1
|
Pause FC, Baufeld A, Urli S, Crociati M, Stradaioli G, Vanselow J, Kalbe C. Exploring the influence of polystyrene-nanoplastics on two distinct in vitro systems in farm animals: A pilot study. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 976:179378. [PMID: 40209587 DOI: 10.1016/j.scitotenv.2025.179378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 02/17/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025]
Abstract
The harmful effects of micro- and nanoplastics (MNPs) on the aquatic ecosystem are already well established, and several studies have demonstrated that MNPs can contaminate the soil. However, the impact of MNPs on farm animals, whose products are intended for human consumption, as well as the accumulation and translocation of these particles in their bodies, is less investigated and not well understood. To address this issue, we evaluated the cellular uptake and the effects of three different concentrations (5, 25, and 75 μg/mL) of 100 nm polystyrene nanoplastics (PS-NPs) on ovarian bovine granulosa cells (GCs) and porcine myoblasts derived from skeletal muscle satellite cells as in vitro primary cell culture models. The uptake of PS-NPs was shown for all the concentrations tested, both for GCs and for myoblasts. The results for GCs reported a significant decrease in cell viability (P < 0.05) for all concentrations of nanoplastics tested compared to the control. However, steroid hormone production and the mRNA expression of GC physiology marker genes were not affected. The results for myoblasts showed a significant decrease in the mean confluence (P < 0.05) after exposure to a concentration of 75 μg/mL of nanoplastics compared to the control. This may be indicative of an initial inhibition of muscle fibre formation. However, cell viability, proliferative capacity, and the mRNA expression of myogenesis-associated genes were not affected. As there is currently no standard method for assessing the quantity of particles that overcome the anatomical barriers and accumulate in various parts of the body, recognizing the implications of exposure to MNPs in farm animals can help us to better comprehend the potential risks to human health. This knowledge is critical for developing informed treatment and avoidance strategies, ensuring the safety of both the food we consume and the environment in which it is produced.
Collapse
Affiliation(s)
- Francesca Corte Pause
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Via Delle, Scienze 206, 33100 Udine, Italy.
| | - Anja Baufeld
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| | - Susy Urli
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Via Delle, Scienze 206, 33100 Udine, Italy.
| | - Martina Crociati
- Department of Veterinary Medicine, University of Perugia, Via S. Costanzo 4, 06126 Perugia, Italy; Centre for Perinatal and Reproductive Medicine, University of Perugia, 06129 Perugia, Italy.
| | - Giuseppe Stradaioli
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Via Delle, Scienze 206, 33100 Udine, Italy.
| | - Jens Vanselow
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| | - Claudia Kalbe
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| |
Collapse
|
2
|
Hoseini R, Hoseini Z, Kamangar A. Myogenic differentiation markers in muscle tissue after aerobic training. Heliyon 2025; 11:e41888. [PMID: 39897925 PMCID: PMC11787638 DOI: 10.1016/j.heliyon.2025.e41888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 11/21/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Aerobic training induces a myriad of adaptations in muscle tissue, encompassing alterations in muscle fiber type composition, hypertrophy, and metabolic capacity. Understanding the potential role of myogenic differentiation markers (MDFs), such as Pax7, MyoD, Myogenin, and myosin heavy chain (MHC) isoforms, in mediating these adaptations is of paramount importance. The review delves into the intricate molecular mechanisms underlying the regulation of MDFs following aerobic training, elucidating the role of key signaling pathways including the MAPK/ERK, PI3K/Akt, and AMPK pathways, among others. These pathways play pivotal roles in orchestrating the expression and activity of MDFs, ultimately influencing muscle adaptation and regeneration. The comprehension of MDFs in the context of aerobic training is far-reaching, offering the potential for targeted interventions to optimize muscle adaptation and regeneration. This review identifies the need for further research to unveil the precise molecular mechanisms of the activation and interaction of myogenic differentiation markers with other signaling pathways, as well as to explore their potential as therapeutic targets for muscle-related conditions. This review article also provides a thorough analysis of MDFs in muscle tissue after aerobic training, highlighting their potential clinical implications and outlining future research directions in this area.
Collapse
Affiliation(s)
- Rastegar Hoseini
- Assistant Professor of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Zahra Hoseini
- PhD of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| | - Ayob Kamangar
- PhD Student of Exercise Physiology, Department of Exercise Physiology, Faculty of Sport Sciences, Razi University, Kermanshah, Iran
| |
Collapse
|
3
|
Liu H, Chehade L, Deguise MO, De Repentigny Y, Kothary R. SMN depletion impairs skeletal muscle formation and maturation in a mouse model of SMA. Hum Mol Genet 2025; 34:21-31. [PMID: 39505369 PMCID: PMC11756284 DOI: 10.1093/hmg/ddae162] [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: 08/14/2024] [Revised: 10/09/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024] Open
Abstract
Spinal muscular atrophy (SMA) is characterized by low levels of the ubiquitously expressed Survival Motor Neuron (SMN) protein, leading to progressive muscle weakness and atrophy. Skeletal muscle satellite cells play a crucial role in muscle fiber maintenance, repair, and remodelling. While the effects of SMN depletion in muscle are well documented, its precise role in satellite cell function remains largely unclear. Using the Smn2B/- mouse model, we investigated SMN-depleted satellite cell biology through single fiber culture studies. Myofibers from Smn2B/- mice were smaller in size, shorter in length, had reduced myonuclear domain size, and reduced sub-synaptic myonuclear clusters-all suggesting impaired muscle function and integrity. These changes were accompanied by a reduction in the number of myonuclei in myofibers from Smn2B/- mice across all disease stages examined. Although the number of satellite cells in myofibers was significantly reduced, those remaining retained their capacity for myogenic activation and proliferation. These findings support the idea that a dysregulated myogenic process could be occurring as early in muscle stem cells during muscle formation and maturation in SMA. Targeting those pathways could offer additional options for combinatorial therapies for SMA.
Collapse
MESH Headings
- Animals
- Muscular Atrophy, Spinal/genetics
- Muscular Atrophy, Spinal/pathology
- Muscular Atrophy, Spinal/metabolism
- Mice
- Disease Models, Animal
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle Development/genetics
- Survival of Motor Neuron 1 Protein/genetics
- Survival of Motor Neuron 1 Protein/metabolism
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Survival of Motor Neuron 2 Protein/genetics
- Mice, Knockout
- Humans
- Motor Neurons/metabolism
- Motor Neurons/pathology
- Cell Proliferation
Collapse
Affiliation(s)
- Hong Liu
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | - Lucia Chehade
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Marc-Olivier Deguise
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
- Center for Neuromuscular Disease, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Department of Pediatrics, Children’s Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Yves De Repentigny
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Center for Neuromuscular Disease, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
- Department of Medicine, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
4
|
Melzener L, Schaeken L, Fros M, Messmer T, Raina D, Kiessling A, van Haaften T, Spaans S, Doǧan A, Post MJ, Flack JE. Optimisation of cell fate determination for cultivated muscle differentiation. Commun Biol 2024; 7:1493. [PMID: 39532984 PMCID: PMC11557827 DOI: 10.1038/s42003-024-07201-6] [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: 01/05/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
Production of cultivated meat requires defined medium formulations for the robust differentiation of myogenic cells into mature skeletal muscle fibres in vitro. Although these formulations can drive myogenic differentiation levels comparable to serum-starvation-based protocols, the resulting cultures are often heterogeneous, with a significant proportion of cells not participating in myofusion, limiting maturation of the muscle. To address this problem, we employed RNA sequencing to analyse heterogeneity in differentiating bovine satellite cells at single-nucleus resolution, identifying distinct cellular subpopulations including proliferative cells that fail to exit the cell cycle and quiescent 'reserve cells' that do not commit to myogenic differentiation. Our findings indicate that the MEK/ERK, NOTCH, and RXR pathways are active during the initial stages of myogenic cell fate determination, and by targeting these pathways, we can promote cell cycle exit while reducing reserve cell formation. This optimised medium formulation consistently yields fusion indices close to 100% in 2D culture. Furthermore, we demonstrate that these conditions enhance myotube formation and actomyosin accumulation in 3D bovine skeletal muscle constructs, providing proof of principle for the generation of highly differentiated cultivated muscle with excellent mimicry to traditional muscle.
Collapse
Affiliation(s)
- Lea Melzener
- Mosa Meat B.V., Maastricht, The Netherlands
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | | | | | - Tobias Messmer
- Mosa Meat B.V., Maastricht, The Netherlands
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | | | | | | | | | - Arin Doǧan
- Mosa Meat B.V., Maastricht, The Netherlands
| | - Mark J Post
- Mosa Meat B.V., Maastricht, The Netherlands
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Joshua E Flack
- Mosa Meat B.V., Maastricht, The Netherlands.
- Department of Biotechnology, Delft University of Technology, Delft, The Netherlands.
| |
Collapse
|
5
|
Hahn O, Peters K, Hartmann A, Dannenberger D, Kalbe C. Potential of animal-welfare compliant and sustainably sourced serum from pig slaughter blood. Cell Tissue Res 2024; 397:205-214. [PMID: 38990342 PMCID: PMC11371839 DOI: 10.1007/s00441-024-03904-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
The animal product most used as a stimulatory additive for cell cultivation is still fetal bovine serum (FBS). Besides the ethical concerns regarding serum collection, the main problems of FBS are batch-to-batch variability and the resulting risk of lower reproducibility, the differences between species, the presence of undefined/unknown components, and the risk of contamination. In contrast, pig blood, which is a by-product of slaughter, is a sufficiently available and sustainable resource with a high degree of standardization in terms of donor age, weight, and genetics. The variations in preparations from pig slaughter blood seem to be comparatively low, and consequently, batch effects might be much smaller, suggesting that the reproducibility of the research data obtained may be increased. Our pilot study aimed to investigate, as a proof of concept, whether adult human and porcine stem cells of different tissue origins proliferate and differentiate adequately when FBS is completely or partially replaced by porcine serum (PS). We could show that the human and porcine stem cells were vital and proliferated under partial and full PS supplementation. Furthermore, using PS, the two cell types studied showed tissue-specific differentiation (i.e., lipid vacuoles as a sign of adipogenic or myotubes as a sign of myogenic differentiation). In conclusion, the pig slaughter blood-derived serum has promising potential to be a replacement for FBS in adult stem cell cultures. Therefore, it could serve as a basis for the development of new cell culture supplements.
Collapse
Affiliation(s)
- Olga Hahn
- Institute for Cell Biology, University Medical Center Rostock, Rostock, Germany
| | - Kirsten Peters
- Institute for Cell Biology, University Medical Center Rostock, Rostock, Germany
| | - Alexander Hartmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Rostock, Rostock, Germany
| | - Dirk Dannenberger
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, D-18196, Dummerstorf, Germany
| | - Claudia Kalbe
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, D-18196, Dummerstorf, Germany.
| |
Collapse
|
6
|
Madigan LA, Jaime D, Chen I, Fallon JR. MuSK-BMP signaling in adult muscle stem cells maintains quiescence and regulates myofiber size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.17.541238. [PMID: 37292636 PMCID: PMC10245747 DOI: 10.1101/2023.05.17.541238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A central question in adult stem cell biology is elucidating the signaling pathways regulating their dynamics and function in diverse physiological and age-related contexts. Muscle stem cells in adults (Satellite Cells; SCs) are generally quiescent but can activate and contribute to muscle repair and growth. Here we tested the role of the MuSK-BMP pathway in regulating adult SC quiescence by deletion of the BMP-binding MuSK Ig3 domain ('ΔIg3-MuSK'). At 3 months of age SC and myonuclei numbers and myofiber size were comparable to WT. However, at 5 months of age SC density was decreased while myofiber size, myonuclear number and grip strength were increased - indicating that SCs had activated and productively fused into the myofibers over this interval. Transcriptomic analysis showed that SCs from uninjured ΔIg3-MuSK mice exhibit signatures of activation. Regeneration experiments showed that ΔIg3-MuSK SCs maintain full stem cell function. Expression of ΔIg3-MuSK in adult SCs was sufficient to break quiescence and increase myofiber size. We conclude that the MuSK-BMP pathway regulates SC quiescence and myofiber size in a cell autonomous, age-dependent manner. Targeting MuSK-BMP signaling in muscle stem cells thus emerges a therapeutic strategy for promoting muscle growth and function in the settings of injury, disease, and aging. Highlights MuSK, in its role as a BMP co-receptor, regulates adult muscle stem cell quiescenceThe MuSK-BMP pathway acts cell autonomouslyIncreased muscle size and function with preservation of myonuclear density and stemness in mice with attenuated MuSK-BMP signaling.
Collapse
|
7
|
Kiperman T, Ma K. Circadian Clock in Muscle Disease Etiology and Therapeutic Potential for Duchenne Muscular Dystrophy. Int J Mol Sci 2024; 25:4767. [PMID: 38731986 PMCID: PMC11083552 DOI: 10.3390/ijms25094767] [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: 03/07/2024] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Circadian clock and clock-controlled output pathways exert temporal control in diverse aspects of skeletal muscle physiology, including the maintenance of muscle mass, structure, function, and metabolism. They have emerged as significant players in understanding muscle disease etiology and potential therapeutic avenues, particularly in Duchenne muscular dystrophy (DMD). This review examines the intricate interplay between circadian rhythms and muscle physiology, highlighting how disruptions of circadian regulation may contribute to muscle pathophysiology and the specific mechanisms linking circadian clock dysregulation with DMD. Moreover, we discuss recent advancements in chronobiological research that have shed light on the circadian control of muscle function and its relevance to DMD. Understanding clock output pathways involved in muscle mass and function offers novel insights into the pathogenesis of DMD and unveils promising avenues for therapeutic interventions. We further explore potential chronotherapeutic strategies targeting the circadian clock to ameliorate muscle degeneration which may inform drug development efforts for muscular dystrophy.
Collapse
Affiliation(s)
| | - Ke Ma
- Department of Diabetes Complications & Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA;
| |
Collapse
|
8
|
Sutcu HH, Rassinoux P, Donnio LM, Neuillet D, Vianna F, Gabillot O, Mari PO, Baldeyron C, Giglia-Mari G. Decline of DNA damage response along with myogenic differentiation. Life Sci Alliance 2024; 7:e202302279. [PMID: 37993260 PMCID: PMC10665522 DOI: 10.26508/lsa.202302279] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023] Open
Abstract
DNA integrity is incessantly confronted to agents inducing DNA lesions. All organisms are equipped with a network of DNA damage response mechanisms that will repair DNA lesions and restore proper cellular activities. Despite DNA repair mechanisms have been revealed in replicating cells, still little is known about how DNA lesions are repaired in postmitotic cells. Muscle fibers are highly specialized postmitotic cells organized in syncytia and they are vulnerable to age-related degeneration and atrophy after radiotherapy treatment. We have studied the DNA repair capacity of muscle fiber nuclei and compared it with the one measured in proliferative myoblasts here. We focused on the DNA repair mechanisms that correct ionizing radiation (IR)-induced lesions, namely the base excision repair, the nonhomologous end joining, and the homologous recombination (HR). We found that in the most differentiated myogenic cells, myotubes, these DNA repair mechanisms present weakened kinetics of recruitment of DNA repair proteins to IR-damaged DNA. For base excision repair and HR, this decline can be linked to reduced steady-state levels of key proteins involved in these processes.
Collapse
Affiliation(s)
- Haser H Sutcu
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRAcc, Fontenay-aux-Roses, France
| | - Phoebe Rassinoux
- Pathophysiology and Genetics of Neuron and Muscle (INMG-PGNM) CNRS UMR 5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Lise-Marie Donnio
- Pathophysiology and Genetics of Neuron and Muscle (INMG-PGNM) CNRS UMR 5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Damien Neuillet
- Pathophysiology and Genetics of Neuron and Muscle (INMG-PGNM) CNRS UMR 5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - François Vianna
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SDOS/LMDN, Saint-Paul-Lez-Durance, France
| | - Olivier Gabillot
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRAcc, Fontenay-aux-Roses, France
| | - Pierre-Olivier Mari
- Pathophysiology and Genetics of Neuron and Muscle (INMG-PGNM) CNRS UMR 5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Céline Baldeyron
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRAcc, Fontenay-aux-Roses, France
| | - Giuseppina Giglia-Mari
- Pathophysiology and Genetics of Neuron and Muscle (INMG-PGNM) CNRS UMR 5261, INSERM U1315, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
9
|
Endo T. Postnatal skeletal muscle myogenesis governed by signal transduction networks: MAPKs and PI3K-Akt control multiple steps. Biochem Biophys Res Commun 2023; 682:223-243. [PMID: 37826946 DOI: 10.1016/j.bbrc.2023.09.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
Skeletal muscle myogenesis represents one of the most intensively and extensively examined systems of cell differentiation, tissue formation, and regeneration. Muscle regeneration provides an in vivo model system of postnatal myogenesis. It comprises multiple steps including muscle stem cell (or satellite cell) quiescence, activation, migration, myogenic determination, myoblast proliferation, myocyte differentiation, myofiber maturation, and hypertrophy. A variety of extracellular signaling and subsequent intracellular signal transduction pathways or networks govern the individual steps of postnatal myogenesis. Among them, MAPK pathways (the ERK, JNK, p38 MAPK, and ERK5 pathways) and PI3K-Akt signaling regulate multiple steps of myogenesis. Ca2+, cytokine, and Wnt signaling also participate in several myogenesis steps. These signaling pathways often control cell cycle regulatory proteins or the muscle-specific MyoD family and the MEF2 family of transcription factors. This article comprehensively reviews molecular mechanisms of the individual steps of postnatal skeletal muscle myogenesis by focusing on signal transduction pathways or networks. Nevertheless, no or only a partial signaling molecules or pathways have been identified in some responses during myogenesis. The elucidation of these unidentified signaling molecules and pathways leads to an extensive understanding of the molecular mechanisms of myogenesis.
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
10
|
Skrivergaard S, Krøyer Rasmussen M, Sahebekhtiari N, Feveile Young J, Therkildsen M. Satellite cells sourced from bull calves and dairy cows differs in proliferative and myogenic capacity - Implications for cultivated meat. Food Res Int 2023; 173:113217. [PMID: 37803537 DOI: 10.1016/j.foodres.2023.113217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 10/08/2023]
Abstract
Cultivated meat produced with primary muscle satellite cells (SCs) will need a continuous supply of isolated cell material from relevant animal donors. Factors such as age, sex, and breed, along with the sustainability and availability of donor animals, could determine the most appropriate donor type for an efficient production. In this study, we focus on the proliferation and differentiation of bovine SCs isolated from bull calf and dairy cow muscle samples. The proliferative performance of bull calf SCs was significantly better than SCs from dairy cows, however a dynamic differentiation assay revealed that the degree of fusion and formation of myotubes were similar between donor types. Furthermore, the proliferation of SCs from both donor types was enhanced using an in-house developed serum-free media compared to 10% FBS, which also delayed myogenic differentiation and increased final cell population density. Using gene chip transcriptomics, we identified several differentially expressed genes between the two donor types, which could help explain the observed cellular differences. This data also revealed a high biological variance between the three replicate animals within donor type, which seemed to be decreased when using our in-house serum-free media. With the use of the powerful imaging modalities of Cytation 5, we developed a novel high contrast brightfield-enabled label-free myotube quantification method along with a more efficient end-point fusion analysis using Phalloidin-staining. The results give new insights into the bovine SC biology and potential use of bull calves and dairy cows as relevant donor animals for cultivated beef cell sourcing. The newly developed differentiation assays will further enhance future research within the field of cultivated meat and SC biology.
Collapse
|
11
|
Sutcu HH, Montagne B, Ricchetti M. DNA-PKcs regulates myogenesis in an Akt-dependent manner independent of induced DNA damage. Cell Death Differ 2023; 30:1900-1915. [PMID: 37400716 PMCID: PMC10406879 DOI: 10.1038/s41418-023-01177-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 07/05/2023] Open
Abstract
Skeletal muscle regeneration relies on muscle stem (satellite) cells. We previously demonstrated that satellite cells efficiently and accurately repair radiation-induced DNA double-strand breaks (DSBs) via the DNA-dependent kinase DNA-PKcs. We show here that DNA-PKcs affects myogenesis independently of its role in DSB repair. Consequently, this process does not require the accumulation of DSBs and it is also independent of caspase-induced DNA damage. We report that in myogenic cells DNA-PKcs is essential for the expression of the differentiation factor Myogenin in an Akt2-dependent manner. DNA-PKcs interacts with the p300-containing complex that activates Myogenin transcription. We show also that SCID mice that are deficient in DNA-PKcs, and are used for transplantation and muscle regeneration studies, display altered myofiber composition and delayed myogenesis upon injury. These defects are exacerbated after repeated injury/regeneration events resulting in reduced muscle size. We thus identify a novel, caspase-independent, regulation of myogenic differentiation, and define a differentiation phase that does not involve the DNA damage/repair process.
Collapse
Affiliation(s)
- Haser Hasan Sutcu
- Institut Pasteur, Team Stability of Nuclear & Mitochondrial DNA, Department of Developmental and Stem Cell Biology, CNRS UMR3738, 75015, Paris, France
- Université Pierre et Marie Curie (Sorbonne Universities, ED515), Paris, France
- Institut de Radioprotection et de Sûrété Nucléaire (IRSN), Radiobiology of Accidental Exposure Laboratory (PSE-SANTE/SERAMED/LRAcc), B.P. 17, 92262 Fontenay-aux-Roses, Cedex, France
| | - Benjamin Montagne
- Institut Pasteur, Team Stability of Nuclear & Mitochondrial DNA, Department of Developmental and Stem Cell Biology, CNRS UMR3738, 75015, Paris, France
- Institut Pasteur, Molecular Mechanisms of Pathological and Physiological Ageing, Department of Developmental and Stem Cell Biology, Paris, France
| | - Miria Ricchetti
- Institut Pasteur, Team Stability of Nuclear & Mitochondrial DNA, Department of Developmental and Stem Cell Biology, CNRS UMR3738, 75015, Paris, France.
- Institut Pasteur, Molecular Mechanisms of Pathological and Physiological Ageing, Department of Developmental and Stem Cell Biology, Paris, France.
| |
Collapse
|
12
|
Johnson AL, Kamal M, Parise G. The Role of Supporting Cell Populations in Satellite Cell Mediated Muscle Repair. Cells 2023; 12:1968. [PMID: 37566047 PMCID: PMC10417507 DOI: 10.3390/cells12151968] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
Skeletal muscle has a high capacity to repair and remodel in response to damage, largely through the action of resident muscle stem cells, termed satellite cells. Satellite cells are required for the proper repair of skeletal muscle through a process known as myogenesis. Recent investigations have observed relationships between satellite cells and other cell types and structures within the muscle microenvironment. These findings suggest that the crosstalk between inflammatory cells, fibrogenic cells, bone-marrow-derived cells, satellite cells, and the vasculature is essential for the restoration of muscle homeostasis. This review will discuss the influence of the cells and structures within the muscle microenvironment on satellite cell function and muscle repair.
Collapse
Affiliation(s)
| | | | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
13
|
Feng X, Wang AH, Juan AH, Ko KD, Jiang K, Riparini G, Ciuffoli V, Kaba A, Lopez C, Naz F, Jarnik M, Aliberti E, Hu S, Segalés J, Khateb M, Acevedo-Luna N, Randazzo D, Cheung TH, Muñoz-Cánoves P, Dell'Orso S, Sartorelli V. Polycomb Ezh1 maintains murine muscle stem cell quiescence through non-canonical regulation of Notch signaling. Dev Cell 2023; 58:1052-1070.e10. [PMID: 37105173 PMCID: PMC10330238 DOI: 10.1016/j.devcel.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/08/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023]
Abstract
Organismal homeostasis and regeneration are predicated on committed stem cells that can reside for long periods in a mitotically dormant but reversible cell-cycle arrest state defined as quiescence. Premature escape from quiescence is detrimental, as it results in stem cell depletion, with consequent defective tissue homeostasis and regeneration. Here, we report that Polycomb Ezh1 confers quiescence to murine muscle stem cells (MuSCs) through a non-canonical function. In the absence of Ezh1, MuSCs spontaneously exit quiescence. Following repeated injuries, the MuSC pool is progressively depleted, resulting in failure to sustain proper muscle regeneration. Rather than regulating repressive histone H3K27 methylation, Ezh1 maintains gene expression of the Notch signaling pathway in MuSCs. Selective genetic reconstitution of the Notch signaling corrects stem cell number and re-establishes quiescence of Ezh1-/- MuSCs.
Collapse
Affiliation(s)
- Xuesong Feng
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - A Hongjun Wang
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - Aster H Juan
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - Kyung Dae Ko
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - Kan Jiang
- Biodata Mining & Discovery Section, NIAMS, NIH, Bethesda, MD, USA
| | - Giulia Riparini
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - Veronica Ciuffoli
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - Aissah Kaba
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - Christopher Lopez
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - Faiza Naz
- Genomic Technology Section, NIAMS, NIH, Bethesda, MD, USA
| | - Michal Jarnik
- Cell Biology and Neurobiology Branch, NICHD, NIH, Bethesda, MD, USA
| | - Elizabeth Aliberti
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - Shenyuan Hu
- Division of Life Sciences, State Key Laboratory of Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jessica Segalés
- Department of Medicine and Life Sciences (MELIS), Pompeu Fabra University (UPF), Barcelona, Spain
| | - Mamduh Khateb
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | - Natalia Acevedo-Luna
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA
| | | | - Tom H Cheung
- Division of Life Sciences, State Key Laboratory of Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Pura Muñoz-Cánoves
- Department of Medicine and Life Sciences (MELIS), Pompeu Fabra University (UPF), Barcelona, Spain; Altos Labs Inc, San Diego, CA, USA
| | | | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells & Gene Regulation, NIAMS, NIH, Bethesda, MD, USA.
| |
Collapse
|
14
|
Cutler AA, Pawlikowski B, Wheeler JR, Dalla Betta N, Elston T, O’Rourke R, Jones K, Olwin BB. The regenerating skeletal muscle niche drives satellite cell return to quiescence. iScience 2022; 25:104444. [PMID: 35733848 PMCID: PMC9207300 DOI: 10.1016/j.isci.2022.104444] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/27/2022] [Accepted: 05/16/2022] [Indexed: 11/27/2022] Open
Abstract
Skeletal muscle stem cells, or satellite cells (SCs), are essential to regenerate and maintain muscle. Quiescent SCs reside in an asymmetric niche between the basal lamina and myofiber membrane. To repair muscle, SCs activate, proliferate, and differentiate, fusing to repair myofibers or reacquiring quiescence to replenish the SC niche. Little is known about when SCs reacquire quiescence during regeneration or the cellular processes that direct SC fate decisions. We find that most SCs reacquire quiescence 5-10 days after muscle injury, following differentiation and fusion of most cells to regenerate myofibers. Single-cell sequencing of myogenic cells in regenerating muscle identifies SCs reacquiring quiescence and reveals that noncell autonomous signaling networks influence SC fate decisions during regeneration. SC transplantation experiments confirm that the regenerating environment influences SC fate. We define a window for SC repopulation of the niche, emphasizing the temporal contribution of the regenerative muscle environment on SC fate.
Collapse
Affiliation(s)
- Alicia A. Cutler
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Bradley Pawlikowski
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80217, USA
| | - Joshua R. Wheeler
- Departments of Pathology and Neuropathology, Stanford University, Palo Alto, CA 94305, USA
| | - Nicole Dalla Betta
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Tiffany Elston
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Rebecca O’Rourke
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80217, USA
| | - Kenneth Jones
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80217, USA
| | - Bradley B. Olwin
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| |
Collapse
|
15
|
Stout AJ, Mirliani AB, Rittenberg ML, Shub M, White EC, Yuen JSK, Kaplan DL. Simple and effective serum-free medium for sustained expansion of bovine satellite cells for cell cultured meat. Commun Biol 2022; 5:466. [PMID: 35654948 PMCID: PMC9163123 DOI: 10.1038/s42003-022-03423-8] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/27/2022] [Indexed: 11/09/2022] Open
Abstract
Cell-cultured meat offers the potential for a more sustainable, ethical, resilient, and healthy food system. However, research and development has been hindered by the lack of serum-free media that enable the robust expansion of relevant cells (e.g., muscle satellite cells) over multiple passages. Recently, a low-cost serum-free media (B8) was described for pluripotent stem cells. Here, B8 is adapted for bovine satellite cells through the addition of a single component, recombinant albumin, which renders it suitable for long-term satellite cell expansion without sacrificing myogenicity. This new media (Beefy-9) maintains cell growth over the entire period tested (seven passages), with an average doubling time of 39 h. Along with demonstrated efficacy for bovine cells, Beefy-9 offers a promising starting-point for developing serum-free media for other meat-relevant species. Ultimately, this work offers a foundation for escaping cultured meat research's reliance on serum, thereby accelerating the field.
Collapse
Affiliation(s)
- Andrew J Stout
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, Medford, MA, USA
| | - Addison B Mirliani
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, Medford, MA, USA
| | - Miriam L Rittenberg
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, Medford, MA, USA
- Biological Engineering Department, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michelle Shub
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, Medford, MA, USA
| | - Eugene C White
- Tufts Veterinary Field Service, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | - John S K Yuen
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, Medford, MA, USA
| | - David L Kaplan
- Biomedical Engineering Department, Tissue Engineering Resource Center, Tufts University, Medford, MA, USA.
| |
Collapse
|
16
|
Fibrous Protein Composite Scaffolds (3D) for Tissue Regeneration: An in vitro Study on Skeletal Muscle Regeneration. Colloids Surf B Biointerfaces 2022; 217:112656. [DOI: 10.1016/j.colsurfb.2022.112656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/04/2022] [Accepted: 06/21/2022] [Indexed: 11/15/2022]
|
17
|
Bosutti A, Giniatullin A, Odnoshivkina Y, Giudice L, Malm T, Sciancalepore M, Giniatullin R, D'Andrea P, Lorenzon P, Bernareggi A. "Time window" effect of Yoda1-evoked Piezo1 channel activity during mouse skeletal muscle differentiation. Acta Physiol (Oxf) 2021; 233:e13702. [PMID: 34097801 PMCID: PMC9286833 DOI: 10.1111/apha.13702] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022]
Abstract
Aim Mechanosensitive Piezo1 ion channels emerged recently as important contributors to various vital functions including modulation of the blood supply to skeletal muscles. The specific Piezo1 channel agonist Yoda1 was shown to regulate the tone of blood vessels similarly to physical exercise. However, the direct role of Piezo1 channels in muscle function has been little studied so far. We therefore investigated the action of Yoda1 on the functional state of skeletal muscle precursors (satellite cells and myotubes) and on adult muscle fibres. Methods Immunostaining, electrophysiological intracellular recordings and Ca2+ imaging experiments were performed to localize and assess the effect of the chemical activation of Piezo1 channels with Yoda1, on myogenic precursors, adult myofibres and at the adult neuromuscular junction. Results Piezo1 channels were detected by immunostaining in satellite cells (SCs) and myotubes as well as in adult myofibres. In the skeletal muscle precursors, Yoda1 treatment stimulated the differentiation and cell fusion rather than the proliferation of SCs. Moreover, in myotubes, Yoda1 induced significant [Ca2+]i transients, without detectable [Ca2+]i response in adult myofibres. Furthermore, although expression of Piezo1 channels was detected around the muscle endplate region, Yoda1 application did not alter either the nerve‐evoked or spontaneous synaptic activity or muscle contractions in adult myofibres. Conclusion Our data indicate that the chemical activation of Piezo1 channels specifically enhances the differentiation of skeletal muscle precursors, suggesting a possible new strategy to promote muscle regeneration.
Collapse
Affiliation(s)
| | - Arthur Giniatullin
- Department of Physiology Kazan State Medical University Kazan Russia
- Laboratory of Biophysics of Synaptic Processes Kazan Institute of Biochemistry and BiophysicsFederal Research Center “Kazan Scientific Center of RAS” Kazan Russia
| | | | - Luca Giudice
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Marina Sciancalepore
- Department of Life Sciences University of Trieste Trieste Italy
- B.R.A.I.N., University of Trieste Centre for Neuroscience Trieste Italy
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
- Institute of Fundamental Medicine and Biology Federal University Kazan Russia
| | - Paola D'Andrea
- Department of Life Sciences University of Trieste Trieste Italy
| | - Paola Lorenzon
- Department of Life Sciences University of Trieste Trieste Italy
- B.R.A.I.N., University of Trieste Centre for Neuroscience Trieste Italy
| | - Annalisa Bernareggi
- Department of Life Sciences University of Trieste Trieste Italy
- B.R.A.I.N., University of Trieste Centre for Neuroscience Trieste Italy
| |
Collapse
|
18
|
The Role of Satellite Cells in Skeletal Muscle Regeneration-The Effect of Exercise and Age. BIOLOGY 2021; 10:biology10101056. [PMID: 34681155 PMCID: PMC8533525 DOI: 10.3390/biology10101056] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/03/2022]
Abstract
Simple Summary Studies describing the effects of various forms of exercise and age on muscle regeneration were reviewed. Satellite cells are a heterogeneous group of cells that includes stem cells and skeletal muscle progenitor cells. Each skeletal muscle fiber has its own pool of satellite cells that remain inactive until the muscle is damaged. Minor damage within the cell membrane of muscle fibers is patched by fusing intracellular vesicles with the damaged sarcolemma. More severe muscle damage initiates a multistep regeneration process in which satellite cells play an essential role. The condition that initiates the cascade of reactions is the formation of inflammation at the structural discontinuity site, resulting in satellite cell activation. The multitude of reactions and pathways occurring during this process means that many different substances are involved in it and control it. Not all of them are well-understood yet. In parallel, the body’s own population of satellite cells is being rebuilt so that more fibers can be regenerated in the future. Athletes and the elderly are primarily at risk for muscle damage, and pathologies in muscle fiber regeneration cause serious diseases. Abstract The population of satellite cells (mSCs) is highly diversified. The cells comprising it differ in their ability to regenerate their own population and differentiate, as well as in the properties they exhibit. The heterogeneity of this group of cells is evidenced by multiple differentiating markers that enable their recognition, classification, labeling, and characterization. One of the main tasks of satellite cells is skeletal muscle regeneration. Myofibers are often damaged during vigorous exercise in people who participate in sports activities. The number of satellite cells and the speed of the regeneration processes that depend on them affect the time structure of an athlete’s training. This process depends on inflammatory cells. The multitude of reactions and pathways that occur during the regeneration process results in the participation and control of many factors that are activated and secreted during muscle fiber damage and at different stages of its regeneration. However, not all of them are well understood yet. This paper presents the current state of knowledge on satellite cell-dependent skeletal muscle regeneration. Studies describing the effects of various forms of exercise and age on this process were reviewed.
Collapse
|
19
|
Arpke RW, Shams AS, Collins BC, Larson AA, Lu N, Lowe DA, Kyba M. Preservation of satellite cell number and regenerative potential with age reveals locomotory muscle bias. Skelet Muscle 2021; 11:22. [PMID: 34481522 PMCID: PMC8418011 DOI: 10.1186/s13395-021-00277-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/24/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Although muscle regenerative capacity declines with age, the extent to which this is due to satellite cell-intrinsic changes vs. environmental changes has been controversial. The majority of aging studies have investigated hindlimb locomotory muscles, principally the tibialis anterior, in caged sedentary mice, where those muscles are abnormally under-exercised. METHODS We analyze satellite cell numbers in 8 muscle groups representing locomotory and non-locomotory muscles in young and 2-year-old mice and perform transplantation assays of low numbers of hind limb satellite cells from young and old mice. RESULTS We find that satellite cell density does not decline significantly by 2 years of age in most muscles, and one muscle, the masseter, shows a modest but statistically significant increase in satellite cell density with age. The tibialis anterior and extensor digitorum longus were clear exceptions, showing significant declines. We quantify self-renewal using a transplantation assay. Dose dilution revealed significant non-linearity in self-renewal above a very low threshold, suggestive of competition between satellite cells for space within the pool. Assaying within the linear range, i.e., transplanting fewer than 1000 cells, revealed no evidence of decline in cell-autonomous self-renewal or regenerative potential of 2-year-old murine satellite cells. CONCLUSION These data demonstrate the value of comparative muscle analysis as opposed to overreliance on locomotory muscles, which are not used physiologically in aging sedentary mice, and suggest that self-renewal impairment with age is precipitously acquired at the geriatric stage, rather than being gradual over time, as previously thought.
Collapse
Affiliation(s)
- Robert W Arpke
- Lillehei Heart Institute and Department of Pediatrics, Medical School, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
- Present address: Division of Biological Sciences, University of Missouri, 105 Tucker Hall, Columbia, MO, 65211, USA
| | - Ahmed S Shams
- Lillehei Heart Institute and Department of Pediatrics, Medical School, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Brittany C Collins
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, MN, 55455, USA
- Present address: Department of Human Genetics, University of Utah, 15 North 2030 East, Salt Lake City, UT, 84112, USA
| | - Alexie A Larson
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Nguyen Lu
- Lillehei Heart Institute and Department of Pediatrics, Medical School, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
| | - Dawn A Lowe
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Michael Kyba
- Lillehei Heart Institute and Department of Pediatrics, Medical School, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
20
|
Khuu S, Fernandez JW, Handsfield GG. A Coupled Mechanobiological Model of Muscle Regeneration In Cerebral Palsy. Front Bioeng Biotechnol 2021; 9:689714. [PMID: 34513808 PMCID: PMC8429491 DOI: 10.3389/fbioe.2021.689714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/06/2021] [Indexed: 01/05/2023] Open
Abstract
Cerebral palsy is a neuromusculoskeletal disorder associated with muscle weakness, altered muscle architecture, and progressive musculoskeletal symptoms that worsen with age. Pathological changes at the level of the whole muscle have been shown; however, it is unclear why this progression of muscle impairment occurs at the cellular level. The process of muscle regeneration is complex, and the interactions between cells in the muscle milieu should be considered in the context of cerebral palsy. In this work, we built a coupled mechanobiological model of muscle damage and regeneration to explore the process of muscle regeneration in typical and cerebral palsy conditions, and whether a reduced number of satellite cells in the cerebral palsy muscle environment could cause the muscle regeneration cycle to lead to progressive degeneration of muscle. The coupled model consisted of a finite element model of a muscle fiber bundle undergoing eccentric contraction, and an agent-based model of muscle regeneration incorporating satellite cells, inflammatory cells, muscle fibers, extracellular matrix, fibroblasts, and secreted cytokines. Our coupled model simulated damage from eccentric contraction followed by 28 days of regeneration within the muscle. We simulated cyclic damage and regeneration for both cerebral palsy and typically developing muscle milieus. Here we show the nonlinear effects of altered satellite cell numbers on muscle regeneration, where muscle repair is relatively insensitive to satellite cell concentration above a threshold, but relatively sensitive below that threshold. With the coupled model, we show that the fiber bundle geometry undergoes atrophy and fibrosis with too few satellite cells and excess extracellular matrix, representative of the progression of cerebral palsy in muscle. This work uses in silico modeling to demonstrate how muscle degeneration in cerebral palsy may arise from the process of cellular regeneration and a reduced number of satellite cells.
Collapse
Affiliation(s)
- Stephanie Khuu
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Justin W. Fernandez
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
21
|
Kostyuk SV, Proskurnina EV, Ershova ES, Kameneva LV, Malinovskaya EM, Savinova EA, Sergeeva VA, Umriukhin PE, Dolgikh OA, Khakina EA, Kraevaya OA, Troshin PA, Kutsev SI, Veiko NN. The Phosphonate Derivative of C 60 Fullerene Induces Differentiation towards the Myogenic Lineage in Human Adipose-Derived Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22179284. [PMID: 34502190 PMCID: PMC8431706 DOI: 10.3390/ijms22179284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 12/26/2022] Open
Abstract
Inductors of myogenic stem cell differentiation attract attention, as they can be used to treat myodystrophies and post-traumatic injuries. Functionalization of fullerenes makes it possible to obtain water-soluble derivatives with targeted biochemical activity. This study examined the effects of the phosphonate C60 fullerene derivatives on the expression of myogenic transcription factors and myogenic differentiation of human mesenchymal stem cells (MSCs). Uptake of the phosphonate C60 fullerene derivatives in human MSCs, intracellular ROS visualization, superoxide scavenging potential, and the expression of myogenic, adipogenic, and osteogenic differentiation genes were studied. The prolonged MSC incubation (within 7–14 days) with the C60 pentaphoshonate potassium salt promoted their differentiation towards the myogenic lineage. The transcription factors and gene expressions determining myogenic differentiation (MYOD1, MYOG, MYF5, and MRF4) increased, while the expression of osteogenic differentiation factors (BMP2, BMP4, RUNX2, SPP1, and OCN) and adipogenic differentiation factors (CEBPB, LPL, and AP2 (FABP4)) was reduced or did not change. The stimulation of autophagy may be one of the factors contributing to the increased expression of myogenic differentiation genes in MSCs. Autophagy may be caused by intracellular alkalosis and/or short-term intracellular oxidative stress.
Collapse
Affiliation(s)
- Svetlana V. Kostyuk
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Elena V. Proskurnina
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
- Correspondence:
| | - Elizaveta S. Ershova
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Larisa V. Kameneva
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Elena M. Malinovskaya
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Ekaterina A. Savinova
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Vasilina A. Sergeeva
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Pavel E. Umriukhin
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
- Department of Normal Physiology, I.M. Sechenov First Moscow State Medical University (Sechenov University) , Mohovaya Str. 11-4, 125009 Moscow, Russia
| | - Olga A. Dolgikh
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Ekaterina A. Khakina
- A.N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavylova St. 28, B-334, 119991 Moscow, Russia;
| | - Olga A. Kraevaya
- Institute of Problems of Chemical Physics of Russian Academy of Sciences, Semenov Prospect 1, 142432 Chernogolovka (Moscow Region), Russia; (O.A.K.); (P.A.T.)
| | - Pavel A. Troshin
- Institute of Problems of Chemical Physics of Russian Academy of Sciences, Semenov Prospect 1, 142432 Chernogolovka (Moscow Region), Russia; (O.A.K.); (P.A.T.)
| | - Sergey I. Kutsev
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Natalia N. Veiko
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| |
Collapse
|
22
|
Gutiérrez J, Gonzalez D, Escalona-Rivano R, Takahashi C, Brandan E. Reduced RECK levels accelerate skeletal muscle differentiation, improve muscle regeneration, and decrease fibrosis. FASEB J 2021; 35:e21503. [PMID: 33811686 DOI: 10.1096/fj.202001646rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/07/2021] [Accepted: 02/19/2021] [Indexed: 12/15/2022]
Abstract
The muscle regeneration process requires a properly assembled extracellular matrix (ECM). Its homeostasis depends on the activity of different matrix-metalloproteinases (MMPs). The reversion-inducing-cysteine-rich protein with kazal motifs (RECK) is a membrane-anchored protein that negatively regulates the activity of different MMPs. However, the role of RECK in the process of skeletal muscle differentiation, regeneration, and fibrosis has not been elucidated. Here, we show that during skeletal muscle differentiation of C2C12 myoblasts and in satellite cells on isolated muscle fibers, RECK is transiently up regulated. C2C12 myoblasts with reduced RECK levels are more prone to enter the differentiation program, showing an accelerated differentiation process. Notch-1 signaling was reduced, while p38 and AKT signaling were augmented in myoblasts with decreased RECK levels. Overexpression of RECK restores the normal differentiation process but diminished the ability to form myotubes. Transient up-regulation of RECK occurs during skeletal muscle regeneration, which was accelerated in RECK-deficient mice (Reck±). RECK, MMPs and ECM proteins augmented in chronically damaged WT muscle, a model of muscle fibrosis. In this model, RECK ± mice showed diminished fibrosis compared to WT. These results strongly suggest that RECK is acting as a potential myogenic repressor during muscle formation and regeneration, emerging as a new player in these processes, and as a potential target to treat individuals with the muscle-wasting disease.
Collapse
Affiliation(s)
- Jaime Gutiérrez
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile.,Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David Gonzalez
- Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Escalona-Rivano
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile
| | - Chiaki Takahashi
- Oncology and Molecular Biology, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Enrique Brandan
- Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Fundación Ciencia & Vida, Santiago, Chile
| |
Collapse
|
23
|
Nuge T, Liu Z, Liu X, Ang BC, Andriyana A, Metselaar HSC, Hoque ME. Recent Advances in Scaffolding from Natural-Based Polymers for Volumetric Muscle Injury. Molecules 2021; 26:699. [PMID: 33572728 PMCID: PMC7865392 DOI: 10.3390/molecules26030699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/03/2021] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Volumetric Muscle Loss (VML) is associated with muscle loss function and often untreated and considered part of the natural sequelae of trauma. Various types of biomaterials with different physical and properties have been developed to treat VML. However, much work remains yet to be done before the scaffolds can pass from the bench to the bedside. The present review aims to provide a comprehensive summary of the latest developments in the construction and application of natural polymers-based tissue scaffolding for volumetric muscle injury. Here, the tissue engineering approaches for treating volumetric muscle loss injury are highlighted and recent advances in cell-based therapies using various sources of stem cells are elaborated in detail. An overview of different strategies of tissue scaffolding and their efficacy on skeletal muscle cells regeneration and migration are presented. Furthermore, the present paper discusses a wide range of natural polymers with a special focus on proteins and polysaccharides that are major components of the extracellular matrices. The natural polymers are biologically active and excellently promote cell adhesion and growth. These bio-characteristics justify natural polymers as one of the most attractive options for developing scaffolds for muscle cell regeneration.
Collapse
Affiliation(s)
- Tamrin Nuge
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, 199 Taikang East Road, Ningbo 315100, China; (T.N.); (Z.L.)
| | - Ziqian Liu
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, 199 Taikang East Road, Ningbo 315100, China; (T.N.); (Z.L.)
| | - Xiaoling Liu
- Department of Mechanical, Materials and Manufacturing Engineering, Faculty of Science and Engineering, University of Nottingham Ningbo China, 199 Taikang East Road, Ningbo 315100, China; (T.N.); (Z.L.)
| | - Bee Chin Ang
- Centre of Advanced Materials, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.A.); (H.S.C.M.)
- Department of Chemical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Andri Andriyana
- Centre of Advanced Materials, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.A.); (H.S.C.M.)
- Department of Mechanical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Hendrik Simon Cornelis Metselaar
- Centre of Advanced Materials, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.A.); (H.S.C.M.)
- Department of Mechanical Engineering, Faculty of Engineering, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Md Enamul Hoque
- Department of Biomedical Engineering, Military Institute of Science and Technology (MIST), Dhaka 1216, Bangladesh;
| |
Collapse
|
24
|
Barlow J, Sfyri PP, Mitchell R, Verpoorten S, Scully D, Andreou C, Papadopoulos P, Patel K, Matsakas A. Platelet releasate normalises the compromised muscle regeneration in a mouse model of hyperlipidaemia. Exp Physiol 2021; 106:700-713. [PMID: 33450106 DOI: 10.1113/ep088937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the impact of obesity-independent hyperlipidaemia on skeletal muscle stem cell function of ApoE-deficient (ApoE-/- ) mice? What is the main finding and its importance? Compromised muscle stem cell function accounts for the impaired muscle regeneration in hyperlipidaemic ApoE-/- mice. Importantly, impaired muscle regeneration is normalised by administration of platelet releasate. ABSTRACT Muscle satellite cells are important stem cells for skeletal muscle regeneration and repair after injury. ApoE-deficient mice, an established mouse model of hyperlipidaemia and atherosclerosis, show evidence of oxidative stress-induced lesions and fat infiltration in skeletal muscle followed by impaired repair after injury. However, the mechanisms underpinning attenuated muscle regeneration remain to be fully defined. Key to addressing the latter is to understand the properties of muscle stem cells from ApoE-deficient mice and their myogenic potential. Muscle stem cells from ApoE-deficient mice were cultured both ex vivo (on single fibres) and in vitro (primary myoblasts) and their myogenic capacity was determined. Skeletal muscle regeneration was studied on days 5 and 10 after cardiotoxin injury. ApoE-deficient muscle stem cells showed delayed activation and differentiation on single muscle fibres ex vivo. Impaired proliferation and differentiation profiles were also evident on isolated primary muscle stem cells in culture. ApoE-deficient mice displayed impaired skeletal muscle regeneration after acute injury in vivo. Administration of platelet releasate in ApoE-deficient mice reversed the deficits of muscle regeneration after acute injury to wild-type levels. These findings indicate that muscle stem cell myogenic potential is perturbed in skeletal muscle of a mouse model of hyperlipidaemia. We propose that platelet releasate could be a therapeutic intervention for conditions with associated myopathy such as peripheral arterial disease.
Collapse
Affiliation(s)
- Joseph Barlow
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Pagona Panagiota Sfyri
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Rob Mitchell
- School of Biological Sciences, University of Reading, Reading, UK
| | - Sandrine Verpoorten
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - David Scully
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Charalampos Andreou
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Petros Papadopoulos
- Department of Hematology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| |
Collapse
|
25
|
Tan CM, Najib NAM, Suhaimi NF, Halid NA, Cho VV, Abdullah SI, Ismail MZ, Khor SC, Jaafar F, Makpol S. Modulation of Ki67 and myogenic regulatory factor expression by tocotrienol-rich fraction ameliorates myogenic program of senescent human myoblasts. Arch Med Sci 2021; 17:752-763. [PMID: 34025846 PMCID: PMC8130490 DOI: 10.5114/aoms.2019.85449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/08/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Replicative senescence results in dysregulation of cell proliferation and differentiation, which plays a role in the regenerative defects observed during age-related muscle atrophy. Vitamin E is a well-known antioxidant, which potentially ameliorates a wide range of age-related manifestations. The aim of this study was to determine the effects of tocotrienol-rich fraction (TRF) in modulating the expression of proliferation- and differentiation-associated proteins in senescent human myoblasts during the differentiation phase. MATERIAL AND METHODS Human skeletal muscle myoblasts were cultured until senescence. Young and senescent cells were treated with TRF for 24 h before and after differentiation induction, followed by evaluation of cellular morphology and efficiency of differentiation. Expression of cell proliferation marker Ki67 protein and myogenic regulatory factors MyoD and myogenin were determined. RESULTS Our findings showed that treatment with TRF significantly improved the morphology of senescent myoblasts. Promotion of differentiation was observed in young and senescent myoblasts with TRF treatment as shown by the increased fusion index and larger size of myotubes. Increased Ki67 and myogenin expression with TRF treatment was also observed in senescent myoblasts, suggesting amelioration of the myogenic program by TRF during replicative senescence. CONCLUSIONS TRF modulates the expression of regulatory factors related to proliferation and differentiation in senescent human myoblasts and could be beneficial for ameliorating the regenerative defects during aging.
Collapse
Affiliation(s)
- Chun Min Tan
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nadwa Aqeela Mohd Najib
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nur Farahin Suhaimi
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nur Alia Halid
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Vi Vien Cho
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Saiful Idham Abdullah
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Muhammad Zulhilmi Ismail
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shy Cian Khor
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Faizul Jaafar
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
26
|
Tidball JG, Flores I, Welc SS, Wehling-Henricks M, Ochi E. Aging of the immune system and impaired muscle regeneration: A failure of immunomodulation of adult myogenesis. Exp Gerontol 2020; 145:111200. [PMID: 33359378 DOI: 10.1016/j.exger.2020.111200] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/17/2020] [Accepted: 12/08/2020] [Indexed: 12/16/2022]
Abstract
Skeletal muscle regeneration that follows acute injury is strongly influenced by interactions with immune cells that invade and proliferate in the damaged tissue. Discoveries over the past 20 years have identified many of the key mechanisms through which myeloid cells, especially macrophages, regulate muscle regeneration. In addition, lymphoid cells that include CD8+ T-cells and regulatory T-cells also significantly affect the course of muscle regeneration. During aging, the regenerative capacity of skeletal muscle declines, which can contribute to progressive loss of muscle mass and function. Those age-related reductions in muscle regeneration are accompanied by systemic, age-related changes in the immune system, that affect many of the myeloid and lymphoid cell populations that can influence muscle regeneration. In this review, we present recent discoveries that indicate that aging of the immune system contributes to the diminished regenerative capacity of aging muscle. Intrinsic, age-related changes in immune cells modify their expression of factors that affect the function of a population of muscle stem cells, called satellite cells, that are necessary for normal muscle regeneration. For example, age-related reductions in the expression of growth differentiation factor-3 (GDF3) or CXCL10 by macrophages negatively affect adult myogenesis, by disrupting regulatory interactions between macrophages and satellite cells. Those changes contribute to a reduction in the numbers and myogenic capacity of satellite cells in old muscle, which reduces their ability to restore damaged muscle. In addition, aging produces changes in the expression of molecules that regulate the inflammatory response to injured muscle, which also contributes to age-related defects in muscle regeneration. For example, age-related increases in the production of osteopontin by macrophages disrupts the normal inflammatory response to muscle injury, resulting in regenerative defects. These nascent findings represent the beginning of a newly-developing field of investigation into mechanisms through which aging of the immune system affects muscle regeneration.
Collapse
Affiliation(s)
- James G Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, United States of America; Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, United States of America; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, United States of America.
| | - Ivan Flores
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, United States of America
| | - Steven S Welc
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, United States of America
| | - Eisuke Ochi
- Hosei University, Faculty of Bioscience and Applied Chemistry, 3-7-2, Kajino, Koganei, Tokyo 184-8584, Japan
| |
Collapse
|
27
|
Immunofluorescence Evaluation of Myf5 and MyoD in Masseter Muscle of Unilateral Posterior Crossbite Patients. J Funct Morphol Kinesiol 2020; 5:jfmk5040080. [PMID: 33467295 PMCID: PMC7739332 DOI: 10.3390/jfmk5040080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/04/2020] [Accepted: 11/04/2020] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED A unilateral posterior crossbite is a malocclusion where the low activity of the affected masseter muscle is compensated by the contralateral muscle hypertrophy. It is still unknown if, in the same condition, myogenesis with new fibre formation takes place. AIM the aim of the present study was to evaluate the expression of myogenesis markers, such as Myf5 and MyoD, in masseter muscles of unilateral posterior crossbite patients. MATERIALS AND METHODS biopsies from fifteen surgical patients with unilateral posterior crossbites have been analysed by immunofluorescence reactions. The results show the expression of Myf5 and MyoD in the contralateral muscle but not in the ipsilateral one. Moreover, statistical analysis shows the higher number of satellite cells in the contralateral side if compared to the ipsilateral one. CONCLUSIONS these results suggest that in contralateral muscle, hyperplastic events take place, as well as hypertrophy.
Collapse
|
28
|
Ganassi M, Badodi S, Wanders K, Zammit PS, Hughes SM. Myogenin is an essential regulator of adult myofibre growth and muscle stem cell homeostasis. eLife 2020; 9:e60445. [PMID: 33001028 PMCID: PMC7599067 DOI: 10.7554/elife.60445] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Growth and maintenance of skeletal muscle fibres depend on coordinated activation and return to quiescence of resident muscle stem cells (MuSCs). The transcription factor Myogenin (Myog) regulates myocyte fusion during development, but its role in adult myogenesis remains unclear. In contrast to mice, myog-/-zebrafish are viable, but have hypotrophic muscles. By isolating adult myofibres with associated MuSCs, we found that myog-/- myofibres have severely reduced nuclear number, but increased myonuclear domain size. Expression of fusogenic genes is decreased, Pax7 upregulated, MuSCs are fivefold more numerous and mis-positioned throughout the length of myog-/-myofibres instead of localising at myofibre ends as in wild-type. Loss of Myog dysregulates mTORC1 signalling, resulting in an 'alerted' state of MuSCs, which display precocious activation and faster cell cycle entry ex vivo, concomitant with myod upregulation. Thus, beyond controlling myocyte fusion, Myog influences the MuSC:niche relationship, demonstrating a multi-level contribution to muscle homeostasis throughout life.
Collapse
Affiliation(s)
- Massimo Ganassi
- Randall Centre for Cell and Molecular Biophysics, King’s College LondonLondonUnited Kingdom
| | - Sara Badodi
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUnited Kingdom
| | - Kees Wanders
- Randall Centre for Cell and Molecular Biophysics, King’s College LondonLondonUnited Kingdom
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King’s College LondonLondonUnited Kingdom
| | - Simon M Hughes
- Randall Centre for Cell and Molecular Biophysics, King’s College LondonLondonUnited Kingdom
| |
Collapse
|
29
|
Gattazzo F, Laurent B, Relaix F, Rouard H, Didier N. Distinct Phases of Postnatal Skeletal Muscle Growth Govern the Progressive Establishment of Muscle Stem Cell Quiescence. Stem Cell Reports 2020; 15:597-611. [PMID: 32763161 PMCID: PMC7486220 DOI: 10.1016/j.stemcr.2020.07.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 02/01/2023] Open
Abstract
Muscle stem cells (or muscle satellite cells [MuSCs]) are required for postnatal growth. Yet, the detailed characterization of myogenic progression and establishment of quiescence during this process remains poorly documented. Here, we provide an overview of myogenic cells heterogeneity and dynamic from birth to adulthood using flow cytometry. We demonstrated that PAX7+ cells acquire an increasing ability to progress in the myogenic program from birth to adulthood. We then simultaneously analyzed the cycling state (KI67 expression) of the MuSCs and progenitors (PAX7+) and their progression into myogenic precursors (PAX7−MYOD+) and differentiating cells (MYOG+) in vivo. We identified two distinct peaks of myogenic differentiation between P7–P10 and P21–P28, and showed that the quiescent MuSC pool is established between 7 and 8 weeks of age. Overall our study provides a comprehensive in vivo characterization of myogenic heterogeneity and demonstrates the highly dynamic nature of skeletal muscle postnatal growth process. Quiescent MuSC pool is fully established between 7 and 8 weeks after birth Postnatal growth comprises two successive phases of differentiation Identification of the complex cellular heterogeneity during myogenic progression Redefinition of the progression of myogenic cells into the differentiation program
Collapse
Affiliation(s)
| | - Béatrice Laurent
- Univ Paris Est Creteil, INSERM, EFS, IMRB, Creteil 94010, France
| | - Frédéric Relaix
- Univ Paris Est Creteil, INSERM, EFS, IMRB, Creteil 94010, France; EnvA, IMRB, Maisons-Alfort 94700, France; AP-HP, Hopital Mondor, Service d'Histologie, Creteil 94010, France.
| | - Hélène Rouard
- Univ Paris Est Creteil, INSERM, EFS, IMRB, Creteil 94010, France
| | - Nathalie Didier
- Univ Paris Est Creteil, INSERM, EFS, IMRB, Creteil 94010, France.
| |
Collapse
|
30
|
Chan SMH, Cerni C, Passey S, Seow HJ, Bernardo I, van der Poel C, Dobric A, Brassington K, Selemidis S, Bozinovski S, Vlahos R. Cigarette Smoking Exacerbates Skeletal Muscle Injury without Compromising Its Regenerative Capacity. Am J Respir Cell Mol Biol 2020; 62:217-230. [PMID: 31461300 DOI: 10.1165/rcmb.2019-0106oc] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle dysfunction in patients with chronic obstructive pulmonary disease negatively impacts quality of life and survival. Cigarette smoking (CS) is the major risk factor for chronic obstructive pulmonary disease and skeletal muscle dysfunction; however, how CS affects skeletal muscle function remains enigmatic. To examine the impact of CS on skeletal muscle inflammation and regeneration, male BALB/c mice were exposed to CS for 8 weeks before muscle injury was induced by barium chloride injection, and were maintained on the CS protocol for up to 21 days after injury. Barium chloride injection resulted in architectural damage to the tibialis anterior muscle, resulting in a decrease contractile function, which was worsened by CS exposure. CS exposure caused muscle atrophy (reduction in gross weight and myofiber cross-sectional area) and altered fiber type composition (31% reduction of oxidative fibers). Both contractile function and loss in myofiber cross-sectional area by CS exposure gradually recovered over time. Satellite cells are muscle stem cells that confer skeletal muscle the plasticity to adapt to changing demands. CS exposure blunted Pax7+ centralized nuclei within satellite cells and thus prevented the activation of these muscle stem cells. Finally, CS triggered muscle inflammation; in particular, there was an exacerbated recruitment of F4/80+ monocytic cells to the site of injury along with enhanced proinflammatory cytokine expression. In conclusion, CS exposure amplified the local inflammatory response at the site of skeletal muscle injury, and this was associated with impaired satellite cell activation, leading to a worsened muscle injury and contractile function without detectable impacts on the recovery outcomes.
Collapse
Affiliation(s)
- Stanley M H Chan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Claudia Cerni
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Samantha Passey
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Huei Jiunn Seow
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Ivan Bernardo
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Chris van der Poel
- Department of Physiology, Anatomy & Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Aleksandar Dobric
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Kurt Brassington
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia; and
| |
Collapse
|
31
|
The micropeptide LEMP plays an evolutionarily conserved role in myogenesis. Cell Death Dis 2020; 11:357. [PMID: 32393776 PMCID: PMC7214441 DOI: 10.1038/s41419-020-2570-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 01/16/2023]
Abstract
In recent years, micropeptides have been increasingly identified as important regulators in various biological processes. However, whether micropeptides are functionally conserved remains largely unknown. Here, we uncovered a micropeptide with evolutionarily conserved roles in myogenesis. RNA-seq data analysis of proliferating mouse satellite cells (SCs) and differentiated myotubes identified a previously annotated lncRNA, MyolncR4 (1500011K16RIK), which is upregulated during muscle differentiation. Significantly, MyolncR4 is highly conserved across vertebrate species. Multiple lines of evidence demonstrate that MyolncR4 encodes a 56-aa micropeptide, which was named as LEMP (lncRNA encoded micropeptide). LEMP promotes muscle formation and regeneration in mouse. In zebrafish, MyolncR4 is enriched in developing somites and elimination of LEMP results in impaired muscle development, which could be efficiently rescued by expression of the mouse LEMP. Interestingly, LEMP is localized at both the plasma membrane and mitochondria, and associated with multiple mitochondrial proteins, suggestive of its involvement in mitochondrial functions. Together, our work uncovers a micropeptide that plays an evolutionarily conserved role in skeletal muscle differentiation, pinpointing the functional importance of this growing family of small peptides.
Collapse
|
32
|
Hillege MMG, Galli Caro RA, Offringa C, de Wit GMJ, Jaspers RT, Hoogaars WMH. TGF-β Regulates Collagen Type I Expression in Myoblasts and Myotubes via Transient Ctgf and Fgf-2 Expression. Cells 2020; 9:E375. [PMID: 32041253 PMCID: PMC7072622 DOI: 10.3390/cells9020375] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 12/27/2022] Open
Abstract
Transforming Growth Factor β (TGF-β) is involved in fibrosis as well as the regulation of muscle mass, and contributes to the progressive pathology of muscle wasting disorders. However, little is known regarding the time-dependent signalling of TGF-β in myoblasts and myotubes, as well as how TGF-β affects collagen type I expression and the phenotypes of these cells. Here, we assessed effects of TGF-β on gene expression in C2C12 myoblasts and myotubes after 1, 3, 9, 24 and 48 h treatment. In myoblasts, various myogenic genes were repressed after 9, 24 and 48 h, while in myotubes only a reduction in Myh3 expression was observed. In both myoblasts and myotubes, TGF-β acutely induced the expression of a subset of genes involved in fibrosis, such as Ctgf and Fgf-2, which was subsequently followed by increased expression of Col1a1. Knockdown of Ctgf and Fgf-2 resulted in a lower Col1a1 expression level. Furthermore, the effects of TGF-β on myogenic and fibrotic gene expression were more pronounced than those of myostatin, and knockdown of TGF-β type I receptor Tgfbr1, but not receptor Acvr1b, resulted in a reduction in Ctgf and Col1a1 expression. These results indicate that, during muscle regeneration, TGF-β induces fibrosis via Tgfbr1 by stimulating the autocrine signalling of Ctgf and Fgf-2.
Collapse
Affiliation(s)
| | | | | | | | - Richard T. Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 BT Amsterdam, The Netherlands; (M.M.G.H.); (R.A.G.C.); (C.O.); (G.M.J.d.W.); (W.M.H.H.)
| | | |
Collapse
|
33
|
Fiore PF, Benedetti A, Sandonà M, Madaro L, De Bardi M, Saccone V, Puri PL, Gargioli C, Lozanoska-Ochser B, Bouché M. Lack of PKCθ Promotes Regenerative Ability of Muscle Stem Cells in Chronic Muscle Injury. Int J Mol Sci 2020; 21:ijms21030932. [PMID: 32023816 PMCID: PMC7037041 DOI: 10.3390/ijms21030932] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disease characterized by muscle wasting and chronic inflammation, leading to impaired satellite cells (SCs) function and exhaustion of their regenerative capacity. We previously showed that lack of PKCθ in mdx mice, a mouse model of DMD, reduces muscle wasting and inflammation, and improves muscle regeneration and performance at early stages of the disease. In this study, we show that muscle regeneration is boosted, and fibrosis reduced in mdxθ−/− mice, even at advanced stages of the disease. This phenotype was associated with a higher number of Pax7 positive cells in mdxθ−/− muscle compared with mdx muscle, during the progression of the disease. Moreover, the expression level of Pax7 and Notch1, the pivotal regulators of SCs self-renewal, were upregulated in SCs isolated from mdxθ−/− muscle compared with mdx derived SCs. Likewise, the expression of the Notch ligands Delta1 and Jagged1 was higher in mdxθ−/− muscle compared with mdx. The expression level of Delta1 and Jagged1 was also higher in PKCθ−/− muscle compared with WT muscle following acute injury. In addition, lack of PKCθ prolonged the survival and sustained the differentiation of transplanted myogenic progenitors. Overall, our results suggest that lack of PKCθ promotes muscle repair in dystrophic mice, supporting stem cells survival and maintenance through increased Delta-Notch signaling.
Collapse
MESH Headings
- Animals
- Cardiotoxins/adverse effects
- Cell Differentiation
- Cells, Cultured
- Male
- Mice
- Mice, Inbred mdx
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/injuries
- Muscle, Skeletal/physiology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/therapy
- PAX7 Transcription Factor/metabolism
- Protein Kinase C-theta/genetics
- Receptor, Notch1/metabolism
- Regeneration
- Signal Transduction
- Stem Cell Transplantation
- Stem Cells/cytology
- Stem Cells/drug effects
- Stem Cells/metabolism
Collapse
Affiliation(s)
- Piera Filomena Fiore
- Department of AHFMO, University of Rome “la Sapienza”, Via A. Scarpa 14, 00161 Rome, Italy; (P.F.F.); (A.B.); (M.S.); (L.M.); (B.L.-O.)
| | - Anna Benedetti
- Department of AHFMO, University of Rome “la Sapienza”, Via A. Scarpa 14, 00161 Rome, Italy; (P.F.F.); (A.B.); (M.S.); (L.M.); (B.L.-O.)
| | - Martina Sandonà
- Department of AHFMO, University of Rome “la Sapienza”, Via A. Scarpa 14, 00161 Rome, Italy; (P.F.F.); (A.B.); (M.S.); (L.M.); (B.L.-O.)
- IRCCS Fondazione Santa Lucia (FSL), e00143 Rome, Italy; (M.D.B.); (V.S.)
| | - Luca Madaro
- Department of AHFMO, University of Rome “la Sapienza”, Via A. Scarpa 14, 00161 Rome, Italy; (P.F.F.); (A.B.); (M.S.); (L.M.); (B.L.-O.)
- IRCCS Fondazione Santa Lucia (FSL), e00143 Rome, Italy; (M.D.B.); (V.S.)
| | - Marco De Bardi
- IRCCS Fondazione Santa Lucia (FSL), e00143 Rome, Italy; (M.D.B.); (V.S.)
| | - Valentina Saccone
- IRCCS Fondazione Santa Lucia (FSL), e00143 Rome, Italy; (M.D.B.); (V.S.)
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA;
| | - Cesare Gargioli
- Department of Biology, Tor Vergata University, 00133 Rome, Italy;
| | - Biliana Lozanoska-Ochser
- Department of AHFMO, University of Rome “la Sapienza”, Via A. Scarpa 14, 00161 Rome, Italy; (P.F.F.); (A.B.); (M.S.); (L.M.); (B.L.-O.)
| | - Marina Bouché
- Department of AHFMO, University of Rome “la Sapienza”, Via A. Scarpa 14, 00161 Rome, Italy; (P.F.F.); (A.B.); (M.S.); (L.M.); (B.L.-O.)
- Correspondence: ; Tel.: +39-06-4976-6755
| |
Collapse
|
34
|
Vézina F, Cornelius Ruhs E, O'Connor ES, Le Pogam A, Régimbald L, Love OP, Jimenez AG. Consequences of being phenotypically mismatched with the environment: rapid muscle ultrastructural changes in cold-shocked black-capped chickadees ( Poecile atricapillus). Am J Physiol Regul Integr Comp Physiol 2019; 318:R274-R283. [PMID: 31823671 DOI: 10.1152/ajpregu.00203.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Phenotypic flexibility has received considerable attention in the last decade; however, whereas many studies have reported amplitude of variation in phenotypic traits, much less attention has focused on the rate at which traits can adjust in response to sudden changes in the environment. We investigated whole animal and muscle phenotypic changes occurring in black-capped chickadees (Poecile atricapillus) acclimated to cold (-5°C) and warm (20°C) temperatures in the first 3 h following a 15°C temperature drop (over 3 h). Before the temperature change, cold-acclimated birds were consuming 95% more food, were carrying twice as much body fat, and had 23% larger pectoralis muscle fiber diameters than individuals kept at 20°C. In the 3 h following the temperature drop, these same birds altered their pectoralis muscle ultrastructure by increasing the number of capillaries per fiber area and the number of nuclei per millimeter of fiber by 22%, consequently leading to a 22% decrease in myonuclear domain (amount of cytoplasm serviced per nucleus), whereas no such changes were observed in the warm-acclimated birds. To our knowledge, this is the first demonstration of such a rapid adjustment in muscle fiber ultrastructure in vertebrates. These results support the hypothesis that chickadees maintaining a cold phenotype are better prepared than warm-phenotype individuals to respond to a sudden decline in temperature, such as what may be experienced in their natural wintering environment.
Collapse
Affiliation(s)
- François Vézina
- Départment de Biologie, Chimie et Géographie, Groupe de Recherche sur les Environnements Nordiques BORÉAS, Centre d'études Nordiques, Centre de la Science de la Biodiversité du Québec Université du Québec à Rimouski, Québec, Canada
| | - Emily Cornelius Ruhs
- Départment de Biologie, Chimie et Géographie, Groupe de Recherche sur les Environnements Nordiques BORÉAS, Centre d'études Nordiques, Centre de la Science de la Biodiversité du Québec Université du Québec à Rimouski, Québec, Canada
| | - Erin S O'Connor
- Department of Biology, Colgate University, Hamilton, New York
| | - Audrey Le Pogam
- Départment de Biologie, Chimie et Géographie, Groupe de Recherche sur les Environnements Nordiques BORÉAS, Centre d'études Nordiques, Centre de la Science de la Biodiversité du Québec Université du Québec à Rimouski, Québec, Canada
| | - Lyette Régimbald
- Départment de Biologie, Chimie et Géographie, Groupe de Recherche sur les Environnements Nordiques BORÉAS, Centre d'études Nordiques, Centre de la Science de la Biodiversité du Québec Université du Québec à Rimouski, Québec, Canada
| | - Oliver P Love
- Department of Integrative Biology, University of Windsor, Ontario, Canada
| | | |
Collapse
|
35
|
Sokołowska E, Błachnio-Zabielska AU. A Critical Review of Electroporation as A Plasmid Delivery System in Mouse Skeletal Muscle. Int J Mol Sci 2019; 20:ijms20112776. [PMID: 31174257 PMCID: PMC6600476 DOI: 10.3390/ijms20112776] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022] Open
Abstract
The gene delivery to skeletal muscles is a promising strategy for the treatment of both muscular disorders (by silencing or overexpression of specific gene) and systemic secretion of therapeutic proteins. The use of a physical method like electroporation with plate or needle electrodes facilitates long-lasting gene silencing in situ. It has been reported that electroporation enhances the expression of the naked DNA gene in the skeletal muscle up to 100 times and decreases the changeability of the intramuscular expression. Coelectransfer of reporter genes such as green fluorescent protein (GFP), luciferase or beta-galactosidase allows the observation of correctly performed silencing in the muscles. Appropriate selection of plasmid injection volume and concentration, as well as electrotransfer parameters, such as the voltage, the length and the number of electrical pulses do not cause long-term damage to myocytes. In this review, we summarized the electroporation methodology as well as the procedure of electrotransfer to the gastrocnemius, tibialis, soleus and foot muscles and compare their advantages and disadvantages.
Collapse
Affiliation(s)
- Emilia Sokołowska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 15-222 Bialystok, Poland.
| | | |
Collapse
|
36
|
Dell'Orso S, Juan AH, Ko KD, Naz F, Perovanovic J, Gutierrez-Cruz G, Feng X, Sartorelli V. Single cell analysis of adult mouse skeletal muscle stem cells in homeostatic and regenerative conditions. Development 2019; 146:dev.174177. [PMID: 30890574 DOI: 10.1242/dev.174177] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/07/2019] [Indexed: 12/15/2022]
Abstract
Dedicated stem cells ensure postnatal growth, repair and homeostasis of skeletal muscle. Following injury, muscle stem cells (MuSCs) exit from quiescence and divide to reconstitute the stem cell pool and give rise to muscle progenitors. The transcriptomes of pooled MuSCs have provided a rich source of information for describing the genetic programs of distinct static cell states; however, bulk microarray and RNA sequencing provide only averaged gene expression profiles, blurring the heterogeneity and developmental dynamics of asynchronous MuSC populations. Instead, the granularity required to identify distinct cell types, states, and their dynamics can be afforded by single cell analysis. We were able to compare the transcriptomes of thousands of MuSCs and primary myoblasts isolated from homeostatic or regenerating muscles by single cell RNA sequencing. Using computational approaches, we could reconstruct dynamic trajectories and place, in a pseudotemporal manner, the transcriptomes of individual MuSC within these trajectories. This approach allowed for the identification of distinct clusters of MuSCs and primary myoblasts with partially overlapping but distinct transcriptional signatures, as well as the description of metabolic pathways associated with defined MuSC states.
Collapse
Affiliation(s)
- Stefania Dell'Orso
- Genome Technology Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA stefania.dell'
| | - Aster H Juan
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Kyung-Dae Ko
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Faiza Naz
- Genome Technology Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Jelena Perovanovic
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Gustavo Gutierrez-Cruz
- Genome Technology Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Xuesong Feng
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, MD 208292, USA stefania.dell'
| |
Collapse
|
37
|
A transient protective effect of low-level laser irradiation against disuse-induced atrophy of rats. Lasers Med Sci 2019; 34:1829-1839. [PMID: 30949786 DOI: 10.1007/s10103-019-02778-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/18/2019] [Indexed: 12/25/2022]
Abstract
Satellite cells, a population of skeletal muscular stem cells, are generally recognized as the main and, possibly, the sole source of postnatal muscle regeneration. Previous studies have revealed the potential of low-level laser (LLL) irradiation in promoting satellite cell proliferation, which, thereby, boosts the recovery of skeletal muscle from atrophy. The purpose of this study is to investigate the beneficial effect of LLL on disuse-induced atrophy. The optimal irradiation condition of LLL (808 nm) enhancing the proliferation of Pax7+ve cells, isolated from tibialis anterior (TA) muscle, was examined and applied on TA muscle of disuse-induced atrophy model of the rats accordingly. Healthy rats were used as the control. On one hand, transiently, LLL was able to postpone the progression of atrophy for 1 week through a reduction of apoptosis in Pax7-veMyoD+ve (myocyte) population. Simultaneously, a significant enhancement was observed in Pax7+veMyoD+ve population; however, most of the increased cells underwent apoptosis since the second week, which suggested an impaired maturation of the population. On the other hand, in normal control rats with LLL irradiation, a significant increase in Pax7+veMyoD+ve cells and a significant decrease of apoptosis were observed. As a result, a strengthened muscle contraction was observed. Our data showed the capability of LLL in postponing the progression of disuse-induced atrophy for the first time. Furthermore, the result of normal rats with LLL irradiation showed the effectiveness of LLL to strengthen muscle contraction in healthy control.
Collapse
|
38
|
Goshu HA, Chu M, Wu X, Pengjia B, Ding XZ, Yan P. Association study and expression analysis of GPC1 gene copy number variation in Chinese Datong yak ( Bos grunniens) breed. ITALIAN JOURNAL OF ANIMAL SCIENCE 2019. [DOI: 10.1080/1828051x.2019.1586456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Habtamu Abera Goshu
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
- Animal Science Department, Oromia Agricultural Research Institute, Bako Agricultural Research Center, Bako, Ethiopia
| | - Min Chu
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
| | - Xiaoyun Wu
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
| | - Bao Pengjia
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
| | - Xue Zhi Ding
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
| | - Ping Yan
- Animal Science Department, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, Gansu, China
| |
Collapse
|
39
|
Cho DS, Doles JD. Skeletal Muscle Progenitor Cell Heterogeneity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1169:179-193. [PMID: 31487024 DOI: 10.1007/978-3-030-24108-7_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissue-specific stem cells contribute to adult tissue maintenance, repair, and regeneration. In skeletal muscle, many different mononuclear cell types are capable of giving rise to differentiated muscle. Of these tissue stem-like cells, satellite cells (SCs) are the most studied muscle stem cell population and are widely considered the main cellular source driving muscle repair and regeneration in adult tissue. Within the satellite cell pool, many distinct subpopulations exist, each exhibiting differential abilities to exit quiescence, expand, differentiate, and self-renew. In this chapter, we discuss the different stem cell types that can give rise to skeletal muscle tissue and then focus on satellite cell heterogeneity during the process of myogenesis/muscle regeneration. Finally, we highlight emerging opportunities to better characterize muscle stem cell heterogeneity, which will ultimately deepen our appreciation of stem cells in muscle development, repair/regeneration, aging, and disease.
Collapse
Affiliation(s)
- Dong Seong Cho
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
40
|
Prüller J, Mannhardt I, Eschenhagen T, Zammit PS, Figeac N. Satellite cells delivered in their niche efficiently generate functional myotubes in three-dimensional cell culture. PLoS One 2018; 13:e0202574. [PMID: 30222770 PMCID: PMC6141091 DOI: 10.1371/journal.pone.0202574] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 08/05/2018] [Indexed: 12/22/2022] Open
Abstract
Biophysical/biochemical cues from the environment contribute to regulation of the regenerative capacity of resident skeletal muscle stem cells called satellites cells. This can be observed in vitro, where muscle cell behaviour is influenced by the particular culture substrates and whether culture is performed in a 2D or 3D environment, with changes including morphology, nuclear shape and cytoskeletal organization. To create a 3D skeletal muscle model we compared collagen I, Fibrin or PEG-Fibrinogen with different sources of murine and human myogenic cells. To generate tension in the 3D scaffold, biomaterials were polymerised between two flexible silicone posts to mimic tendons. This 3D culture system has multiple advantages including being simple, fast to set up and inexpensive, so providing an accessible tool to investigate myogenesis in a 3D environment. Immortalised human and murine myoblast lines, and primary murine satellite cells showed varying degrees of myogenic differentiation when cultured in these biomaterials, with C2 myoblasts in particular forming large multinucleated myotubes in collagen I or Fibrin. However, murine satellite cells retained in their niche on a muscle fibre and embedded in 3D collagen I or Fibrin gels generated aligned, multinucleated and contractile myotubes.
Collapse
Affiliation(s)
- Johanna Prüller
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, England
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.,DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
| | - Peter S Zammit
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, England
| | - Nicolas Figeac
- King's College London, Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, England
| |
Collapse
|
41
|
Mattyasovszky SG, Langendorf EK, Ritz U, Schmitz C, Schmidtmann I, Nowak TE, Wagner D, Hofmann A, Rommens PM, Drees P. Exposure to radial extracorporeal shock waves modulates viability and gene expression of human skeletal muscle cells: a controlled in vitro study. J Orthop Surg Res 2018; 13:75. [PMID: 29625618 PMCID: PMC5889540 DOI: 10.1186/s13018-018-0779-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 03/23/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recent clinical and animal studies have shown that extracorporeal shock wave therapy has a promoting influence on the healing process of musculoskeletal disorders. However, the underlying biological effects of extracorporeal shock wave therapy on human skeletal muscle cells have not yet been investigated. METHODS In this study, we investigated human skeletal muscle cells after exposure to radial extracorporeal shock waves in a standardized in vitro setup. Cells were isolated from muscle specimens taken from adult patients undergoing spine surgery. Primary muscle cells were exposed once or twice to radial extracorporeal shock waves in vitro with different energy flux densities. Cell viability and gene expression of the paired box protein 7 (Pax7), neural cell adhesion molecule (NCAM), and myogenic factor 5 (Myf5) and MyoD as muscle cell markers were compared to non-treated muscle cells that served as controls. RESULTS Isolated muscle cells were positive for the hallmark protein of satellite cells, Pax7, as well as for the muscle cell markers NCAM, MyoD, and Myf5. Exposure to radial extracorporeal shock waves at low energy flux densities enhanced cell viability, whereas higher energy flux densities had no further significant impact. Gene expression analyses of muscle specific genes (Pax7, NCAM, Myf5, and MyoD) demonstrated a significant increase after single exposure to the highest EFD (4 bar, 0.19 mJ/mm2) and after double exposure with the medium EFDs (2 and 3 bar; 0.09 and 0.14 mJ/mm2, respectively). Double exposure of the highest EFD, however, results in a significant down-regulation when compared to single exposure with this EFD. CONCLUSIONS This is the first study demonstrating that radial extracorporal shock wave therapy has the potential to modulate the biological function of human skeletal muscle cells. Based on our experimental findings, we hypothesize that radial extracorporal shock wave therapy could be a promising therapeutic modality to improve the healing process of sports-related structural muscle injuries.
Collapse
Affiliation(s)
- Stefan G Mattyasovszky
- Department of Orthopedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany.
| | - Eva K Langendorf
- Department of Orthopedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Christoph Schmitz
- Extracorporeal Shock Wave Research Unit, Department of Anatomy II, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Irene Schmidtmann
- Institue for Medical Biometry, Epidemiology and Computer Science, University Medical Centre of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Tobias E Nowak
- Department of Orthopedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Daniel Wagner
- Department of Orthopedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Alexander Hofmann
- Department of Orthopedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Pol M Rommens
- Department of Orthopedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Philipp Drees
- Department of Orthopedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|
42
|
Yamamoto M, Legendre NP, Biswas AA, Lawton A, Yamamoto S, Tajbakhsh S, Kardon G, Goldhamer DJ. Loss of MyoD and Myf5 in Skeletal Muscle Stem Cells Results in Altered Myogenic Programming and Failed Regeneration. Stem Cell Reports 2018; 10:956-969. [PMID: 29478898 PMCID: PMC5918368 DOI: 10.1016/j.stemcr.2018.01.027] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 11/22/2022] Open
Abstract
MyoD and Myf5 are fundamental regulators of skeletal muscle lineage determination in the embryo, and their expression is induced in satellite cells following muscle injury. MyoD and Myf5 are also expressed by satellite cell precursors developmentally, although the relative contribution of historical and injury-induced expression to satellite cell function is unknown. We show that satellite cells lacking both MyoD and Myf5 (double knockout [dKO]) are maintained with aging in uninjured muscle. However, injured muscle fails to regenerate and dKO satellite cell progeny accumulate in damaged muscle but do not undergo muscle differentiation. dKO satellite cell progeny continue to express markers of myoblast identity, although their myogenic programming is labile, as demonstrated by dramatic morphological changes and increased propensity for non-myogenic differentiation. These data demonstrate an absolute requirement for either MyoD or Myf5 in muscle regeneration and indicate that their expression after injury stabilizes myogenic identity and confers the capacity for muscle differentiation. MyoD or Myf5 expression in satellite cells is essential for muscle regeneration Satellite cells lacking both regulatory genes exhibit labile myogenic programming A single functional allele of either MyoD or Myf5 can support muscle regeneration Satellite cells lacking both MyoD and Myf5 are maintained with aging
Collapse
Affiliation(s)
- Masakazu Yamamoto
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Nicholas P Legendre
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Arpita A Biswas
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Alexander Lawton
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Shoko Yamamoto
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA
| | - Shahragim Tajbakhsh
- Institut Pasteur, Stem Cells & Development, CNRS URA 2578, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, 15 North 2030 East, Salt Lake City, UT 84112, USA
| | - David J Goldhamer
- Department of Molecular & Cell Biology, University of Connecticut Stem Cell Institute, University of Connecticut, 91 N. Eagleville Road, Storrs, CT 06269, USA.
| |
Collapse
|
43
|
Recapitulation of Extracellular LAMININ Environment Maintains Stemness of Satellite Cells In Vitro. Stem Cell Reports 2018; 10:568-582. [PMID: 29337118 PMCID: PMC5830886 DOI: 10.1016/j.stemcr.2017.12.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 12/15/2017] [Accepted: 12/15/2017] [Indexed: 12/15/2022] Open
Abstract
Satellite cells function as precursor cells in mature skeletal muscle homeostasis and regeneration. In healthy tissue, these cells are maintained in a state of quiescence by a microenvironment formed by myofibers and basement membrane in which LAMININs (LMs) form a major component. In the present study, we evaluated the satellite cell microenvironment in vivo and found that these cells are encapsulated by LMα2–5. We sought to recapitulate this satellite cell niche in vitro by culturing satellite cells in the presence of recombinant LM-E8 fragments. We show that treatment with LM-E8 promotes proliferation of satellite cells in an undifferentiated state, through reduced phosphorylation of JNK and p38. On transplantation into injured muscle tissue, satellite cells cultured with LM-E8 promoted the regeneration of skeletal muscle. These findings represent an efficient method of culturing satellite cells for use in transplantation through the recapitulation of the satellite cell niche using recombinant LM-E8 fragments. Satellite cells are encapsulated by LMα2–5 LM-E8 promotes proliferation of satellite cells in an undifferentiated state Satellite cells cultured with LM-E8 enhanced the regeneration of skeletal muscle
Collapse
|
44
|
Ambrosio R, De Stefano MA, Di Girolamo D, Salvatore D. Thyroid hormone signaling and deiodinase actions in muscle stem/progenitor cells. Mol Cell Endocrinol 2017; 459:79-83. [PMID: 28630021 DOI: 10.1016/j.mce.2017.06.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/01/2017] [Accepted: 06/15/2017] [Indexed: 11/30/2022]
Abstract
Thyroid hormone (TH) regulates such crucial biological functions as normal growth, development and metabolism of nearly all vertebrate tissues. In skeletal muscle, TH plays a critical role in regulating the function of satellite cells, the bona fide skeletal muscle stem cells. Deiodinases (D2 and D3) have been found to modulate the expression of various TH target genes in satellite cells. Regulation of the expression and activity of the deiodinases constitutes a cell-autonomous, pre-receptor mechanism that controls crucial steps during the various phases of myogenesis. Here, we review the roles of deiodinases in skeletal muscle stem cells, particularly in muscle homeostasis and upon regeneration. We focus on the role of T3 in stem cell functions and in commitment towards lineage progression. We also discuss how deiodinases might be therapeutically exploited to improve satellite-cell-mediated muscle repair in skeletal muscle disorders or injury.
Collapse
Affiliation(s)
- Raffaele Ambrosio
- Istituto di Ricovero e Cura a Carattere Scientifico SDN, Naples, Italy
| | - Maria Angela De Stefano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Daniela Di Girolamo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Domenico Salvatore
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
45
|
Abstract
Our understanding of satellite cells, now known to be the obligate stem cells of skeletal muscle, has increased dramatically in recent years due to the introduction of new molecular, genetic, and technical resources. In addition to their role in acute repair of damaged muscle, satellite cells are of interest in the fields of aging, exercise, neuromuscular disease, and stem cell therapy, and all of these applications have driven a dramatic increase in our understanding of the activity and potential of satellite cells. However, many fundamental questions of satellite cell biology remain to be answered, including their emergence as a specific lineage, the degree and significance of heterogeneity within the satellite cell population, the roles of their interactions with other resident and infiltrating cell types during homeostasis and regeneration, and the relative roles of intrinsic vs extrinsic factors that may contribute to satellite cell dysfunction in the context of aging or disease. This review will address the current state of these open questions in satellite cell biology.
Collapse
Affiliation(s)
- Ddw Cornelison
- University of Missouri, Columbia, MO, United States; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
46
|
Zammit PS. Function of the myogenic regulatory factors Myf5, MyoD, Myogenin and MRF4 in skeletal muscle, satellite cells and regenerative myogenesis. Semin Cell Dev Biol 2017; 72:19-32. [PMID: 29127046 DOI: 10.1016/j.semcdb.2017.11.011] [Citation(s) in RCA: 508] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/03/2017] [Accepted: 11/06/2017] [Indexed: 12/19/2022]
Abstract
Discovery of the myogenic regulatory factor family of transcription factors MYF5, MYOD, Myogenin and MRF4 was a seminal step in understanding specification of the skeletal muscle lineage and control of myogenic differentiation during development. These factors are also involved in specification of the muscle satellite cell lineage, which becomes the resident stem cell compartment inadult skeletal muscle. While MYF5, MYOD, Myogenin and MRF4 have subtle roles in mature muscle, they again play a crucial role in directing satellite cell function to regenerate skeletal muscle: linking the genetic control of developmental and regenerative myogenesis. Here, I review the role of the myogenic regulatory factors in developing and mature skeletal muscle, satellite cell specification and muscle regeneration.
Collapse
Affiliation(s)
- Peter S Zammit
- King's College London, Randall Centre for Cell and Molecular Biophysics, London, SE1 1UL, UK.
| |
Collapse
|
47
|
Staufen1 inhibits MyoD translation to actively maintain muscle stem cell quiescence. Proc Natl Acad Sci U S A 2017; 114:E8996-E9005. [PMID: 29073096 DOI: 10.1073/pnas.1708725114] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Tissue regeneration depends on the timely activation of adult stem cells. In skeletal muscle, the adult stem cells maintain a quiescent state and proliferate upon injury. We show that muscle stem cells (MuSCs) use direct translational repression to maintain the quiescent state. High-resolution single-molecule and single-cell analyses demonstrate that quiescent MuSCs express high levels of Myogenic Differentiation 1 (MyoD) transcript in vivo, whereas MyoD protein is absent. RNA pulldowns and costainings show that MyoD mRNA interacts with Staufen1, a potent regulator of mRNA localization, translation, and stability. Staufen1 prevents MyoD translation through its interaction with the MyoD 3'-UTR. MuSCs from Staufen1 heterozygous (Staufen1+/-) mice have increased MyoD protein expression, exit quiescence, and begin proliferating. Conversely, blocking MyoD translation maintains the quiescent phenotype. Collectively, our data show that MuSCs express MyoD mRNA and actively repress its translation to remain quiescent yet primed for activation.
Collapse
|
48
|
Milanesi A, Lee JW, Yang A, Liu YY, Sedrakyan S, Cheng SY, Perin L, Brent GA. Thyroid Hormone Receptor Alpha is Essential to Maintain the Satellite Cell Niche During Skeletal Muscle Injury and Sarcopenia of Aging. Thyroid 2017; 27:1316-1322. [PMID: 28847239 PMCID: PMC5649408 DOI: 10.1089/thy.2017.0021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Myopathic changes are commonly described in hypothyroid and hyperthyroid patients, including muscular atrophy and weakness. Satellite cells (SCs) play a major role in skeletal muscle maintenance and regeneration after injury. A mouse model of resistance to thyroid hormone-TRα1PV demonstrated impaired skeletal muscle regeneration after injury with significant reduction of SCs, suggesting that exhaustion of the SC pool contributes to the impaired regeneration. To test this hypothesis, SC activation and proliferation were analyzed in vivo in response to skeletal muscle injury and during aging. METHODS SCs of TRα1PV male mice were analyzed four days after cardiotoxin-induced muscle injury, and they were compared to wild-type (WT) male animals. TRα-knockdown C2C12 myoblasts were injected into injured skeletal muscle, and four days after transplantation, the in vivo behavior was compared to control C2C12 myoblasts. Skeletal muscle regeneration was compared in younger and older TRα1PV and WT animals. RESULTS The total number of SCs in skeletal muscle of TRα1PV mice was significantly lower than control, both before and shortly after muscle injury, with significant impairment of SC activation, consistent with SC pool exhaustion. TRα-knockdown myoblasts showed impaired in vivo proliferation and migration. TRα1PV mice had skeletal muscle loss and significant impairment in skeletal muscle regeneration with aging. This translated to a significant reduction of the SC pool with aging compared to WT mice. CONCLUSION TRα plays an important role in the maintenance of the SC pool. Impaired skeletal muscle regeneration in TRα1PV mice is associated with insufficient SC activation and proliferation, as well as the progressive loss of the SC pool with aging. Regulation of the SC pool and SC proliferation provides a therapeutic target to enhance skeletal muscle regeneration and possibly slow age-associated sarcopenia.
Collapse
Affiliation(s)
- Anna Milanesi
- Division of Endocrinology, Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jang-Won Lee
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California
| | - An Yang
- Division of Endocrinology, Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Yan-Yun Liu
- Division of Endocrinology, Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Sargis Sedrakyan
- Department of Urology, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Sheue-yann Cheng
- Laboratory of Molecular Biology, National Cancer Institute, Bethesda, Maryland
| | - Laura Perin
- Department of Urology, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California
| | - Gregory A. Brent
- Division of Endocrinology, Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
49
|
Ishii K, Suzuki N, Mabuchi Y, Sekiya I, Akazawa C. Technical advantage of recombinant collagenase for isolation of muscle stem cells. Regen Ther 2017; 7:1-7. [PMID: 30271846 PMCID: PMC6134918 DOI: 10.1016/j.reth.2017.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/19/2017] [Accepted: 06/02/2017] [Indexed: 02/07/2023] Open
Abstract
Background Muscle satellite cells are resident skeletal muscle stem cells responsible for muscle regeneration. Isolation of satellite cells is a critical process for clinical application such as drug screening and cell transplantation. Fluorescence-activated cell sorting (FACS) enables the direct isolation of satellite cells from muscle tissue. During the process used to isolate satellite cells from skeletal muscle, enzymatic digestion is the first step. Therefore, the evaluation and standardization of enzymes is important not only for reproducibility of cellular yield and viability, but also for traceability of material used in protocols. Methods The comparison of muscle digestion was performed either by a mixture of recombinant collagenase G (ColG) and collagenase H (ColH) or by a conventional collagenase II. The degree of cell damage and surface antigen expression upon collagenase treatment were analyzed by FACS. To investigate whether satellite cells isolated using recombinant collagenase can regenerate injured muscle, satellite cells were cultured, transplanted into injured muscles, and analyzed by immunostaining. Results We show that ColG and ColH were efficient to isolate satellite cells from mouse skeletal muscle tissue. Digestion with a combination of ColG and ColH enriched satellite cells with intact surface antigens such as α7 and β1 integrins. Furthermore, satellite cells isolated using ColG and ColH dramatically proliferated and remained undifferentiated in vitro. When transplanted, satellite cells isolated using ColG and ColH enhanced the therapeutic efficacy in vivo. Conclusions Our results provide an efficient method of satellite cell preparation using recombinant collagenases with a high cell yield, viability of cells, and regeneration potency to fit the biological raw material criteria.
Collapse
Key Words
- CTX, cardiotoxin
- ColG, collagenase G
- ColH, collagenase H
- Collagenase
- Ct, cycle threshold
- ECM, extracellular matrix
- FACS, fluorescence-activated cell sorting
- Muscle stem cell
- PBS, phosphate-buffered saline
- PE, phycoerythrin
- PI, propidium iodide
- Regeneration
- Satellite cell
- TA, tibialis anterior
- Transplantation
Collapse
Affiliation(s)
- Kana Ishii
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Nobuharu Suzuki
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yo Mabuchi
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Chihiro Akazawa
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
50
|
Reed KM, Mendoza KM, Abrahante JE, Barnes NE, Velleman SG, Strasburg GM. Response of turkey muscle satellite cells to thermal challenge. I. transcriptome effects in proliferating cells. BMC Genomics 2017; 18:352. [PMID: 28477619 PMCID: PMC5420122 DOI: 10.1186/s12864-017-3740-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/27/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Climate change poses a multi-dimensional threat to food and agricultural systems as a result of increased risk to animal growth, development, health, and food product quality. This study was designed to characterize transcriptional changes induced in turkey muscle satellite cells cultured under cold or hot thermal challenge to better define molecular mechanisms by which thermal stress alters breast muscle ultrastructure. RESULTS Satellite cells isolated from the pectoralis major muscle of 7-weeks-old male turkeys from two breeding lines (16 weeks body weight-selected and it's randombred control) were proliferated in culture at 33 °C, 38 °C or 43 °C for 72 h. Total RNA was isolated and 12 libraries subjected to RNAseq analysis. Statistically significant differences in gene expression were observed among treatments and between turkey lines with a greater number of genes altered by cold treatment than by hot and fewer differences observed between lines than between temperatures. Pathway analysis found that cold treatment resulted in an overrepresentation of genes involved in cell signaling/signal transduction and cell communication/cell signaling as compared to control (38 °C). Heat-treated muscle satellite cells showed greater tendency towards expression of genes related to muscle system development and differentiation. CONCLUSIONS This study demonstrates significant transcriptome effects on turkey skeletal muscle satellite cells exposed to thermal challenge. Additional effects on gene expression could be attributed to genetic selection for 16 weeks body weight (muscle mass). New targets are identified for further research on the differential control of satellite cell proliferation in poultry.
Collapse
Affiliation(s)
- Kent M. Reed
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN USA
| | - Kristelle M. Mendoza
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN USA
| | - Juan E. Abrahante
- University of Minnesota Informatics Institute, University of Minnesota, Minneapolis, MN USA
| | - Natalie E. Barnes
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN USA
| | - Sandra G. Velleman
- Department of Animal Sciences, The Ohio State University, Columbus, OH USA
- Ohio Agricultural Research and Development Center, Wooster, OH USA
| | - Gale M. Strasburg
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI USA
| |
Collapse
|