reduction in the area and increase in the number of type I muscle fibres; increase in myofibrillar disorganization and Z-line disorganization and dissolution; and reduction in the area and largest and smallest diameters of neuromuscular junctions. Thus this animal model of CP produced morphofunctional alterations in skeletal muscle, that were associated with evidence of motor deficits as demonstrated by the open-field test.
Collapse
Affiliation(s)
- Pâmela Buratti
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| | - Caroline Covatti
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| | - Lígia Aline Centenaro
- Centro de Ciências Médicas e Farmacêuticas, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| | - Rose Meire Costa Brancalhão
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| | - Marcia Miranda Torrejais
- Programa de Pós-Graduação em Biociências e Saúde, Universidade Estadual do Oeste do Paraná - UNIOESTE, Cascavel, Paraná, Brazil
| |
Collapse
|
15
|
Kneppers A, Leermakers P, Pansters N, Backx E, Gosker H, van Loon L, Schols A, Langen R, Verdijk L. Coordinated regulation of skeletal muscle mass and metabolic plasticity during recovery from disuse. FASEB J 2018; 33:1288-1298. [PMID: 30133324 DOI: 10.1096/fj.201701403rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Skeletal muscle regeneration after disuse is essential for muscle maintenance and involves the regulation of both mass- and metabolic plasticity-related processes. However, the relation between these processes during recovery from disuse remains unclear. In this study, we explored the potential interrelationship between the molecular regulation of muscle mass and oxidative metabolism during recovery from disuse. Molecular profiles were measured in biopsies from the vastus lateralis of healthy men after 1-leg cast immobilization and after 1 wk reloading, and in mouse gastrocnemius obtained before and after hindlimb suspension and during reloading (RL-1, -2, -3, -5, and -8 d). Cluster analysis of the human recovery response revealed correlations between myogenesis and autophagy markers in 2 clusters, which were distinguished by the presence of markers of early myogenesis, autophagosome formation, and mitochondrial turnover vs. markers of late myogenesis, autophagy initiation, and mitochondrial mass. In line with these findings, an early transient increase in B-cell lymphoma-2 interacting protein-3 and sequestosome-1 protein, and GABA type A receptor-associated protein like-1 protein and mRNA and a late increase in myomaker and myosin heavy chain-8 mRNA, microtubule-associated protein 1 light chain 3-II:I ratio, and FUN14 domain-containing-1 mRNA and protein were observed in mice. In summary, the regulatory profiles of protein, mitochondrial, and myonuclear turnover are correlated and temporally associated, suggesting a coordinated regulation of muscle mass- and oxidative metabolism-related processes during recovery from disuse.-Kneppers, A., Leermakers, P., Pansters, N., Backx, E., Gosker, H., van Loon, L., Schols, A., Langen, R., Verdijk, L. Coordinated regulation of skeletal muscle mass and metabolic plasticity during recovery from disuse.
Collapse
Affiliation(s)
- Anita Kneppers
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands; and
| | - Pieter Leermakers
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands; and
| | - Nicholas Pansters
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands; and
| | - Evelien Backx
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Harry Gosker
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands; and
| | - Luc van Loon
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Annemie Schols
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands; and
| | - Ramon Langen
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands; and
| | - Lex Verdijk
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
16
|
Fukada SI. The roles of muscle stem cells in muscle injury, atrophy and hypertrophy. J Biochem 2018; 163:353-358. [PMID: 29394360 DOI: 10.1093/jb/mvy019] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/29/2017] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle is composed of multinuclear cells called myofibers. Muscular dystrophy (a genetic muscle disorder) induces instability in the cell membrane of myofibers and eventually causes myofibre damage. Non-genetic muscle disorders, including sarcopenia, diabetes, bedridden immobility and cancer cachexia, lead to atrophy of myofibres. In contrast, resistance training induces myofibre hypertrophy. Thus, myofibres exhibit a plasticity that is strongly affected by both intrinsic and extrinsic factors. There is no doubt that muscle stem cells (MuSCs, also known as muscle satellite cells) are indispensable for muscle repair/regeneration, but their contributions to atrophy and hypertrophy are still controversial. The present review focuses on the relevance of MuSCs to (i) muscle diseases and (ii) hypertrophy. Further, this review addresses fundamental questions about MuSCs to clarify the onset or progression of these diseases and which might lead to development of a MuSC-based therapy.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita City, Osaka 565-0871, Japan
| |
Collapse
|
17
|
Guitart M, Lloreta J, Mañas‐Garcia L, Barreiro E. Muscle regeneration potential and satellite cell activation profile during recovery following hindlimb immobilization in mice. J Cell Physiol 2018; 233:4360-4372. [DOI: 10.1002/jcp.26282] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 11/13/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Maria Guitart
- Department of Pulmonology‐Muscle Wasting Cachexia in Chronic Respiratory Diseases Lung Cancer Research GroupIMIM‐Hospital del Mar Parc de Salut Mar, Health and Sciences Experimental Department (CEXS)Universitat Pompeu Fabra (UPF)Barcelona Biomedical Research Park (PRBB)BarcelonaSpain
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES)Instituto de Salud Carlos III (ISCIII)BarcelonaSpain
| | - Josep Lloreta
- Department of PathologyHospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS)Universitat Pompeu Fabra (UPF)Barcelona Biomedical Research Park (PRBB)BarcelonaSpain
| | - Laura Mañas‐Garcia
- Department of Pulmonology‐Muscle Wasting Cachexia in Chronic Respiratory Diseases Lung Cancer Research GroupIMIM‐Hospital del Mar Parc de Salut Mar, Health and Sciences Experimental Department (CEXS)Universitat Pompeu Fabra (UPF)Barcelona Biomedical Research Park (PRBB)BarcelonaSpain
| | - Esther Barreiro
- Department of Pulmonology‐Muscle Wasting Cachexia in Chronic Respiratory Diseases Lung Cancer Research GroupIMIM‐Hospital del Mar Parc de Salut Mar, Health and Sciences Experimental Department (CEXS)Universitat Pompeu Fabra (UPF)Barcelona Biomedical Research Park (PRBB)BarcelonaSpain
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES)Instituto de Salud Carlos III (ISCIII)BarcelonaSpain
| |
Collapse
|
18
|
Mashinchian O, Pisconti A, Le Moal E, Bentzinger CF. The Muscle Stem Cell Niche in Health and Disease. Curr Top Dev Biol 2017; 126:23-65. [PMID: 29305000 DOI: 10.1016/bs.ctdb.2017.08.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Nestlé Institute of Health Sciences, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Doctoral Program in Biotechnology and Bioengineering, Lausanne, Switzerland
| | - Addolorata Pisconti
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emmeran Le Moal
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - C Florian Bentzinger
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
19
|
Yousuf Y, Jeschke MG, Shah A, Sadri AR, Datu AK, Samei P, Amini-Nik S. The response of muscle progenitor cells to cutaneous thermal injury. Stem Cell Res Ther 2017; 8:234. [PMID: 29041952 PMCID: PMC5646146 DOI: 10.1186/s13287-017-0686-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/02/2017] [Accepted: 10/02/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Severe burn results in a systemic response that leads to significant muscle wasting. It is believed that this rapid loss in muscle mass occurs due to increased protein degradation combined with reduced protein synthesis. Alterations in the microenvironment of muscle progenitor cells may partially account for this pathology. The aim of this study was to ascertain the response of muscle progenitor cells following thermal injury in mice and to enlighten the cellular cascades that contribute to the muscle wasting. METHODS C57BL/6 mice received a 20% total body surface area (TBSA) thermal injury. Gastrocnemius muscle was harvested at days 2, 7, and 14 following injury for protein and histological analysis. RESULTS We observed a decrease in myofiber cross-sectional area at 2 days post-burn. This muscle atrophy was compensated for by an increase in myofiber cross-sectional area at 7 and 14 days post-burn. Myeloperoxidase (MPO)-positive cells (neutrophils) increased significantly at 2 days. Moreover, through Western blot analysis of two key mediators of the proteolytic pathway, we show there is an increase in Murf1 and NF-κB 2 days post-burn. MPO-positive cells were also positive for NF-κB, suggesting that neutrophils attain NF-κB activity in the muscle. Unlike inflammatory and proteolytic pathways, the number of Pax7-positive muscle progenitor cells decreased significantly 2 days post-burn. This was followed by a recovery in the number of Pax7-positive cells at 7 and 14 days, suggesting proliferation of muscle progenitors that accompanied regrowth. CONCLUSION Our data show a biphasic response in the muscles of mice exposed to burn injury, with phenotypic characteristics of muscle atrophy at 2 days while compensation was observed later with a change in Pax7-positive muscle progenitor cells. Targeting muscle progenitors may be of therapeutic benefit in muscle wasting observed after burn injury.
Collapse
Affiliation(s)
- Yusef Yousuf
- Institute of Medicine Science, University of Toronto, Toronto, Canada.,Sunnybrook Research Institute, Sunnybrook's Trauma, Emergency & Critical Care (TECC) Program, Ross Tilley Burn Centre, M7-161, Lab: M7-140, 2075 Bayview Ave., Toronto, ON, M4N 3 M5, Canada
| | - Marc G Jeschke
- Institute of Medicine Science, University of Toronto, Toronto, Canada.,Sunnybrook Research Institute, Sunnybrook's Trauma, Emergency & Critical Care (TECC) Program, Ross Tilley Burn Centre, M7-161, Lab: M7-140, 2075 Bayview Ave., Toronto, ON, M4N 3 M5, Canada.,Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada.,Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Ahmed Shah
- Sunnybrook Research Institute, Sunnybrook's Trauma, Emergency & Critical Care (TECC) Program, Ross Tilley Burn Centre, M7-161, Lab: M7-140, 2075 Bayview Ave., Toronto, ON, M4N 3 M5, Canada
| | - Ali-Reza Sadri
- Institute of Medicine Science, University of Toronto, Toronto, Canada.,Sunnybrook Research Institute, Sunnybrook's Trauma, Emergency & Critical Care (TECC) Program, Ross Tilley Burn Centre, M7-161, Lab: M7-140, 2075 Bayview Ave., Toronto, ON, M4N 3 M5, Canada
| | - Andrea-Kaye Datu
- Sunnybrook Research Institute, Sunnybrook's Trauma, Emergency & Critical Care (TECC) Program, Ross Tilley Burn Centre, M7-161, Lab: M7-140, 2075 Bayview Ave., Toronto, ON, M4N 3 M5, Canada
| | - Pantea Samei
- Sunnybrook Research Institute, Sunnybrook's Trauma, Emergency & Critical Care (TECC) Program, Ross Tilley Burn Centre, M7-161, Lab: M7-140, 2075 Bayview Ave., Toronto, ON, M4N 3 M5, Canada
| | - Saeid Amini-Nik
- Sunnybrook Research Institute, Sunnybrook's Trauma, Emergency & Critical Care (TECC) Program, Ross Tilley Burn Centre, M7-161, Lab: M7-140, 2075 Bayview Ave., Toronto, ON, M4N 3 M5, Canada. .,Laboratory in Medicine and Pathobiology, University of Toronto, Toronto, Canada. .,Department of Surgery, Division of Plastic Surgery, University of Toronto, Toronto, Canada.
| |
Collapse
|
20
|
de Sousa HA, da Silva MDG, Barbosa KDP, Vianna LMDS, Pacheco YG, de Godoy JRP, Kuckelhaus SAS. Electrical stimulation structurally affects the tissues of the rectum and anus of nulliparous rats. J Anat 2017; 231:398-404. [PMID: 28547814 PMCID: PMC5554829 DOI: 10.1111/joa.12635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2017] [Indexed: 12/20/2022] Open
Abstract
Considering that the muscles of the anus perform a critical role in maintaining continence, losses in their structure can negatively affect the physiological control of the intestinal contents. Anorectal electro-stimulation (ARES) has been reported to have a positive effect on the functionality of treated patients, but how ARES affects the structural tissues of the anorectal segment remains unknown. Because the study of how ARES structurally affects human tissues is not possible, this study aimed to clarify these effects in a murine model, which has a similar anorectal segment (structure and physiology) to humans. For the descriptive and comparative study, randomly selected nulliparous adult Wistar rats (n = 5) were submitted to 30 anorectal sessions of ARES with a biphasic current (700 μs, 50 Hz from 2 to 4 mA). After treatment, the animals were euthanized, and the anorectal segments were dissected and processed for histopathological analysis. Our results showed that ARES increased the widths of the mucosal, submucosal and muscle layers of the rectum, as well as the number of leukocytes in the mucosa. ARES also caused hyperplasia of the smooth muscle of the internal anal sphincter and hypertrophy of the external anal sphincter muscle. In conclusion, our results showed that ARES had not only a positive effect on the structure (morphology) of all tissues associated with the rectum and anus but, more importantly, on the structural gain of the muscles (hyperplasia and hypertrophy), which could point to a functional gain of the anal sphincter, reinforcing the applicability of ARES as a non-invasive treatment for anal incontinence.
Collapse
Affiliation(s)
- Hugo Alves de Sousa
- Laboratory of Molecular Anatomy and NanomedicineArea of MorphologyFaculty of MedicineUniversity of BrasiliaBrasiliaFederal DistrictBrazil
| | - Maria da Glória da Silva
- Laboratory of HistologyArea of MorphologyFaculty of MedicineUniversity of BrasiliaBrasiliaFederal DistrictBrazil
| | | | | | - Yolanda Galindo Pacheco
- Laboratory of Molecular Anatomy and NanomedicineArea of MorphologyFaculty of MedicineUniversity of BrasiliaBrasiliaFederal DistrictBrazil
| | - José Roberto Pimenta de Godoy
- Laboratory of Molecular Anatomy and NanomedicineArea of MorphologyFaculty of MedicineUniversity of BrasiliaBrasiliaFederal DistrictBrazil
| | - Selma Aparecida Souza Kuckelhaus
- Laboratory of Molecular Anatomy and NanomedicineArea of MorphologyFaculty of MedicineUniversity of BrasiliaBrasiliaFederal DistrictBrazil
- Laboratory of Cell ImmunologyArea of PathologyFaculty of MedicineUniversity of BrasiliaBrasiliaFederal DistrictBrazil
| |
Collapse
|
21
|
Lima ARR, Pagan LU, Damatto RL, Cezar MDM, Bonomo C, Gomes MJ, Martinez PF, Guizoni DM, Campos DHS, Damatto FC, Okoshi K, Okoshi MP. Effects of growth hormone on cardiac remodeling and soleus muscle in rats with aortic stenosis-induced heart failure. Oncotarget 2017; 8:83009-83021. [PMID: 29137319 PMCID: PMC5669945 DOI: 10.18632/oncotarget.20583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/29/2017] [Indexed: 12/18/2022] Open
Abstract
Background Skeletal muscle wasting is often observed in heart failure (HF). The growth hormone (GH)/insulin-like growth factor-1 (IGF-1) axis is impaired in HF. In this study, we evaluated the effects of GH on soleus muscle and cardiac remodeling in rats with aortic stenosis (AS)-induced HF. Methods AS was created by placing a stainless-steel clip on the ascending aorta. After clinically detecting HF, GH (2 mg/kg/day) was subcutaneously injected for 14 days (AS-GH group). Results were compared with those from Sham and non-treated AS groups. Transthoracic echocardiogram was performed before and after treatment. Protein expression was evaluated by Western blot and satellite cells activation by immunofluorescence. Statistical analyzes: ANOVA and Tukey or Kruskal-Wallis and Student-Newman-Keuls. Results Before treatment both AS groups presented a similar degree of cardiac injury. GH prevented body weight loss and attenuated systolic dysfunction. Soleus cross-sectional fiber areas were lower in both AS groups than Sham (Sham 3,556±447; AS 2,882±422; AS-GH 2,868±591 μm2; p=0.016). GH increased IGF-1 serum concentration (Sham 938±83; AS 866±116; AS-GH 1167±166 ng/mL; p<0.0001) and IGF-1 muscle protein expression and activated PI3K protein. Neural cell adhesion molecule (NCAM) immunofluorescence was increased in both AS groups. Catabolism-related intracellular pathways did not differ between groups. Conclusion Short-term growth hormone attenuates left ventricular systolic dysfunction in rats with aortic stenosis-induced HF. Despite preserving body weight, increasing serum and muscular IGF-1 levels, and stimulating PI3K muscle expression, GH does not modulate soleus muscle trophism, satellite cells activation or intracellular pathways associated with muscle catabolism.
Collapse
Affiliation(s)
- Aline R R Lima
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Luana U Pagan
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Ricardo L Damatto
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Marcelo D M Cezar
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Camila Bonomo
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Mariana J Gomes
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Paula F Martinez
- School of Physical Therapy, Federal University of Mato Grosso do Sul, Campo Grande, Brazil
| | - Daniele M Guizoni
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Dijon H S Campos
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Felipe C Damatto
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Katashi Okoshi
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| | - Marina P Okoshi
- Botucatu Medical School, Internal Medicine Departament, Sao Paulo State University, UNESP, Botucatu, Brazil
| |
Collapse
|
22
|
Takase F, Inui A, Mifune Y, Sakata R, Muto T, Harada Y, Ueda Y, Kokubu T, Kurosaka M. Effect of platelet-rich plasma on degeneration change of rotator cuff muscles: In vitro and in vivo evaluations. J Orthop Res 2017; 35:1806-1815. [PMID: 27684960 DOI: 10.1002/jor.23451] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 09/23/2016] [Indexed: 02/04/2023]
Abstract
Atrophy with fatty degeneration is often seen in rotator cuff muscles with torn tendons. PRP has been reported to enhance tissue repair processes after tendon ruptures. However, the effect of PRP on atrophy and fatty degeneration of the muscle is not yet known. The aim of this study is to examine the effect of PRP on degeneration change of rotator cuff muscles in vitro and in vivo. A murine myogenic cell line and a rat rotator cuff tear model were used in this study and PRP was administrated into subacromial space which is widely used in clinical practice. In in vitro study, administration of PRP to C2C12 cells stimulated cell proliferation while inhibited both myogenic and adipogenic differentiation. In in vivo study, administration of PRP suppressed Oil Red-O positive lipid droplet formation. The expression of adipogenic genes was also decreased by PRP administration. In conclusion, PRP promoted proliferation of myoblast cells, while inhibiting adipogenic differentiation of myoblast cells and suppressing fatty degeneration change in rat torn rotator cuff muscles. Further investigations are needed to determine the clinical applicability of the PRP. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1806-1815, 2017.
Collapse
Affiliation(s)
- Fumiaki Takase
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Atsuyuki Inui
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yutaka Mifune
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Ryosuke Sakata
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tomoyuki Muto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yoshifumi Harada
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yasuhiro Ueda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takeshi Kokubu
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masahiro Kurosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
23
|
Li TS, Shi H, Wang L, Yan CZ. Effect of Bone Marrow Mesenchymal Stem Cells on Satellite Cell Proliferation and Apoptosis in Immobilization-Induced Muscle Atrophy in Rats. Med Sci Monit 2016; 22:4651-4660. [PMID: 27898654 PMCID: PMC5132424 DOI: 10.12659/msm.898137] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/11/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Muscle atrophy due to disuse occurs along with adverse physiological and functional changes, but bone marrow stromal cells (MSCs) may be able to act as muscle satellite cells to restore myofibers. Thus, we investigated whether MSCs could enhance the proliferation of satellite cells and suppress myonuclear apoptosis during immobilization. MATERIAL AND METHODS We isolated, purified, amplified, and identified MSCs. Rats (n=48) were randomized into 3 groups: WB group (n=16), IM-PBS group (n=16), and IM-MSC (n=16). Rat hind limbs were immobilized for 14 d, treated with MSCs or phosphate-buffered saline (PBS), and then studied using immunohistochemistry and Western blot analysis to characterize the proteins involved. Apoptosis was assessed by terminal deoxynucleotidyl transferase (TdT)-mediated deoxy-UTP nick end labeling (TUNEL) method. RESULTS We compared muscle mass, cross-sectional areas, and peak tetanic forces and noted insignificant differences between PBS- and MSC-treated animals, but satellite cell proliferation was significantly greater after MSC treatment (p<0.05). Apoptotic myonuclei were reduced (p<0.05) after MSC treatment as well. Pro-apoptotic Bax was down-regulated and anti-apoptotic Bcl-2 and p-Akt protein were upregulated (p<0.05). CONCLUSIONS MSCs injected during hind limb immobilization can maintain satellite cell activity by suppressing myonuclear apoptosis.
Collapse
Affiliation(s)
- Tie-Shan Li
- Department of Neurology and Neuromuscular Center, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
- Department of Rehabilitation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Hao Shi
- Shandong Rehabilitation Research Center, Jinan, Shandong, P.R. China
| | - Lin Wang
- Department of Rehabilitation, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, P.R. China
| | - Chuan-Zhu Yan
- Department of Neurology and Neuromuscular Center, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
- Brain Science Research Institute, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
24
|
Randolph ME, Phillips BL, Choo HJ, Vest KE, Vera Y, Pavlath GK. Pharyngeal Satellite Cells Undergo Myogenesis Under Basal Conditions and Are Required for Pharyngeal Muscle Maintenance. Stem Cells 2016; 33:3581-95. [PMID: 26178867 DOI: 10.1002/stem.2098] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/15/2015] [Accepted: 06/01/2015] [Indexed: 12/19/2022]
Abstract
The pharyngeal muscles of the nasal, oral, and laryngeal pharynxes are required for swallowing. Pharyngeal muscles are preferentially affected in some muscular dystrophies yet spared in others. Muscle stem cells, called satellite cells, may be critical factors in the development of pharyngeal muscle disorders; however, very little is known about pharyngeal satellite cells (PSC) and their role in pharyngeal muscles. We show that PSC are distinct from the commonly studied hindlimb satellite cells both transcriptionally and biologically. Under basal conditions PSC proliferate, progress through myogenesis, and fuse with pharyngeal myofibers. Furthermore, PSC exhibit biologic differences dependent on anatomic location in the pharynx. Importantly, PSC are required to maintain myofiber size and myonuclear number in pharyngeal myofibers. Together, these results demonstrate that PSC are critical for pharyngeal muscle maintenance and suggest that satellite cell impairment could contribute to pharyngeal muscle pathology associated with various muscular dystrophies and aging.
Collapse
Affiliation(s)
| | | | - Hyo-Jung Choo
- Department of Pharmacology, Emory University, Atlanta, Georgia, USA
| | - Katherine E Vest
- Department of Pharmacology, Emory University, Atlanta, Georgia, USA
| | - Yandery Vera
- Department of Pharmacology, Emory University, Atlanta, Georgia, USA
| | - Grace K Pavlath
- Department of Pharmacology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
25
|
Yoshida T, Delafontaine P. An Intronic Enhancer Element Regulates Angiotensin II Type 2 Receptor Expression during Satellite Cell Differentiation, and Its Activity Is Suppressed in Congestive Heart Failure. J Biol Chem 2016; 291:25578-25590. [PMID: 27756842 DOI: 10.1074/jbc.m116.752501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/17/2016] [Indexed: 12/20/2022] Open
Abstract
Patients with advanced congestive heart failure (CHF) or chronic kidney disease often have increased angiotensin II (Ang II) levels and cachexia. We previously demonstrated that Ang II, via its type 1 receptor, causes muscle protein breakdown and apoptosis and inhibits satellite cell (SC) proliferation and muscle regeneration, likely contributing to cachexia in CHF and chronic kidney disease. In contrast, Ang II, via its type 2 receptor (AT2R) expression, is robustly induced during SC differentiation, and it potentiates muscle regeneration. To understand the mechanisms regulating AT2R expression and its potential role in muscle regeneration in chronic diseases, we used a mouse model of CHF and found that muscle regeneration was markedly reduced and that this was accompanied by blunted increase of AT2R expression. We performed AT2R promoter reporter analysis during satellite cell differentiation and found that the 70 bp upstream of the AT2R transcription start site contain a core promoter region, and regions upstream of 70 bp to 3 kbp are dispensable for AT2R induction. Instead, AT2R intron 2 acts as a transcriptional enhancer during SC differentiation. Further deletion/mutation analysis revealed that multiple transcription factor binding sites in the +286/+690 region within intron 2 coordinately regulate AT2R transcription. Importantly, +286/+690 enhancer activity was suppressed in CHF mouse skeletal muscle, suggesting that AT2R expression is suppressed in CHF via inhibition of AT2R intronic enhancer activity, leading to lowered muscle regeneration. Thus targeting intron 2 enhancer element could lead to the development of a novel intervention to increase AT2R expression in SCs and potentiate skeletal muscle regenerative capacity in chronic diseases.
Collapse
Affiliation(s)
- Tadashi Yoshida
- From the Department of Medicine and Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri 65212
| | - Patrice Delafontaine
- From the Department of Medicine and Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri 65212
| |
Collapse
|
26
|
Esteves de Lima J, Bonnin MA, Birchmeier C, Duprez D. Muscle contraction is required to maintain the pool of muscle progenitors via YAP and NOTCH during fetal myogenesis. eLife 2016; 5. [PMID: 27554485 PMCID: PMC5030091 DOI: 10.7554/elife.15593] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 08/23/2016] [Indexed: 12/27/2022] Open
Abstract
The importance of mechanical activity in the regulation of muscle progenitors during chick development has not been investigated. We show that immobilization decreases NOTCH activity and mimics a NOTCH loss-of-function phenotype, a reduction in the number of muscle progenitors and increased differentiation. Ligand-induced NOTCH activation prevents the reduction of muscle progenitors and the increase of differentiation upon immobilization. Inhibition of NOTCH ligand activity in muscle fibers suffices to reduce the progenitor pool. Furthermore, immobilization reduces the activity of the transcriptional co-activator YAP and the expression of the NOTCH ligand JAG2 in muscle fibers. YAP forced-activity in muscle fibers prevents the decrease of JAG2 expression and the number of PAX7+ cells in immobilization conditions. Our results identify a novel mechanism acting downstream of muscle contraction, where YAP activates JAG2 expression in muscle fibers, which in turn regulates the pool of fetal muscle progenitors via NOTCH in a non-cell-autonomous manner. DOI:http://dx.doi.org/10.7554/eLife.15593.001 Skeletal muscle is attached to the skeleton and allows the body to move. Making a new muscle, or repairing an existing one, relies on stem cells that are present inside muscles. A major goal of skeletal muscle research is to understand the signals that regulate the abilities of muscle stem cells to divide and give rise to more stem cells or to become muscle cells. Molecular signals are known to regulate the numbers of stem cells in the muscle. Skeletal muscles become larger if they are exercised, but it is not clear if mechanical forces generated by muscle contractions directly affect the number of muscle stem cells. The NOTCH signaling pathway contributes to maintaining the population of stem cells in muscles by forcing the stem cells to divide and preventing them from becoming muscle cells. Here, Esteves de Lima et al. investigated whether muscle contraction regulates NOTCH signaling during muscle formation in chick fetuses. The experiments show that muscle contraction stimulates the activity of a protein called YAP in muscle cells, which in turn, activates a gene in the NOTCH signaling pathway known as JAG2. This increases NOTCH signaling activity in the neighboring stem cells and maintains the number of stem cells in the muscle. The next step following this work will be to establish if this mechanism also operates during muscle formation and regeneration in other animals such as mice and zebrafish. DOI:http://dx.doi.org/10.7554/eLife.15593.002
Collapse
Affiliation(s)
- Joana Esteves de Lima
- CNRS UMR 7622, F-75005 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, F-75005, Paris, France
| | - Marie-Ange Bonnin
- CNRS UMR 7622, F-75005 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, F-75005, Paris, France
| | - Carmen Birchmeier
- Developmental Biology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Delphine Duprez
- CNRS UMR 7622, F-75005 Paris, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,IBPS-Developmental Biology Laboratory, Paris, France.,Inserm U1156, F-75005, Paris, France
| |
Collapse
|
27
|
Haralampieva D, Betzel T, Dinulovic I, Salemi S, Stoelting M, Krämer SD, Schibli R, Sulser T, Handschin C, Eberli D, Ametamey SM. Noninvasive PET Imaging and Tracking of Engineered Human Muscle Precursor Cells for Skeletal Muscle Tissue Engineering. J Nucl Med 2016; 57:1467-73. [PMID: 27199355 DOI: 10.2967/jnumed.115.170548] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 02/08/2016] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Transplantation of human muscle precursor cells (hMPCs) is envisioned for the treatment of various muscle diseases. However, a feasible noninvasive tool to monitor cell survival, migration, and integration into the host tissue is still missing. METHODS In this study, we designed an adenoviral delivery system to genetically modify hMPCs to express a signaling-deficient form of human dopamine D2 receptor (hD2R). The gene expression levels of the receptor were evaluated by reverse transcriptase polymerase chain reaction, and infection efficiency was evaluated by fluorescent microscopy. The viability, proliferation, and differentiation capacity of the transduced cells, as well as their myogenic phenotype, were determined by flow cytometry analysis and fluorescent microscopy. (18)F-fallypride and (18)F-fluoromisonidazole, two well-established PET radioligands, were assessed for their potential to image engineered hMPCs in a mouse model and their uptakes were evaluated at different time points after cell inoculation in vivo. Biodistribution studies, autoradiography, and PET experiments were performed to determine the extent of signal specificity. To address feasibility for tracking hMPCs in an in vivo model, the safety of the adenoviral gene delivery was evaluated. Finally, the harvested tissues were histologically examined to determine whether survival of the transplanted cells was sustained at different time points. RESULTS Adenoviral gene delivery was shown to be safe, with no detrimental effects on the primary human cells. The viability, proliferation, and differentiation capacity of the transduced cells were confirmed, and flow cytometry analysis and fluorescent microscopy showed that their myogenic phenotype was sustained. (18)F-fallypride and (18)F-fluoromisonidazole were successfully synthesized. Specific binding of (18)F-fallypride to hD2R hMPCs was demonstrated in vitro and in vivo. Furthermore, the (18)F-fluoromisonidazole signal was high at the early stages. Finally, sustained survival of the transplanted cells at different time points was confirmed histologically, with formation of muscle tissue at the site of injection. CONCLUSION Our proposed use of a signaling-deficient hD2R as a potent reporter for in vivo hMPC PET tracking by (18)F-fallypride is a significant step toward potential noninvasive tracking of hD2R hMPCs and bioengineered muscle tissues in the clinic.
Collapse
Affiliation(s)
- Deana Haralampieva
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland Zurich Center for Integrative Human Physiology, Zurich, Switzerland; and
| | - Thomas Betzel
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Ivana Dinulovic
- Focal Area of Growth and Development, Biozentrum, University of Basel, Basel, Switzerland
| | - Souzan Salemi
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Meline Stoelting
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stefanie D Krämer
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Roger Schibli
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Tullio Sulser
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Christoph Handschin
- Focal Area of Growth and Development, Biozentrum, University of Basel, Basel, Switzerland
| | - Daniel Eberli
- Laboratory for Tissue Engineering and Stem Cell Therapy, Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland Zurich Center for Integrative Human Physiology, Zurich, Switzerland; and
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland Zurich Center for Integrative Human Physiology, Zurich, Switzerland; and
| |
Collapse
|
28
|
Arentson-Lantz EJ, English KL, Paddon-Jones D, Fry CS. Fourteen days of bed rest induces a decline in satellite cell content and robust atrophy of skeletal muscle fibers in middle-aged adults. J Appl Physiol (1985) 2016; 120:965-75. [PMID: 26796754 DOI: 10.1152/japplphysiol.00799.2015] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 01/20/2016] [Indexed: 01/06/2023] Open
Abstract
Bed rest, a ground-based spaceflight analog, induces robust atrophy of skeletal muscle, an effect that is exacerbated with increasing age. We examined the effect of 14 days of bed rest on skeletal muscle satellite cell content and fiber type atrophy in middle-aged adults, an understudied age demographic with few overt signs of muscle aging that is representative of astronauts who perform long-duration spaceflight. Muscle biopsies were obtained from the vastus lateralis of healthy middle-aged adults [n= 7 (4 male, 3 female); age: 51 ± 1 yr] before (Pre-BR) and after (Post-BR) 14 days of bed rest. Immunohistochemical analyses were used to quantify myosin heavy chain (MyHC) isoform expression, cross-sectional area (CSA), satellite cell and myonuclear content, and capillary density. Peak oxygen consumption, knee extensor strength, and body composition were also measured Pre-BR and Post-BR. Post-BR MyHC type 2a fiber percentage was reduced, and mean CSA decreased in all fiber types (-24 ± 5%;P< 0.05). Satellite cell content was also reduced Post-BR (-39 ± 9%;P< 0.05), and the change in satellite cell content was significantly correlated with the change in mean fiber CSA (r(2)= 0.60;P< 0.05). A decline in capillary density was observed Post-BR (-23 ± 6%;P< 0.05), and Post-BR capillary content was significantly associated with Post-BR peak aerobic capacity (r(2)= 0.59;P< 0.05). A subtle decline in myonuclear content occurred during bed rest (-5 ± 1%;P< 0.05). The rapid maladaptation of skeletal muscle to 14 days of mechanical unloading in middle-aged adults emphasizes the need for robust countermeasures to preserve muscle function in astronauts.
Collapse
Affiliation(s)
- Emily J Arentson-Lantz
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Kirk L English
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Douglas Paddon-Jones
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas; Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas
| | - Christopher S Fry
- Department of Nutrition and Metabolism, University of Texas Medical Branch, Galveston, Texas; Division of Rehabilitation Sciences, University of Texas Medical Branch, Galveston, Texas; Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
29
|
Optimizing Electrical Stimulation for Promoting Satellite Cell Proliferation in Muscle Disuse Atrophy. Am J Phys Med Rehabil 2016; 95:28-38. [DOI: 10.1097/phm.0000000000000307] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
30
|
Expression of CCAAT/Enhancer Binding Protein Beta in Muscle Satellite Cells Inhibits Myogenesis in Cancer Cachexia. PLoS One 2015; 10:e0145583. [PMID: 26709824 PMCID: PMC4692409 DOI: 10.1371/journal.pone.0145583] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/04/2015] [Indexed: 01/09/2023] Open
Abstract
Cancer cachexia is a paraneoplastic syndrome that causes profound weight loss and muscle mass atrophy and is estimated to be the cause of up to 30% of cancer deaths. Though the exact cause is unknown, patients with cancer cachexia have increased muscle protein catabolism. In healthy muscle, injury activates skeletal muscle stem cells, called satellite cells, to differentiate and promote regeneration. Here, we provide evidence that this mechanism is inhibited in cancer cachexia due to persistent expression of CCAAT/Enhancer Binding Protein beta (C/EBPβ) in muscle myoblasts. C/EBPβ is a bzip transcription factor that is expressed in muscle satellite cells and is normally downregulated upon differentiation. However, in myoblasts exposed to a cachectic milieu, C/EBPβ expression remains elevated, despite activation to differentiate, resulting in the inhibition of myogenin expression and myogenesis. In vivo, cancer cachexia results in increased number of Pax7+ cells that also express C/EBPβ and the inhibition of normal repair mechanisms. Loss of C/EBPβ expression in primary myoblasts rescues differentiation under cachectic conditions without restoring myotube size, indicating that C/EBPβ is an important inhibitor of myogenesis in cancer cachexia.
Collapse
|
31
|
Functional Overload Enhances Satellite Cell Properties in Skeletal Muscle. Stem Cells Int 2015; 2016:7619418. [PMID: 26779264 PMCID: PMC4686724 DOI: 10.1155/2016/7619418] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/29/2015] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle represents a plentiful and accessible source of adult stem cells. Skeletal-muscle-derived stem cells, termed satellite cells, play essential roles in postnatal growth, maintenance, repair, and regeneration of skeletal muscle. Although it is well known that the number of satellite cells increases following physical exercise, functional alterations in satellite cells such as proliferative capacity and differentiation efficiency following exercise and their molecular mechanisms remain unclear. Here, we found that functional overload, which is widely used to model resistance exercise, causes skeletal muscle hypertrophy and converts satellite cells from quiescent state to activated state. Our analysis showed that functional overload induces the expression of MyoD in satellite cells and enhances the proliferative capacity and differentiation potential of these cells. The changes in satellite cell properties coincided with the inactivation of Notch signaling and the activation of Wnt signaling and likely involve modulation by transcription factors of the Sox family. These results indicate the effects of resistance exercise on the regulation of satellite cells and provide insight into the molecular mechanism of satellite cell activation following physical exercise.
Collapse
|
32
|
Talbert EE, Guttridge DC. Impaired regeneration: A role for the muscle microenvironment in cancer cachexia. Semin Cell Dev Biol 2015; 54:82-91. [PMID: 26385617 DOI: 10.1016/j.semcdb.2015.09.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 09/11/2015] [Indexed: 12/17/2022]
Abstract
While changes in muscle protein synthesis and degradation have long been known to contribute to muscle wasting, a body of literature has arisen which suggests that regulation of the satellite cell and its ensuing regenerative program are impaired in atrophied muscle. Lessons learned from cancer cachexia suggest that this regulation is simply not a consequence, but a contributing factor to the wasting process. In addition to satellite cells, evidence from mouse models of cancer cachexia also suggests that non-satellite progenitor cells from the muscle microenvironment are also involved. This chapter in the series reviews the evidence of dysfunctional muscle repair in multiple wasting conditions. Potential mechanisms for this dysfunctional regeneration are discussed, particularly in the context of cancer cachexia.
Collapse
Affiliation(s)
- Erin E Talbert
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, and the Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Denis C Guttridge
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, and the Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
33
|
Biressi S, Gopinath SD. The quasi-parallel lives of satellite cells and atrophying muscle. Front Aging Neurosci 2015; 7:140. [PMID: 26257645 PMCID: PMC4510774 DOI: 10.3389/fnagi.2015.00140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/06/2015] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle atrophy or wasting accompanies various chronic illnesses and the aging process, thereby reducing muscle function. One of the most important components contributing to effective muscle repair in postnatal organisms, the satellite cells (SCs), have recently become the focus of several studies examining factors participating in the atrophic process. We critically examine here the experimental evidence linking SC function with muscle loss in connection with various diseases as well as aging, and in the subsequent recovery process. Several recent reports have investigated the changes in SCs in terms of their differentiation and proliferative capacity in response to various atrophic stimuli. In this regard, we review the molecular changes within SCs that contribute to their dysfunctional status in atrophy, with the intention of shedding light on novel potential pharmacological targets to counteract the loss of muscle mass.
Collapse
Affiliation(s)
- Stefano Biressi
- Dulbecco Telethon Institute and Centre for Integrative Biology (CIBIO), University of TrentoTrento, Italy
| | | |
Collapse
|
34
|
Meyer GA, Farris AL, Sato E, Gibbons M, Lane JG, Ward SR, Engler AJ. Muscle progenitor cell regenerative capacity in the torn rotator cuff. J Orthop Res 2015; 33:421-9. [PMID: 25410765 PMCID: PMC4346515 DOI: 10.1002/jor.22786] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/06/2014] [Indexed: 02/04/2023]
Abstract
Chronic rotator cuff (RC) tears affect a large portion of the population and result in substantial upper extremity impairment, shoulder weakness, pain, and limited range of motion. Regardless of surgical or conservative treatment, persistent atrophic muscle changes limit functional restoration and may contribute to surgical failure. We hypothesized that deficits in the skeletal muscle progenitor (SMP) cell pool could contribute to poor muscle recovery following tendon repair. Biopsies were obtained from patients undergoing arthroscopic RC surgery. The SMP population was quantified, isolated, and assayed in culture for its ability to proliferate and fuse in vitro and in vivo. The SMP population was larger in muscles from cuffs with partial tears compared with no tears or full thickness tears. However, SMPs from muscles in the partial tear group also exhibited reduced proliferative ability. Cells from all cuff states were able to fuse robustly in culture and engraft when injected into injured mouse muscle, suggesting that when given the correct signals, SMPs are capable of contributing to muscle hypertrophy and regeneration regardless of tear severity. The fact that this does not appear to happen in vivo helps focus future therapeutic targets for promoting muscle recovery following rotator cuff repairs and may help improve clinical outcomes.
Collapse
Affiliation(s)
- Gretchen A. Meyer
- Investigation performed at the Department of Bioengineering, University of California, San Diego; La Jolla, CA 92093
| | - Ashley L. Farris
- Investigation performed at the Department of Bioengineering, University of California, San Diego; La Jolla, CA 92093
| | - Eugene Sato
- Investigation performed at the Department of Bioengineering, University of California, San Diego; La Jolla, CA 92093
| | - Michael Gibbons
- Investigation performed at the Department of Bioengineering, University of California, San Diego; La Jolla, CA 92093
| | - John G. Lane
- Investigation performed at the Department of Orthopaedic Surgery, University of California, San Diego; La Jolla, CA 92093
| | - Samuel R. Ward
- Investigation performed at the Department of Orthopaedic Surgery, University of California, San Diego; La Jolla, CA 92093,Investigation performed at the Department of Radiology, University of California, San Diego; La Jolla, CA 92093,Corresponding Authors: Dr. Adam J. Engler, 9500 Gilman Drive / MC 0695, La Jolla, CA 92093, Tel: 858-246-0678 Fax: 858-246-1469, , Dr. Samuel R. Ward, 9500 Gilman Drive / MC 0610, La Jolla, CA 92093, Tel: 858-534-4918 Fax: 858-822-3807,
| | - Adam J. Engler
- Investigation performed at the Department of Bioengineering, University of California, San Diego; La Jolla, CA 92093,Sanford Consortium for Regenerative Medicine; La Jolla, CA 92037,Corresponding Authors: Dr. Adam J. Engler, 9500 Gilman Drive / MC 0695, La Jolla, CA 92093, Tel: 858-246-0678 Fax: 858-246-1469, , Dr. Samuel R. Ward, 9500 Gilman Drive / MC 0610, La Jolla, CA 92093, Tel: 858-534-4918 Fax: 858-822-3807,
| |
Collapse
|
35
|
Abstract
Muscle regeneration recapitulates many aspects of embryonic myogenesis and is an important homeostatic process of the adult skeletal muscle, which, after development, retains the capacity to regenerate in response to appropriate stimuli, activating the muscle compartment of stem cells, namely, satellite cells, as well as other precursor cells. Moreover, significant evidence suggests that while stem cells represent an important determinant for tissue regeneration, a “qualified” environment is necessary to guarantee and achieve functional results. It is therefore plausible that the loss of control over these cell fate decisions could lead to a pathological transdifferentiation, leading to pathologic defects in the regenerative process. This review provides an overview about the general aspects of muscle development and discusses the cellular and molecular aspects that characterize the five interrelated and time-dependent phases of muscle regeneration, namely, degeneration, inflammation, regeneration, remodeling, and maturation/functional repair.
Collapse
|
36
|
Brooks NE, Myburgh KH. Skeletal muscle wasting with disuse atrophy is multi-dimensional: the response and interaction of myonuclei, satellite cells and signaling pathways. Front Physiol 2014; 5:99. [PMID: 24672488 PMCID: PMC3955994 DOI: 10.3389/fphys.2014.00099] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/27/2014] [Indexed: 12/25/2022] Open
Abstract
Maintenance of skeletal muscle is essential for health and survival. There are marked losses of skeletal muscle mass as well as strength and physiological function under conditions of low mechanical load, such as space flight, as well as ground based models such as bed rest, immobilization, disuse, and various animal models. Disuse atrophy is caused by mechanical unloading of muscle and this leads to reduced muscle mass without fiber attrition. Skeletal muscle stem cells (satellite cells) and myonuclei are integrally involved in skeletal muscle responses to environmental changes that induce atrophy. Myonuclear domain size is influenced differently in fast and slow twitch muscle, but also by different models of muscle wasting, a factor that is not yet understood. Although the myonuclear domain is 3-dimensional this is rarely considered. Apoptosis as a mechanism for myonuclear loss with atrophy is controversial, whereas cell death of satellite cells has not been considered. Molecular signals such as myostatin/SMAD pathway, MAFbx, and MuRF1 E3 ligases of the ubiquitin proteasome pathway and IGF1-AKT-mTOR pathway are 3 distinctly different contributors to skeletal muscle protein adaptation to disuse. Molecular signaling pathways activated in muscle fibers by disuse are rarely considered within satellite cells themselves despite similar exposure to unloading or low mechanical load. These molecular pathways interact with each other during atrophy and also when various interventions are applied that could alleviate atrophy. Re-applying mechanical load is an obvious method to restore muscle mass, however how nutrient supplementation (e.g., amino acids) may further enhance recovery (or reduce atrophy despite unloading or ageing) is currently of great interest. Satellite cells are particularly responsive to myostatin and to growth factors. Recently, the hibernating squirrel has been identified as an innovative model to study resistance to atrophy.
Collapse
Affiliation(s)
- Naomi E Brooks
- Health and Exercise Science Research Group, School of Sport, University of Stirling Stirling, UK
| | - Kathryn H Myburgh
- Muscle Research Group, Department of Physiological Sciences, Stellenbosch University Stellenbosch, South Africa
| |
Collapse
|
37
|
Microvesicles containing miRNAs promote muscle cell death in cancer cachexia via TLR7. Proc Natl Acad Sci U S A 2014; 111:4525-9. [PMID: 24616506 DOI: 10.1073/pnas.1402714111] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression and, in cancers, are often packaged within secreted microvesicles. The cachexia syndrome is a debilitating state of cancer that predominantly results from the loss of skeletal muscle mass, which is in part associated with apoptosis. How tumors promote apoptosis in distally located skeletal muscles has not been explored. Using both tumor cell lines and patient samples, we show that tumor-derived microvesicles induce apoptosis of skeletal muscle cells. This proapoptotic activity is mediated by a microRNA cargo, miR-21, which signals through the Toll-like 7 receptor (TLR7) on murine myoblasts to promote cell death. Furthermore, tumor microvesicles and miR-21 require c-Jun N-terminal kinase activity to regulate this apoptotic response. Together, these results describe a unique pathway by which tumor cells promote muscle loss, which might provide a great insight into elucidating the causes and treatment options of cancer cachexia.
Collapse
|
38
|
He WA, Berardi E, Cardillo VM, Acharyya S, Aulino P, Thomas-Ahner J, Wang J, Bloomston M, Muscarella P, Nau P, Shah N, Butchbach MER, Ladner K, Adamo S, Rudnicki MA, Keller C, Coletti D, Montanaro F, Guttridge DC. NF-κB-mediated Pax7 dysregulation in the muscle microenvironment promotes cancer cachexia. J Clin Invest 2014; 123:4821-35. [PMID: 24084740 DOI: 10.1172/jci68523] [Citation(s) in RCA: 264] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 08/06/2013] [Indexed: 01/09/2023] Open
Abstract
Cachexia is a debilitating condition characterized by extreme skeletal muscle wasting that contributes significantly to morbidity and mortality. Efforts to elucidate the underlying mechanisms of muscle loss have predominantly focused on events intrinsic to the myofiber. In contrast, less regard has been given to potential contributory factors outside the fiber within the muscle microenvironment. In tumor-bearing mice and patients with pancreatic cancer, we found that cachexia was associated with a type of muscle damage resulting in activation of both satellite and nonsatellite muscle progenitor cells. These muscle progenitors committed to a myogenic program, but were inhibited from completing differentiation by an event linked with persistent expression of the self-renewing factor Pax7. Overexpression of Pax7 was sufficient to induce atrophy in normal muscle, while under tumor conditions, the reduction of Pax7 or exogenous addition of its downstream target, MyoD, reversed wasting by restoring cell differentiation and fusion with injured fibers. Furthermore, Pax7 was induced by serum factors from cachectic mice and patients, in an NF-κB-dependent manner, both in vitro and in vivo. Together, these results suggest that Pax7 responds to NF-κB by impairing the regenerative capacity of myogenic cells in the muscle microenvironment to drive muscle wasting in cancer.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Animals
- Cachexia/etiology
- Cachexia/metabolism
- Cachexia/pathology
- Case-Control Studies
- Cell Line, Tumor
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Mice
- Mice, Inbred mdx
- Mice, Nude
- Mice, Transgenic
- Microscopy, Electron, Transmission
- Middle Aged
- Muscle Development
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/pathology
- NF-kappa B/metabolism
- PAX7 Transcription Factor/genetics
- PAX7 Transcription Factor/metabolism
- Pancreatic Neoplasms/complications
- Pancreatic Neoplasms/metabolism
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Tumor Microenvironment
- Young Adult
Collapse
|
39
|
Chen W, Lv YT, Zhang HX, Ruan D, Wang S, Lin YC. Developmental specificity in skeletal muscle of late-term avian embryos and its potential manipulation. Poult Sci 2013; 92:2754-64. [PMID: 24046424 DOI: 10.3382/ps.2013-03099] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Unlike the mammalian fetus, development of the avian embryo is independent of the maternal uterus and is potentially vulnerable to physiological and environmental stresses close to hatch. In contrast to the fetus of late gestation in mammals, skeletal muscle in avian embryos during final incubation shows differential developmental characteristics: 1) muscle mobilization (also called atrophy) is selectively enhanced in the type II fibers (pectoral muscle) but not in the type I fibers (biceps femoris and semimembranosus muscle), involving activation of ubiquitin-mediated protein degradation and suppression of S6K1-mediated protein translation; 2) the proliferative activity of satellite cells is decreased in the atrophied muscle of late-term embryos but enhanced at the day of hatch, probably preparing for the postnatal growth. The mobilization of muscle may represent an adaptive response of avian embryos to external (environmental) or internal (physiological) changes, considering there are developmental transitions both in hormones and requirements for glycolytic substrates from middle-term to late-term incubation. Although the exact mechanism triggering muscle fiber atrophy is still unknown, nutritional and endocrine changes may be of importance. The atrophied muscle fiber recovers as soon as feed and water are available to the hatchling. In ovo feeding of late-term embryos has been applied to improve the nutritional status and therein enhances muscle development. Similarly, in ovo exposure to higher temperature or green light during the critical period of muscle development are also demonstrated to be potential strategies to promote pre- and posthatch muscle growth.
Collapse
Affiliation(s)
- W Chen
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | | | | | | | | | | |
Collapse
|
40
|
Mohamed JS, Lopez MA, Cox GA, Boriek AM. Ankyrin repeat domain protein 2 and inhibitor of DNA binding 3 cooperatively inhibit myoblast differentiation by physical interaction. J Biol Chem 2013; 288:24560-8. [PMID: 23824195 DOI: 10.1074/jbc.m112.434423] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Ankyrin repeat domain protein 2 (ANKRD2) translocates from the nucleus to the cytoplasm upon myogenic induction. Overexpression of ANKRD2 inhibits C2C12 myoblast differentiation. However, the mechanism by which ANKRD2 inhibits myoblast differentiation is unknown. We demonstrate that the primary myoblasts of mdm (muscular dystrophy with myositis) mice (pMB(mdm)) overexpress ANKRD2 and ID3 (inhibitor of DNA binding 3) proteins and are unable to differentiate into myotubes upon myogenic induction. Although suppression of either ANKRD2 or ID3 induces myoblast differentiation in mdm mice, overexpression of ANKRD2 and inhibition of ID3 or vice versa is insufficient to inhibit myoblast differentiation in WT mice. We identified that ANKRD2 and ID3 cooperatively inhibit myoblast differentiation by physical interaction. Interestingly, although MyoD activates the Ankrd2 promoter in the skeletal muscles of wild-type mice, SREBP-1 (sterol regulatory element binding protein-1) activates the same promoter in the skeletal muscles of mdm mice, suggesting the differential regulation of Ankrd2. Overall, we uncovered a novel pathway in which SREBP-1/ANKRD2/ID3 activation inhibits myoblast differentiation, and we propose that this pathway acts as a critical determinant of the skeletal muscle developmental program.
Collapse
Affiliation(s)
- Junaith S Mohamed
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
41
|
Suetta C, Frandsen U, Mackey AL, Jensen L, Hvid LG, Bayer ML, Petersson SJ, Schrøder HD, Andersen JL, Aagaard P, Schjerling P, Kjaer M. Ageing is associated with diminished muscle re-growth and myogenic precursor cell expansion early after immobility-induced atrophy in human skeletal muscle. J Physiol 2013; 591:3789-804. [PMID: 23732643 DOI: 10.1113/jphysiol.2013.257121] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Recovery of skeletal muscle mass from immobilisation-induced atrophy is faster in young than older individuals, yet the cellular mechanisms remain unknown. We examined the cellular and molecular regulation of muscle recovery in young and older human subjects subsequent to 2 weeks of immobility-induced muscle atrophy. Retraining consisted of 4 weeks of supervised resistive exercise in 9 older (OM: mean age) 67.3, range 61-74 yrs) and 11 young (YM: mean age 24.4, range 21-30 yrs) males. Measures of myofibre area (MFA), Pax7-positive satellite cells (SCs) associated with type I and type II muscle fibres, as well as gene expression analysis of key growth and transcription factors associated with local skeletal muscle milieu, were performed after 2 weeks immobility (Imm) and following 3 days (+3d) and 4 weeks (+4wks) of retraining. OM demonstrated no detectable gains in MFA (vastus lateralis muscle) and no increases in number of Pax7-positive SCs following 4wks retraining, whereas YM increased their MFA (P < 0.05), number of Pax7-positive cells, and had more Pax7-positive cells per type II fibre than OM at +3d and +4wks (P < 0.05). No age-related differences were observed in mRNA expression of IGF-1Ea, MGF, MyoD1 and HGF with retraining, whereas myostatin expression levels were more down-regulated in YM compared to OM at +3d (P < 0.05). In conclusion, the diminished muscle re-growth after immobilisation in elderly humans was associated with a lesser response in satellite cell proliferation in combination with an age-specific regulation of myostatin. In contrast, expression of local growth factors did not seem to explain the age-related difference in muscle mass recovery.
Collapse
Affiliation(s)
- C Suetta
- Institute of Sports Medicine and Center of Healthy Ageing, Faculty of Health and Medical Sciences, University of Copenhagen, Bispebjerg Hospital, Denmark.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sirabella D, De Angelis L, Berghella L. Sources for skeletal muscle repair: from satellite cells to reprogramming. J Cachexia Sarcopenia Muscle 2013; 4:125-36. [PMID: 23314905 PMCID: PMC3684700 DOI: 10.1007/s13539-012-0098-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 11/28/2012] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle regeneration is the process that ensures tissue repair after damage by injury or in degenerative diseases such as muscular dystrophy. Satellite cells, the adult skeletal muscle progenitor cells, are commonly considered to be the main cell type involved in skeletal muscle regeneration. Their mechanism of action in this process is extensively characterized. However, evidence accumulated in the last decade suggests that other cell types may participate in skeletal muscle regeneration. Although their actual contribution to muscle formation and regeneration is still not clear; if properly manipulated, these cells may become new suitable and powerful sources for cell therapy of skeletal muscle degenerative diseases. Mesoangioblasts, vessel associated stem/progenitor cells with high proliferative, migratory and myogenic potential, are very good candidates for clinical applications and are already in clinical experimentation. In addition, pluripotent stem cells are very promising sources for regeneration of most tissues, including skeletal muscle. Conditions such as muscle cachexia or aging that severely alter homeostasis may be counteracted by transplantation of donor and/or recruitment and activation of resident muscle stem/progenitor cells. Advantages and limitations of different cell therapy approaches will be discussed.
Collapse
Affiliation(s)
- Dario Sirabella
- />Department of Biomedical Engineering, Columbia University, 2920 Broadway, New York, NY 10027-7164 USA
| | - Luciana De Angelis
- />DAHFMO, Unit of Histology and Medical Embryology, University of Roma “La Sapienza”, Via Scarpa, 14, 00161 Rome, Italy
| | - Libera Berghella
- />IRCCS Fondazione S. Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
- />HudsonAlpha Institute for Biotechnology, 601 Genome Way, Huntsville, AL 35806 USA
| |
Collapse
|
43
|
Dong Y, Pan JS, Zhang L. Myostatin suppression of Akirin1 mediates glucocorticoid-induced satellite cell dysfunction. PLoS One 2013; 8:e58554. [PMID: 23516508 PMCID: PMC3596298 DOI: 10.1371/journal.pone.0058554] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 02/05/2013] [Indexed: 11/28/2022] Open
Abstract
Glucocorticoids production is increased in many pathological conditions that are associated with muscle loss, but their role in causing muscle wasting is not fully understood. We have demonstrated a new mechanism of glucocorticoid-induced muscle atrophy: Dexamethasone (Dex) suppresses satellite cell function contributing to the development of muscle atrophy. Specifically, we found that Dex decreases satellite cell proliferation and differentiation in vitro and in vivo. The mechanism involved Dex-induced upregulation of myostatin and suppression of Akirin1, a promyogenic gene. When myostatin was inhibited in Dex-treated mice, Akirin1 expression increased as did satellite cell activity, muscle regeneration and muscle growth. In addition, silencing myostatin in myoblasts or satellite cells prevented Dex from suppressing Akirin1 expression and cellular proliferation and differentiation. Finally, overexpression of Akirin1 in myoblasts increased their expression of MyoD and myogenin and improved cellular proliferation and differentiation, theses improvements were no longer suppressed by Dex. We conclude that glucocorticoids stimulate myostatin which inhibits Akirin1 expression and the reparative functions of satellite cells. These responses attribute to muscle atrophy. Thus, inhibition of myostatin or increasing Akirin1 expression could lead to therapeutic strategies for improving satellite cell activation and enhancing muscle growth in diseases associated with increased glucocorticoid production.
Collapse
Affiliation(s)
- Yanjun Dong
- Department of Medicine, Nephrology Division, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jenny S. Pan
- Department of Medicine, Nephrology Division, Baylor College of Medicine, Houston, Texas, United States of America
| | - Liping Zhang
- Department of Medicine, Nephrology Division, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
44
|
Kuraitis D, Berardinelli MG, Suuronen EJ, Musarò A. A necrotic stimulus is required to maximize matrix-mediated myogenesis in mice. Dis Model Mech 2013; 6:793-801. [PMID: 23471914 PMCID: PMC3634661 DOI: 10.1242/dmm.011072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Biomaterials that are similar to skeletal muscle extracellular matrix have been shown to augment regeneration in ischemic muscle. In this study, treatment with a collagen-based matrix stimulated molecular myogenesis in an mdx murine model of necrosis. Matrix-treated animals ran ≥ 40% further, demonstrating functional regeneration, and expressed increased levels of myogenic transcripts. By contrast, matrix treatment was unable to induce transcriptional or functional changes in an MLC/SOD1(G93A) atrophic mouse model. In vitro, satellite cells were cultured under standard conditions, on matrix, in the presence of myocyte debris (to simulate a necrotic-like environment) or with both matrix and necrotic stimuli. Exposure to both matrix and necrotic stimuli induced the greatest increases in mef2c, myf5, myoD and myogenin transcripts. Furthermore, conditioned medium collected from satellite cells cultured with both stimuli contained elevated levels of factors that modulate satellite cell activation and proliferation, such as FGF-2, HGF and SDF-1. Application of the conditioned medium to C2C12 myoblasts accelerated maturation, as demonstrated by increased mef2c, myf5 and myogenin transcripts and fusion indexes. In summary, the collagen matrix required a necrotic stimulus to enhance the maturation of satellite cells and their secretion of a myogenic cocktail. Considering that matrix treatment supports myogenesis only in in vivo models that exhibit necrosis, this study demonstrates that a necrotic environment is required to maximize matrix-mediated myogenesis.
Collapse
Affiliation(s)
- Drew Kuraitis
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM, Sapienza University of Rome, Via Scarpa 14, Rome 00161, Italy
| | | | | | | |
Collapse
|
45
|
Ye F, Mathur S, Liu M, Borst SE, Walter GA, Sweeney HL, Vandenborne K. Overexpression of insulin-like growth factor-1 attenuates skeletal muscle damage and accelerates muscle regeneration and functional recovery after disuse. Exp Physiol 2013; 98:1038-52. [PMID: 23291913 DOI: 10.1113/expphysiol.2012.070722] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Skeletal muscle is a highly dynamic tissue that responds to endogenous and external stimuli, including alterations in mechanical loading and growth factors. In particular, the antigravity soleus muscle experiences significant muscle atrophy during disuse and extensive muscle damage upon reloading. Given that insulin-like growth factor-1 (IGF-1) has been implicated as a central regulator of muscle repair and modulation of muscle size, we examined the effect of virally mediated overexpression of IGF-1 on the soleus muscle following hindlimb cast immobilization and upon reloading. Recombinant IGF-1 cDNA virus was injected into one of the posterior hindlimbs of the mice, while the contralateral limb was injected with saline (control). At 20 weeks of age, both hindlimbs were immobilized for 2 weeks to induce muscle atrophy in the soleus and ankle plantarflexor muscle group. Subsequently, the mice were allowed to reambulate, and muscle damage and recovery were monitored over a period of 2-21 days. The primary finding of this study was that IGF-1 overexpression attenuated reloading-induced muscle damage in the soleus muscle, and accelerated muscle regeneration and force recovery. Muscle T2 assessed by magnetic resonance imaging, a non-specific marker of muscle damage, was significantly lower in IGF-1-injected compared with contralateral soleus muscles at 2 and 5 days reambulation (P<0.05). The reduced prevalence of muscle damage in IGF-1-injected soleus muscles was confirmed on histology, with a lower fractional area of abnormal muscle tissue in IGF-1-injected muscles at 2 days reambulation (33.2±3.3 versus 54.1±3.6%, P<0.05). Evidence of the effect of IGF-1 on muscle regeneration included timely increases in the number of central nuclei (21% at 5 days reambulation), paired-box transcription factor 7 (36% at 5 days), embryonic myosin (37% at 10 days) and elevated MyoD mRNA (7-fold at 2 days) in IGF-1-injected limbs (P<0.05). These findings demonstrate a potential role of IGF-1 in protecting unloaded skeletal muscles from damage and accelerating muscle repair and regeneration.
Collapse
Affiliation(s)
- Fan Ye
- Department of Physical Therapy, PO Box 100154, Room 1142, PHHP Building, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Schenk HC, Krampfl K, Baumgärtner W, Tipold A. Canine muscle cell culture and consecutive patch-clamp measurements - a new approach to characterize muscular diseases in dogs. BMC Vet Res 2012; 8:227. [PMID: 23171640 PMCID: PMC3539935 DOI: 10.1186/1746-6148-8-227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 11/18/2012] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The recognition of functional muscular disorders, (e.g. channelopathies like Myotonia) is rising in veterinary neurology. Morphologic (e.g. histology) and even genetic based studies in these diseases are not able to elucidate the functional pathomechanism. As there is a deficit of knowledge and skills considering this special task, the aim of the current pilot study was to develop a canine muscle cell culture system derived from muscle biopsies of healthy client-owned dogs, which allows sampling of the biopsies under working conditions in the daily veterinary practise. RESULTS Muscular biopsies from 16 dogs of different age and breed were taken during standard surgical procedures and were stored for one to three days at 4°C in a transport medium in order to simulate shipping conditions. Afterwards biopsies were professionally processed, including harvesting of satellite cells, inducing their proliferation, differentiating them into myotubes and recultivating myotubes after long-term storage in liquid nitrogen. Myogenic origin of cultured cells was determined by immunofluorescence, immunohistology and by their typical morphology after inducing differentiation. Subsequent to the differentiation into myotubes feasibility of patch-clamp recordings of voltage gated ion channels was successfully. CONCLUSION We have developed a canine muscle cell culture system, which allows sampling of biopsies from young and old dogs of different breeds under practical conditions. Patch clamp measurements can be carried out with the cultured myotubes demonstrating potential of these cells as source for functional research.
Collapse
Affiliation(s)
- Henning Christian Schenk
- Department of Small Animal Medicine and Surgery, University of Veterinary Medicine, Hannover, Germany.
| | | | | | | |
Collapse
|
47
|
Wu X, Walters TJ, Rathbone CR. Skeletal muscle satellite cell activation following cutaneous burn in rats. Burns 2012; 39:736-44. [PMID: 23146573 DOI: 10.1016/j.burns.2012.10.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 10/10/2012] [Accepted: 10/15/2012] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cutaneous burn distant from skeletal muscles induces atrophy; however, its effect on muscle stem cells resident in skeletal muscle (satellite cells) distal to burn is not known. METHODS Satellite cell activation was measured in predominantly fast-twitch [tibialis anterior, extensor digitorum longus (EDL), plantaris, and gastrocnemius] and slow-twitch (soleus) muscles of rats that received either 40% total body surface area full-thickness scald burn or sham burn to the trunk area by determining bromodeoxyuridine incorporation, MyoD, and Pax7 immunohistochemistry in vivo ≤48 h after burn. To determine the effects of circulating factors on satellite cell activation, satellite cell cultures were treated with serum from sham or burn rats. RESULTS In vivo activation of satellite cells was increased in fast muscles isolated from burn as compared to sham animals, whereas a significant response was not seen in slow muscles. Serum taken from animals in the burn group increased the activation of satellite cells isolated from both sham and burn animals in vitro, suggesting that circulating factors have the potential to increase satellite cell activation following burn. CONCLUSIONS Increases in satellite cell activation in muscles distal to burn are fiber-type-dependent, and circulating factors may play a role in the activation of satellite cells following burn. A better understanding of the impact of burn on satellite cell functionality will allow us to identify the cellular mechanisms of long-term muscle atrophy.
Collapse
Affiliation(s)
- Xiaowu Wu
- Extremity Trauma and Regenerative Medicine, US Army Institute of Surgical Research, Fort Sam Houston, TX 78234-6315, USA.
| | | | | |
Collapse
|
48
|
Jackson JR, Mula J, Kirby TJ, Fry CS, Lee JD, Ubele MF, Campbell KS, McCarthy JJ, Peterson CA, Dupont-Versteegden EE. Satellite cell depletion does not inhibit adult skeletal muscle regrowth following unloading-induced atrophy. Am J Physiol Cell Physiol 2012; 303:C854-61. [PMID: 22895262 DOI: 10.1152/ajpcell.00207.2012] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Resident muscle stem cells, known as satellite cells, are thought to be the main mediators of skeletal muscle plasticity. Satellite cells are activated, replicate, and fuse into existing muscle fibers in response to both muscle injury and mechanical load. It is generally well-accepted that satellite cells participate in postnatal growth, hypertrophy, and muscle regeneration following injury; however, their role in muscle regrowth following an atrophic stimulus remains equivocal. The current study employed a genetic mouse model (Pax7-DTA) that allowed for the effective depletion of >90% of satellite cells in adult muscle upon the administration of tamoxifen. Vehicle and tamoxifen-treated young adult female mice were either hindlimb suspended for 14 days to induce muscle atrophy or hindlimb suspended for 14 days followed by 14 days of reloading to allow regrowth, or they remained ambulatory for the duration of the experimental protocol. Additionally, 5-bromo-2'-deoxyuridine (BrdU) was added to the drinking water to track cell proliferation. Soleus muscle atrophy, as measured by whole muscle wet weight, fiber cross-sectional area, and single-fiber width, occurred in response to suspension and did not differ between satellite cell-depleted and control muscles. Furthermore, the depletion of satellite cells did not attenuate muscle mass or force recovery during the 14-day reloading period, suggesting that satellite cells are not required for muscle regrowth. Myonuclear number was not altered during either the suspension or the reloading period in soleus muscle fibers from vehicle-treated or satellite cell-depleted animals. Thus, myonuclear domain size was reduced following suspension due to decreased cytoplasmic volume and was completely restored following reloading, independent of the presence of satellite cells. These results provide convincing evidence that satellite cells are not required for muscle regrowth following atrophy and that, instead, the myonuclear domain size changes as myofibers adapt.
Collapse
Affiliation(s)
- Janna R Jackson
- Department of Rehabilitation Sciences, College of Health Sciences, University of Kentucky, Lexington, 40536-0200, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Park S, Nozaki K, Guyton MK, Smith JA, Ray SK, Banik NL. Calpain inhibition attenuated morphological and molecular changes in skeletal muscle of experimental allergic encephalomyelitis rats. J Neurosci Res 2012; 90:2134-45. [PMID: 22715087 DOI: 10.1002/jnr.23096] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 04/30/2012] [Accepted: 05/09/2012] [Indexed: 12/22/2022]
Abstract
Muscle weakness and atrophy are important manifestations of multiple sclerosis (MS). To investigate the pathophysiological mechanisms of skeletal muscle change in MS, we induced experimental autoimmune encephalomyelitis (EAE) in Lewis male rats and examined morphological and molecular changes in skeletal muscle. We also treated EAE rats with calpepetin, a calpain inhibitor, to examine its beneficial effects on skeletal muscle damage. Morphological changes in muscle tissue of EAE rats included smaller and irregularly shaped muscle fibers and fibrosis. Western blot analysis demonstrated increased calpain:calpastatin ratio, inflammation-related transcription factors (nuclear factor-κB:inhibitor of κB α ratio), and proinflammatory enzymes (cyclooxygenase-2). TUNEL-positive myonuclei in skeletal muscle cells of EAE rats indicated cell death. In addition, markers of apoptotic cell death (Bax:Bcl-2 ratio and caspase-12 protein levels) were elevated. Expression of muscle-specific ubiquitin ligases (muscle atrophy F-box and muscle ring finger protein 1), was upregulated in muscle tissue of EAE-vehicle animals. Both prophylactic and therapeutic treatment with calpeptin partially attenuated muscle changes noted in EAE animals. These results indicate that morphological and molecular changes including apoptotic cell death and protein breakdown develop in skeletal muscle of EAE animals and that these changes can be reversed by calpain inhibition.
Collapse
Affiliation(s)
- Sookyoung Park
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | |
Collapse
|
50
|
Kudryashova E, Kramerova I, Spencer MJ. Satellite cell senescence underlies myopathy in a mouse model of limb-girdle muscular dystrophy 2H. J Clin Invest 2012; 122:1764-76. [PMID: 22505452 DOI: 10.1172/jci59581] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 02/29/2012] [Indexed: 12/25/2022] Open
Abstract
Mutations in the E3 ubiquitin ligase tripartite motif-containing 32 (TRIM32) are responsible for the disease limb-girdle muscular dystrophy 2H (LGMD2H). Previously, we generated Trim32 knockout mice (Trim32-/- mice) and showed that they display a myopathic phenotype accompanied by neurogenic features. Here, we used these mice to investigate the muscle-specific defects arising from the absence of TRIM32, which underlie the myopathic phenotype. Using 2 models of induced atrophy, we showed that TRIM32 is dispensable for muscle atrophy. Conversely, TRIM32 was necessary for muscle regrowth after atrophy. Furthermore, TRIM32-deficient primary myoblasts underwent premature senescence and impaired myogenesis due to accumulation of PIAS4, an E3 SUMO ligase and TRIM32 substrate that was previously shown to be associated with senescence. Premature senescence of myoblasts was also observed in vivo in an atrophy/regrowth model. Trim32-/- muscles had substantially fewer activated satellite cells, increased PIAS4 levels, and growth failure compared with wild-type muscles. Moreover, Trim32-/- muscles exhibited features of premature sarcopenia, such as selective type II fast fiber atrophy. These results imply that premature senescence of muscle satellite cells is an underlying pathogenic feature of LGMD2H and reveal what we believe to be a new mechanism of muscular dystrophy associated with reductions in available satellite cells and premature sarcopenia.
Collapse
Affiliation(s)
- Elena Kudryashova
- Department of Neurology, Center for Duchenne Muscular Dystrophy at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1606, USA
| | | | | |
Collapse
|