1
|
Suzuki T, Nishi Y, Koyama T, Nakada M, Arimatsu R, Komiya Y, Ogawa A, Osaki R, Maeno T, Egusa AS, Nakamura M, Tatsumi R, Ojima K, Nishimura T. Reduced myogenic differentiation capacity of satellite cell-derived myoblasts in male ICR mice compared with male C57BL/6 and BALB/c mice. In Vitro Cell Dev Biol Anim 2025:10.1007/s11626-025-01035-0. [PMID: 40387981 DOI: 10.1007/s11626-025-01035-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/27/2025] [Indexed: 05/20/2025]
Abstract
Many strains of wild-type laboratory mice have been developed for studies in the life sciences, including skeletal muscle cell biology. Muscle regeneration capacity differs among wild-type mouse strains. However, few studies have focused on whether myogenic stem cells (satellite cells) are directly related to mouse strain-dependent myoregeneration gaps using in vitro culture models. In this study, we selected three major wild-type mouse strains, CD1 (outbred; Jcl:ICR [ICR]), C57BL/6NJcl (inbred; B6), and BALB/cAJcl (inbred; C), which are widely used in laboratory experiments. Initially, we compared myotube fusion capabilities using satellite cell-derived myoblasts. The results showed that cell cultures isolated from male ICR mice could not efficiently form myotubes owing to low expression levels of myogenic regulatory factors (e.g., MyoD, myogenin, myocyte enhancer factor [MEF] 2A, and MEF2C) compared with B6 and C mouse strains. Next, we compared the myofiber-type compositions of muscle tissues and cultured myotubes among male mice from each of the three strains. Although each muscle tissue used for satellite cell isolation similarly expressed fast-twitch myofiber markers in all mouse strains, male ICR-derived myoblasts formed abundant amounts of slow-type myotubes. By contrast, myotubes from male B6 and C mice expressed substantial levels of fast-twitch myofiber markers. We also performed a comparative experiment in female ICR, B6, and C mouse strains, similar to the male mouse experiments. The myogenic differentiation potencies of myoblasts and myofiber-type compositions of myotubes in female mouse strains were similar. Thus, male ICR-derived satellite cells (myoblasts) had low myogenic differentiation potential, which may be associated with the tendency slow-twitch myotube formation.
Collapse
Affiliation(s)
- Takahiro Suzuki
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan.
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan.
| | - Yuriko Nishi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Taku Koyama
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Minori Nakada
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Rio Arimatsu
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Yusuke Komiya
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Aoi Ogawa
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Rika Osaki
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takahiro Maeno
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Ai Saiga Egusa
- Department of Food Science and Technology, Nippon Veterinary and Life Science University, Musashino, Japan
| | - Mako Nakamura
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Ryuichi Tatsumi
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Koichi Ojima
- Muscle Biology Research Unit, Division of Animal Products Research, Institute of Livestock and Grassland Science, Naro, Tsukuba, Japan
| | - Takanori Nishimura
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|
2
|
Shi DL, Grifone R, Zhang X, Li H. Rbm24-mediated post-transcriptional regulation of skeletal and cardiac muscle development, function and regeneration. J Muscle Res Cell Motil 2025; 46:53-65. [PMID: 39614020 DOI: 10.1007/s10974-024-09685-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
RNA-binding proteins are critically involved in the post-transcriptional control of gene expression during embryonic development and in adult life, contributing to regulating cell differentiation and maintaining tissue homeostasis. Compared to the relatively well documented functions of transcription factors, the regulatory roles of RNA-binding proteins in muscle development and function remain largely elusive. However, deficiency of many RNA-binding proteins has been associated with muscular defects, neuromuscular disorders and heart diseases, such as myotonic dystrophy, amyotrophic lateral sclerosis, and cardiomyopathy. Rbm24 is highly conserved among vertebrates and is one of the best characterized RNA-binding proteins with crucial implication in the myogenic and cardiomyogenic programs. It presents the distinctive particularity of displaying highly restricted expression in both skeletal and cardiac muscles, with changes in subcellular localization during the process of differentiation. Functional analyses using different vertebrate models have clearly demonstrated its requirement for skeletal muscle differentiation and regeneration as well as for myocardium organization and cardiac function, by regulating the expression of both common and distinct target genes in these tissues. The challenge remains to decipher the dynamic feature of post-transcriptional circuits regulated by Rbm24 during skeletal myogenesis, cardiomyogenesis, and muscle repair. This review discusses current understanding of its function in striated muscles and its possible implication in human disease, with the aim of identifying research gaps for future investigation.
Collapse
Affiliation(s)
- De-Li Shi
- Laboratoire de Biologie du Développement, Sorbonne Université, CNRS UMR7622, INSERM U1156, LBD, Paris, F-75005, France.
| | - Raphaëlle Grifone
- Laboratoire de Biologie du Développement, Sorbonne Université, CNRS UMR7622, INSERM U1156, LBD, Paris, F-75005, France
| | - Xiangmin Zhang
- College of Marine Life Sciences, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Hongyan Li
- College of Marine Life Sciences, Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| |
Collapse
|
3
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
4
|
Matusova Z, Dykstra W, de Pablo Y, Zetterdahl OG, Canals I, van Gelder CAGH, Vos HR, Pérez-Sala D, Kubista M, Abaffy P, Ahlenius H, Valihrach L, Hol EM, Pekny M. Aberrant neurodevelopment in human iPS cell-derived models of Alexander disease. Glia 2025; 73:57-79. [PMID: 39308436 DOI: 10.1002/glia.24618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 12/21/2024]
Abstract
Alexander disease (AxD) is a rare and severe neurodegenerative disorder caused by mutations in glial fibrillary acidic protein (GFAP). While the exact disease mechanism remains unknown, previous studies suggest that mutant GFAP influences many cellular processes, including cytoskeleton stability, mechanosensing, metabolism, and proteasome function. While most studies have primarily focused on GFAP-expressing astrocytes, GFAP is also expressed by radial glia and neural progenitor cells, prompting questions about the impact of GFAP mutations on central nervous system (CNS) development. In this study, we observed impaired differentiation of astrocytes and neurons in co-cultures of astrocytes and neurons, as well as in neural organoids, both generated from AxD patient-derived induced pluripotent stem (iPS) cells with a GFAPR239C mutation. Leveraging single-cell RNA sequencing (scRNA-seq), we identified distinct cell populations and transcriptomic differences between the mutant GFAP cultures and a corrected isogenic control. These findings were supported by results obtained with immunocytochemistry and proteomics. In co-cultures, the GFAPR239C mutation resulted in an increased abundance of immature cells, while in unguided neural organoids and cortical organoids, we observed altered lineage commitment and reduced abundance of astrocytes. Gene expression analysis revealed increased stress susceptibility, cytoskeletal abnormalities, and altered extracellular matrix and cell-cell communication patterns in the AxD cultures, which also exhibited higher cell death after stress. Overall, our results point to altered cell differentiation in AxD patient-derived iPS-cell models, opening new avenues for AxD research.
Collapse
Affiliation(s)
- Zuzana Matusova
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Werner Dykstra
- Department of Translational Neuroscience, University Medical Centre Utrecht Brain Centre, Utrecht University, Utrecht, The Netherlands
| | - Yolanda de Pablo
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Oskar G Zetterdahl
- Stem Cells, Aging and Neurodegeneration Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
- Glial and Neuronal Biology Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Isaac Canals
- Glial and Neuronal Biology Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
- Division of Metabolism, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
- ITINERARE-Innovative therapies in rare diseases, University Research Priority Program, University of Zurich, Zurich, Switzerland
| | - Charlotte A G H van Gelder
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Harmjan R Vos
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
- Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Henrik Ahlenius
- Stem Cells, Aging and Neurodegeneration Lab, Department of Experimental Medical Science, Faculty of Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Centre Utrecht Brain Centre, Utrecht University, Utrecht, The Netherlands
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
5
|
Chinvattanachot G, Rivas D, Duque G. Mechanisms of muscle cells alterations and regeneration decline during aging. Ageing Res Rev 2024; 102:102589. [PMID: 39566742 DOI: 10.1016/j.arr.2024.102589] [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: 07/13/2024] [Revised: 10/27/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024]
Abstract
Skeletal muscles are essential for locomotion and body metabolism regulation. As muscles age, they lose strength, elasticity, and metabolic capability, leading to ineffective motion and metabolic derangement. Both cellular and extracellular alterations significantly influence muscle aging. Satellite cells (SCs), the primary muscle stem cells responsible for muscle regeneration, become exhausted, resulting in diminished population and functionality during aging. This decline in SC function impairs intercellular interactions as well as extracellular matrix production, further hindering muscle regeneration. Other muscle-resident cells, such as fibro-adipogenic progenitors (FAPs), pericytes, and immune cells, also deteriorate with age, reducing local growth factor activities and responsiveness to stress or injury. Systemic signaling, including hormonal changes, contributes to muscle cellular catabolism and disrupts muscle homeostasis. Collectively, these cellular and environmental components interact, disrupting muscle homeostasis and regeneration in advancing age. Understanding these complex interactions offers insights into potential regenerative strategies to mitigate age-related muscle degeneration.
Collapse
Affiliation(s)
- Guntarat Chinvattanachot
- Department of Orthopedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| | - Daniel Rivas
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Gustavo Duque
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, QC, Canada; Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
6
|
Hurley-Novatny A, Chang D, Murakami K, Wang L, Li H. Poor bone health in Duchenne muscular dystrophy: a multifactorial problem beyond corticosteroids and loss of ambulation. Front Endocrinol (Lausanne) 2024; 15:1398050. [PMID: 39669499 PMCID: PMC11634624 DOI: 10.3389/fendo.2024.1398050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/31/2024] [Indexed: 12/14/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive, fatal muscle wasting disease caused by X-linked mutations in the dystrophin gene. Alongside the characteristic muscle weakness, patients face a myriad of skeletal complications, including osteoporosis/osteopenia, high susceptibility to vertebral and long bone fractures, fat embolism post-fracture, scoliosis, and growth retardation. Those skeletal abnormalities significantly compromise quality of life and are sometimes life-threatening. These issues were traditionally attributed to loss of ambulation and chronic corticosteroid use, but recent investigations have unveiled a more intricate etiology. Factors such as vitamin D deficiency, hormonal imbalances, systemic inflammation, myokine release from dystrophic muscle, and vascular dysfunction are emerging as significant contributors as well. This expanded understanding illuminates the multifaceted pathogenesis underlying skeletal issues in DMD. Present therapeutic options are limited and lack specificity. Advancements in understanding the pathophysiology of bone complications in DMD will offer promising avenues for novel treatment modalities. In this review, we summarize the current understanding of factors contributing to bone problems in DMD and delineate contemporary and prospective multidisciplinary therapeutic approaches.
Collapse
Affiliation(s)
- Amelia Hurley-Novatny
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - David Chang
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Katsuhiro Murakami
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - Ling Wang
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
| | - Hongshuai Li
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA, United States
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
7
|
Xue S, Benvie AM, Blum JE, Kolba NJ, Cosgrove BD, Thalacker-Mercer A, Berry DC. Suppressing PDGFRβ Signaling Enhances Myocyte Fusion to Promote Skeletal Muscle Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618247. [PMID: 39464006 PMCID: PMC11507758 DOI: 10.1101/2024.10.15.618247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Muscle cell fusion is critical for forming and maintaining multinucleated myotubes during skeletal muscle development and regeneration. However, the molecular mechanisms directing cell-cell fusion are not fully understood. Here, we identify platelet-derived growth factor receptor beta (PDGFRβ) signaling as a key modulator of myocyte fusion in adult muscle cells. Our findings demonstrate that genetic deletion of Pdgfrβ enhances muscle regeneration and increases myofiber size, whereas PDGFRβ activation impairs muscle repair. Inhibition of PDGFRβ activity promotes myonuclear accretion in both mouse and human myotubes, whereas PDGFRβ activation stalls myotube development by preventing cell spreading to limit fusion potential. Transcriptomics analysis show that PDGFRβ signaling cooperates with TGFβ signaling to direct myocyte size and fusion. Mechanistically, PDGFRβ signaling requires STAT1 activation, and blocking STAT1 phosphorylation enhances myofiber repair and size during regeneration. Collectively, PDGFRβ signaling acts as a regenerative checkpoint and represents a potential clinical target to rapidly boost skeletal muscle repair.
Collapse
Affiliation(s)
- Siwen Xue
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | - Abigail M Benvie
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | - Jamie E Blum
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Current address: Department of Chemical Engineering; Stanford University; Stanford, CA
| | - Nikolai J Kolba
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
| | | | - Anna Thalacker-Mercer
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Department of Cell, Development and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel C Berry
- The Divisional of Nutritional Sciences at Cornell University, Ithaca, NY
- Corresponding author
| |
Collapse
|
8
|
Ugorets V, Mendez PL, Zagrebin D, Russo G, Kerkhoff Y, Kotsaris G, Jatzlau J, Stricker S, Knaus P. Dynamic remodeling of septin structures fine-tunes myogenic differentiation. iScience 2024; 27:110630. [PMID: 39246450 PMCID: PMC11380178 DOI: 10.1016/j.isci.2024.110630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 06/02/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024] Open
Abstract
Controlled myogenic differentiation is integral to the development, maintenance and repair of skeletal muscle, necessitating precise regulation of myogenic progenitors and resident stem cells. The transformation of proliferative muscle progenitors into multinuclear syncytia involves intricate cellular processes driven by cytoskeletal reorganization. While actin and microtubles have been extensively studied, we illuminate the role of septins, an essential yet still often overlooked cytoskeletal component, in myoblast architecture. Notably, Septin9 emerges as a critical regulator of myoblast differentiation during the initial commitment phase. Knock-down of Septin9 in C2C12 cells and primary mouse myoblasts accelerates the transition from proliferation to committed progenitor transcriptional programs. Furthermore, we unveil significant reorganization and downregulation of Septin9 during myogenic differentiation. Collectively, we propose that filmamentous septin structures and their orchestrated reorganization in myoblasts are part of a temporal regulatory mechanism governing the differentiation of myogenic progenitors. This study sheds light on the dynamic interplay between cytoskeletal components underlying controlled myogenic differentiation.
Collapse
Affiliation(s)
- Vladimir Ugorets
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
| | - Paul-Lennard Mendez
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
- Max Planck Institute for Molecular Genetics, IMPRS-Biology and Computation, 14195 Berlin, Germany
| | - Dmitrii Zagrebin
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
| | - Giulia Russo
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), 13125 Berlin, Germany
| | - Yannic Kerkhoff
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Bionanointerfaces Group, 14195 Berlin, Germany
| | - Georgios Kotsaris
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, 14195 Berlin, Germany
| | - Jerome Jatzlau
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
| | - Sigmar Stricker
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, 14195 Berlin, Germany
| | - Petra Knaus
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Signal Transduction Group, 14195 Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
9
|
Verma M, Asakura Y, Wang X, Zhou K, Ünverdi M, Kann AP, Krauss RS, Asakura A. Endothelial cell signature in muscle stem cells validated by VEGFA-FLT1-AKT1 axis promoting survival of muscle stem cell. eLife 2024; 13:e73592. [PMID: 38842166 PMCID: PMC11216748 DOI: 10.7554/elife.73592] [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: 09/02/2021] [Accepted: 06/05/2024] [Indexed: 06/07/2024] Open
Abstract
Endothelial and skeletal muscle lineages arise from common embryonic progenitors. Despite their shared developmental origin, adult endothelial cells (ECs) and muscle stem cells (MuSCs; satellite cells) have been thought to possess distinct gene signatures and signaling pathways. Here, we shift this paradigm by uncovering how adult MuSC behavior is affected by the expression of a subset of EC transcripts. We used several computational analyses including single-cell RNA-seq (scRNA-seq) to show that MuSCs express low levels of canonical EC markers in mice. We demonstrate that MuSC survival is regulated by one such prototypic endothelial signaling pathway (VEGFA-FLT1). Using pharmacological and genetic gain- and loss-of-function studies, we identify the FLT1-AKT1 axis as the key effector underlying VEGFA-mediated regulation of MuSC survival. All together, our data support that the VEGFA-FLT1-AKT1 pathway promotes MuSC survival during muscle regeneration, and highlights how the minor expression of select transcripts is sufficient for affecting cell behavior.
Collapse
Affiliation(s)
- Mayank Verma
- Department of Pediatrics & Neurology, Division of Pediatric Neurology, The University of Texas Southwestern Medical CenterDallasUnited States
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Yoko Asakura
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Xuerui Wang
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Kasey Zhou
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Mahmut Ünverdi
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| | - Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Graduate School of Biomedical Sciencesf, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
- Graduate School of Biomedical Sciencesf, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Atsushi Asakura
- Stem Cell Institute, University of Minnesota Medical SchoolMinneapolisUnited States
- Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical SchoolMinneapolisUnited States
- Department of Neurology, University of Minnesota Medical SchoolMinneapolisUnited States
| |
Collapse
|
10
|
Tam LM, Rand MD. Review: myogenic and muscle toxicity targets of environmental methylmercury exposure. Arch Toxicol 2024; 98:1645-1658. [PMID: 38546836 PMCID: PMC11105986 DOI: 10.1007/s00204-024-03724-3] [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: 12/12/2023] [Accepted: 02/29/2024] [Indexed: 05/01/2024]
Abstract
A number of environmental toxicants are noted for their activity that leads to declined motor function. However, the role of muscle as a proximal toxicity target organ for environmental agents has received considerably less attention than the toxicity targets in the nervous system. Nonetheless, the effects of conventional neurotoxicants on processes of myogenesis and muscle maintenance are beginning to resolve a concerted role of muscle as a susceptible toxicity target. A large body of evidence from epidemiological, animal, and in vitro studies has established that methylmercury (MeHg) is a potent developmental toxicant, with the nervous system being a preferred target. Despite its well-recognized status as a neurotoxicant, there is accumulating evidence that MeHg also targets muscle and neuromuscular development as well as contributes to the etiology of motor defects with prenatal MeHg exposure. Here, we summarize evidence for targets of MeHg in the morphogenesis and maintenance of skeletal muscle that reveal effects on MeHg distribution, myogenesis, myotube formation, myotendinous junction formation, neuromuscular junction formation, and satellite cell-mediated muscle repair. We briefly recapitulate the molecular and cellular mechanisms of skeletal muscle development and highlight the pragmatic role of alternative model organisms, Drosophila and zebrafish, in delineating the molecular underpinnings of muscle development and MeHg-mediated myotoxicity. Finally, we discuss how toxicity targets in muscle development may inform the developmental origins of health and disease theory to explain the etiology of environmentally induced adult motor deficits and accelerated decline in muscle fitness with aging.
Collapse
Affiliation(s)
- Lok Ming Tam
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY, 14642, USA.
- Clinical and Translational Science Institute, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA.
| | - Matthew D Rand
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Rochester, NY, 14642, USA
| |
Collapse
|
11
|
Sales Conniff A, Tur J, Kohena K, Zhang M, Gibbons J, Heller LC. DNA Electrotransfer Regulates Molecular Functions in Skeletal Muscle. Bioelectricity 2024; 6:80-90. [PMID: 39119567 PMCID: PMC11304878 DOI: 10.1089/bioe.2022.0041] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
Background Tissues, such as skeletal muscle, have been targeted for the delivery of plasmid DNA (pDNA) encoding vaccines and therapeutics. The application of electric pulses (electroporation or electrotransfer) increases cell membrane permeability to enhance plasmid delivery and expression. However, the molecular effects of DNA electrotransfer on the muscle tissue are poorly characterized. Materials and Methods Four hours after intramuscular plasmid electrotransfer, we evaluated gene expression changes by RNA sequencing. Differentially expressed genes were analyzed by gene ontology (GO) pathway enrichment analysis. Results GO analysis highlighted many enriched molecular functions. The terms regulated by pulse application were related to muscle stress, the cytoskeleton and inflammation. The terms regulated by pDNA injection were related to a DNA-directed response and its control. Several terms regulated by pDNA electrotransfer were similar to those regulated by pulse application. However, the terms related to pDNA injection differed, focusing on entry of the plasmid into the cells and intracellular trafficking. Conclusion Each muscle stimulus resulted in specific regulated molecular functions. Identifying the unique intrinsic molecular changes driven by intramuscular DNA electrotransfer will aid in the design of preventative and therapeutic gene therapies.
Collapse
Affiliation(s)
- Amanda Sales Conniff
- Department of Medical Engineering, University of South Florida, Tampa, Florida, USA
| | - Jared Tur
- Department of Medical Engineering, University of South Florida, Tampa, Florida, USA
| | - Kristopher Kohena
- Department of Medical Engineering, University of South Florida, Tampa, Florida, USA
| | - Min Zhang
- USF Genomics Core, University of South Florida, Tampa, Florida, USA
| | - Justin Gibbons
- USF Omics Hub, University of South Florida, Tampa, Florida, USA
| | - Loree C. Heller
- Department of Medical Engineering, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
12
|
Li C, Cao H, Ren Y, Jia J, Yang G, Jin J, Shi X. Eicosapentaenoic acid-mediated activation of PGAM2 regulates skeletal muscle growth and development via the PI3K/AKT pathway. Int J Biol Macromol 2024; 268:131547. [PMID: 38641281 DOI: 10.1016/j.ijbiomac.2024.131547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/20/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024]
Abstract
Eicosapentaenoic acid regulates glucose uptake in skeletal muscle and significantly affects whole-body energy metabolism. However, the underlying molecular mechanism remains unclear. Here we report that eicosapentaenoic acid activates phosphoglycerate mutase 2, which mediates the conversion of 2-phosphoglycerate into 3-phosphoglycerate. This enzyme plays a pivotal role in glycerol degradation, thereby facilitating the proliferation and differentiation of satellite cells in skeletal muscle. Interestingly, phosphoglycerate mutase 2 inhibits mitochondrial metabolism, promoting the formation of fast-type muscle fibers. Treatment with eicosapentaenoic acid and phosphoglycerate mutase 2 knockdown induced opposite transcriptomic changes, most of which were enriched in the PI3K-AKT signaling pathway. Phosphoglycerate mutase 2 activated the PI3K-AKT signaling pathway, which inhibited the phosphorylation of FOXO1, and, in turn, inhibited mitochondrial function and promoted the formation of fast-type muscle fibers. Our results suggest that eicosapentaenoic acid promotes skeletal muscle growth and regulates glucose metabolism by targeting phosphoglycerate mutase 2 and activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Chenchen Li
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China
| | - Haigang Cao
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China
| | - Yingchun Ren
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China
| | - Jinrui Jia
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China
| | - Gongshe Yang
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China
| | - Jianjun Jin
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China.
| | - Xin'e Shi
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, PR China.
| |
Collapse
|
13
|
Wang X, Zong X, Ye M, Jin C, Xu T, Yang J, Gao C, Wang X, Yan H. Lysine Distinctively Manipulates Myogenic Regulatory Factors and Wnt/Ca 2+ Pathway in Slow and Fast Muscles, and Their Satellite Cells of Postnatal Piglets. Cells 2024; 13:650. [PMID: 38607088 PMCID: PMC11011516 DOI: 10.3390/cells13070650] [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: 12/05/2023] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024] Open
Abstract
Muscle regeneration, representing an essential homeostatic process, relies mainly on the myogenic progress of resident satellite cells, and it is modulated by multiple physical and nutritional factors. Here, we investigated how myogenic differentiation-related factors and pathways respond to the first limiting amino acid lysine (Lys) in the fast and slow muscles, and their satellite cells (SCs), of swine. Thirty 28-day-old weaned piglets with similar body weights were subjected to three diet regimens: control group (d 0-28: 1.31% Lys, n = 12), Lys-deficient group (d 0-28: 0.83% Lys, n = 12), and Lys rescue group (d 0-14: 0.83% Lys; d 15-28: 1.31% Lys, n = 6). Pigs on d 15 and 29 were selectively slaughtered for muscular parameters evaluation. Satellite cells isolated from fast (semimembranosus) and slow (semitendinosus) muscles were also selected to investigate differentiation ability variations. We found Lys deficiency significantly hindered muscle development in both fast and slow muscles via the distinct manipulation of myogenic regulatory factors and the Wnt/Ca2+ pathway. In the SC model, Lys deficiency suppressed the Wnt/Ca2+ pathways and myosin heavy chain, myogenin, and myogenic regulatory factor 4 in slow muscle SCs but stimulated them in fast muscle SCs. When sufficient Lys was attained, the fast muscle-derived SCs Wnt/Ca2+ pathway (protein kinase C, calcineurin, calcium/calmodulin-dependent protein kinase II, and nuclear factor of activated T cells 1) was repressed, while the Wnt/Ca2+ pathway of its counterpart was stimulated to further the myogenic differentiation. Lys potentially manipulates the differentiation of porcine slow and fast muscle myofibers via the Wnt/Ca2+ pathway in opposite trends.
Collapse
Affiliation(s)
- Xiaofan Wang
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Xiaoyin Zong
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Mao Ye
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Chenglong Jin
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Tao Xu
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Jinzeng Yang
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI 96822, USA;
| | - Chunqi Gao
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Xiuqi Wang
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Huichao Yan
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| |
Collapse
|
14
|
Nguyen J, Gilbert PM. Decoding the forces that shape muscle stem cell function. Curr Top Dev Biol 2024; 158:279-306. [PMID: 38670710 DOI: 10.1016/bs.ctdb.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Skeletal muscle is a force-producing organ composed of muscle tissues, connective tissues, blood vessels, and nerves, all working in synergy to enable movement and provide support to the body. While robust biomechanical descriptions of skeletal muscle force production at the body or tissue level exist, little is known about force application on microstructures within the muscles, such as cells. Among various cell types, skeletal muscle stem cells reside in the muscle tissue environment and play a crucial role in driving the self-repair process when muscle damage occurs. Early evidence indicates that the fate and function of skeletal muscle stem cells are controlled by both biophysical and biochemical factors in their microenvironments, but much remains to accomplish in quantitatively describing the biophysical muscle stem cell microenvironment. This book chapter aims to review current knowledge on the influence of biophysical stresses and landscape properties on muscle stem cells in heath, aging, and diseases.
Collapse
Affiliation(s)
- Jo Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
15
|
Yu IS, Choi YR, Choi J, Kim MK, Jung CH, Um MY, Kim MJ. Discovery of Novel Stimulators of Pax7 and/or MyoD: Enhancing the Efficacy of Cultured Meat Production through Culture Media Enrichment. BIOSENSORS 2023; 14:24. [PMID: 38248401 PMCID: PMC10813534 DOI: 10.3390/bios14010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/26/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024]
Abstract
The principles of myogenesis play crucial roles in the production of cultured meat, and identifying protein stimulators associated with myogenesis holds great potential to enhance the efficiency of this process. In this study, we used surface plasmon resonance (SPR)-based screening of a natural product library to discover ligands for Pax7 and MyoD, key regulators of satellite cells (SCs), and performed cell-based assays on Hanwoo SCs (HWSCs) to identify substances that promote cell proliferation and/or differentiation. Through an SPR analysis, we found that six chemicals, including one Pax7+/MyoD- chemical, four Pax7+/MyoD+ chemicals, and one Pax7-/MyoD+ chemical, bound to Pax7 and/or MyoD proteins. Among four Pax7+/MyoD+ chemicals, parthenolide (0.5 and 1 µM) and rutin (100 and 200 µM) stimulated cell proliferation in the medium with 10% FBS similar to the medium with 20% FBS, without affecting differentiation. Adenosine, a Pax7-/MyoD+ chemical, accelerated differentiation. These chemicals could be potential additives to reduce the reliance of FBS required for HWSC proliferation and differentiation in cultured meat production.
Collapse
Affiliation(s)
- In-Sun Yu
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (I.-S.Y.); (Y.R.C.); (C.H.J.); (M.Y.U.)
- Department of Food Science and Human Nutrition and K-Food Research Center, Jeonbuk National University, Jeonju-si 54896, Republic of Korea;
| | - Yae Rim Choi
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (I.-S.Y.); (Y.R.C.); (C.H.J.); (M.Y.U.)
- Department of Food Science and Engineering, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jungseok Choi
- Department of Animal Science, Chungbuk National University, Cheongju-si 28644, Republic of Korea;
| | - Mina K. Kim
- Department of Food Science and Human Nutrition and K-Food Research Center, Jeonbuk National University, Jeonju-si 54896, Republic of Korea;
| | - Chang Hwa Jung
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (I.-S.Y.); (Y.R.C.); (C.H.J.); (M.Y.U.)
| | - Min Young Um
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (I.-S.Y.); (Y.R.C.); (C.H.J.); (M.Y.U.)
| | - Min Jung Kim
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea; (I.-S.Y.); (Y.R.C.); (C.H.J.); (M.Y.U.)
| |
Collapse
|
16
|
Taravatfard AZ, Ceballos-Gonzalez C, Siddique AB, Bolivar-Monsalve J, Madadelahi M, Trujillo-de Santiago G, Moisés Alvarez M, Pramanick AK, Martinez Guerra E, Kulinsky L, Madou MJ, Martinez SO, Ray M. Nitrogen-functionalized graphene quantum dot incorporated GelMA microgels as fluorescent 3D-tissue Constructs. NANOSCALE 2023; 15:16277-16286. [PMID: 37650749 DOI: 10.1039/d3nr02612d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Biopolymer microgels present many opportunities in biomedicine and tissue engineering. To understand their in vivo behavior in therapeutic interventions, long-term monitoring is critical, which is usually achieved by incorporating fluorescent materials within the hydrogel matrix. Current research is limited due to issues concerning the biocompatibility and instability of the conventional fluorescent species, which also tend to adversely affect the bio-functionality of the hydrogels. Here, we introduce a microfluidic-based approach to generate nitrogen-functionalized graphene quantum dot (NGQD) incorporated gelatin methacryloyl (GelMA) hydrogel microspheres, capable of long-term monitoring while preserving or enhancing the other favorable features of 3D cell encapsulation. A multilayer droplet-based microfluidic device was designed and fabricated to make monodisperse NGQD-loaded GelMA hydrogel microspheres encapsulating skeletal muscle cells (C2C12). Control over the sizes of microspheres could be achieved by tuning the flow rates in the microfluidic device. Skeletal muscle cells encapsulated in these microgels exhibited high cell viability from day 1 (82.9 ± 6.50%) to day 10 (92.1 ± 3.90%). The NGQD-loaded GelMA microgels encapsulating the cells demonstrated higher metabolic activity compared to the GelMA microgels. Presence of sarcomeric α-actin was verified by immunofluorescence staining on day 10. A fluorescence signal was observed from the NGQD-loaded microgels during the entire period of the study. The investigation reveals the advantages of integrating NGQDs in microgels for non-invasive imaging and monitoring of cell-laden microspheres and presents new opportunities for future therapeutic applications.
Collapse
Affiliation(s)
- Aida Zahra Taravatfard
- School of Engineering and Sciences, Tecnológico de Monterrey, Monterrey, 64849, Mexico.
- Department of Mechanical and Aerospace Engineering, University of California, Irvine, CA 92697, USA
| | | | - Abu Bakar Siddique
- School of Engineering and Sciences, Tecnológico de Monterrey, Monterrey, 64849, Mexico.
| | | | - Masoud Madadelahi
- School of Engineering and Sciences, Tecnológico de Monterrey, Monterrey, 64849, Mexico.
| | - Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey 64849, Mexico
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, Monterrey 64849, Mexico
| | - Mario Moisés Alvarez
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey 64849, Mexico
- Departamento de Ingeniería Mecatrónica y Eléctrica, Tecnológico de Monterrey, Monterrey 64849, Mexico
| | | | - Eduardo Martinez Guerra
- Centro de Investigaciones en Materiales Avanzados, CIMAV Unidad Monterrey, Alianza Norte 202, Apodaca, Nuevo León, C.P. 66628, Mexico
| | - Lawrence Kulinsky
- Department of Mechanical and Aerospace Engineering, University of California, Irvine, CA 92697, USA
| | - Marc J Madou
- School of Engineering and Sciences, Tecnológico de Monterrey, Monterrey, 64849, Mexico.
- Department of Mechanical and Aerospace Engineering, University of California, Irvine, CA 92697, USA
| | - Sergio O Martinez
- School of Engineering and Sciences, Tecnológico de Monterrey, Monterrey, 64849, Mexico.
| | - Mallar Ray
- School of Engineering and Sciences, Tecnológico de Monterrey, Monterrey, 64849, Mexico.
| |
Collapse
|
17
|
Motohashi N, Minegishi K, Aoki Y. Inherited myogenic abilities in muscle precursor cells defined by the mitochondrial complex I-encoding protein. Cell Death Dis 2023; 14:689. [PMID: 37857600 PMCID: PMC10587152 DOI: 10.1038/s41419-023-06192-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023]
Abstract
Skeletal muscle comprises different muscle fibers, including slow- and fast-type muscles, and satellite cells (SCs), which exist in individual muscle fibers and possess different myogenic properties. Previously, we reported that myoblasts (MBs) from slow-type enriched soleus (SOL) had a high potential to self-renew compared with cells derived from fast-type enriched tibialis anterior (TA). However, whether the functionality of myogenic cells in adult muscles is attributed to the muscle fiber in which they reside and whether the characteristics of myogenic cells derived from slow- and fast-type fibers can be distinguished at the genetic level remain unknown. Global gene expression analysis revealed that the myogenic potential of MBs was independent of the muscle fiber type they reside in but dependent on the region of muscles they are derived from. Thus, in this study, proteomic analysis was conducted to clarify the molecular differences between MBs derived from TA and SOL. NADH dehydrogenase (ubiquinone) iron-sulfur protein 8 (Ndufs8), a subunit of NADH dehydrogenase in mitochondrial complex I, significantly increased in SOL-derived MBs compared with that in TA-derived cells. Moreover, the expression level of Ndufs8 in MBs significantly decreased with age. Gain- and loss-of-function experiments revealed that Ndufs8 expression in MBs promoted differentiation, self-renewal, and apoptosis resistance. In particular, Ndufs8 suppression in MBs increased p53 acetylation, followed by a decline in NAD/NADH ratio. Nicotinamide mononucleotide treatment, which restores the intracellular NAD+ level, could decrease p53 acetylation and increase myogenic cell self-renewal ability in vivo. These results suggested that the functional differences in MBs derived from SOL and TA governed by the mitochondrial complex I-encoding gene reflect the magnitude of the decline in SC number observed with aging, indicating that the replenishment of NAD+ is a possible approach for improving impaired cellular functions caused by aging or diseases.
Collapse
Affiliation(s)
- Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan.
| | - Katsura Minegishi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan.
| |
Collapse
|
18
|
Gong Y, Dale R, Fung HF, Amador GO, Smit ME, Bergmann DC. A cell size threshold triggers commitment to stomatal fate in Arabidopsis. SCIENCE ADVANCES 2023; 9:eadf3497. [PMID: 37729402 PMCID: PMC10881030 DOI: 10.1126/sciadv.adf3497] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 08/15/2023] [Indexed: 09/22/2023]
Abstract
How flexible developmental programs integrate information from internal and external factors to modulate stem cell behavior is a fundamental question in developmental biology. Cells of the Arabidopsis stomatal lineage modify the balance of stem cell proliferation and differentiation to adjust the size and cell type composition of mature leaves. Here, we report that meristemoids, one type of stomatal lineage stem cell, trigger the transition from asymmetric self-renewing divisions to commitment and terminal differentiation by crossing a critical cell size threshold. Through computational simulation, we demonstrate that this cell size-mediated transition allows robust, yet flexible termination of stem cell proliferation, and we observe adjustments in the number of divisions before the differentiation threshold under several genetic manipulations. We experimentally evaluate several mechanisms for cell size sensing, and our data suggest that this stomatal lineage transition is dependent on a nuclear factor that is sensitive to DNA content.
Collapse
Affiliation(s)
- Yan Gong
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Renee Dale
- Donald Danforth Plant Science Center, St. Louis, MO 63132 USA
| | - Hannah F. Fung
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Gabriel O. Amador
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Margot E. Smit
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Dominique C. Bergmann
- Department of Biology, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
19
|
Wang P, Liu X, Yao Z, Chen Y, Luo L, Liang K, Tan JHE, Chua MWJ, Chua YJB, Ma S, Zhang L, Ma W, Liu S, Cao W, Guo L, Guang L, Wang Y, Zhao H, Ai N, Li Y, Li C, Wang RR, Teh BT, Jiang L, Yu K, Shyh-Chang N. Lin28a maintains a subset of adult muscle stem cells in an embryonic-like state. Cell Res 2023; 33:712-726. [PMID: 37188880 PMCID: PMC10474071 DOI: 10.1038/s41422-023-00818-y] [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: 09/21/2022] [Accepted: 04/23/2023] [Indexed: 05/17/2023] Open
Abstract
During homeostasis and after injury, adult muscle stem cells (MuSCs) activate to mediate muscle regeneration. However, much remains unclear regarding the heterogeneous capacity of MuSCs for self-renewal and regeneration. Here, we show that Lin28a is expressed in embryonic limb bud muscle progenitors, and that a rare reserve subset of Lin28a+Pax7- skeletal MuSCs can respond to injury at adult stage by replenishing the Pax7+ MuSC pool to drive muscle regeneration. Compared with adult Pax7+ MuSCs, Lin28a+ MuSCs displayed enhanced myogenic potency in vitro and in vivo upon transplantation. The epigenome of adult Lin28a+ MuSCs showed resemblance to embryonic muscle progenitors. In addition, RNA-sequencing revealed that Lin28a+ MuSCs co-expressed higher levels of certain embryonic limb bud transcription factors, telomerase components and the p53 inhibitor Mdm4, and lower levels of myogenic differentiation markers compared to adult Pax7+ MuSCs, resulting in enhanced self-renewal and stress-response signatures. Functionally, conditional ablation and induction of Lin28a+ MuSCs in adult mice revealed that these cells are necessary and sufficient for efficient muscle regeneration. Together, our findings connect the embryonic factor Lin28a to adult stem cell self-renewal and juvenile regeneration.
Collapse
Affiliation(s)
- Peng Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xupeng Liu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ziyue Yao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu Chen
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lanfang Luo
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kun Liang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jun-Hao Elwin Tan
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Laboratory of Cancer Therapeutics, Program in Cancer and Stem Cell Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
| | - Min-Wen Jason Chua
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Laboratory of Cancer Therapeutics, Program in Cancer and Stem Cell Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
| | - Yan-Jiang Benjamin Chua
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
- Laboratory of Cancer Therapeutics, Program in Cancer and Stem Cell Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
| | - Shilin Ma
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Liping Zhang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wenwu Ma
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuqing Liu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wenhua Cao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Luyao Guo
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lu Guang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuefan Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - He Zhao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Na Ai
- University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yun Li
- University of Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Chunwei Li
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ruiqi Rachel Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Bin Tean Teh
- Laboratory of Cancer Therapeutics, Program in Cancer and Stem Cell Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Science, National Cancer Centre Singapore, Singapore, Singapore
| | - Lan Jiang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Kang Yu
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ng Shyh-Chang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
20
|
Okafor AE, Lin X, Situ C, Wei X, Xiang Y, Wei X, Wu Z, Diao Y. Single-cell chromatin accessibility profiling reveals a self-renewing muscle satellite cell state. J Cell Biol 2023; 222:e202211073. [PMID: 37382627 PMCID: PMC10309185 DOI: 10.1083/jcb.202211073] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/30/2023] [Accepted: 05/17/2023] [Indexed: 06/30/2023] Open
Abstract
A balance between self-renewal and differentiation is critical for the regenerative capacity of tissue-resident stem cells. In skeletal muscle, successful regeneration requires the orchestrated activation, proliferation, and differentiation of muscle satellite cells (MuSCs) that are normally quiescent. A subset of MuSCs undergoes self-renewal to replenish the stem cell pool, but the features that identify and define self-renewing MuSCs remain to be elucidated. Here, through single-cell chromatin accessibility analysis, we reveal the self-renewal versus differentiation trajectories of MuSCs over the course of regeneration in vivo. We identify Betaglycan as a unique marker of self-renewing MuSCs that can be purified and efficiently contributes to regeneration after transplantation. We also show that SMAD4 and downstream genes are genetically required for self-renewal in vivo by restricting differentiation. Our study unveils the identity and mechanisms of self-renewing MuSCs, while providing a key resource for comprehensive analysis of muscle regeneration.
Collapse
Affiliation(s)
- Arinze E. Okafor
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Xin Lin
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
| | - Chenghao Situ
- Division of Life Science, Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Xiaolin Wei
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
| | - Yu Xiang
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
| | - Xiuqing Wei
- Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Zhenguo Wu
- Division of Life Science, Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Yarui Diao
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC, USA
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC, USA
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| |
Collapse
|
21
|
Amato I, Meurant S, Renard P. The Key Role of Mitochondria in Somatic Stem Cell Differentiation: From Mitochondrial Asymmetric Apportioning to Cell Fate. Int J Mol Sci 2023; 24:12181. [PMID: 37569553 PMCID: PMC10418455 DOI: 10.3390/ijms241512181] [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: 06/30/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
The study of the mechanisms underlying stem cell differentiation is under intensive research and includes the contribution of a metabolic switch from glycolytic to oxidative metabolism. While mitochondrial biogenesis has been previously demonstrated in number of differentiation models, it is only recently that the role of mitochondrial dynamics has started to be explored. The discovery of asymmetric distribution of mitochondria in stem cell progeny has strengthened the interest in the field. This review attempts to summarize the regulation of mitochondrial asymmetric apportioning by the mitochondrial fusion, fission, and mitophagy processes as well as emphasize how asymmetric mitochondrial apportioning in stem cells affects their metabolism, and thus epigenetics, and determines cell fate.
Collapse
Affiliation(s)
- Ilario Amato
- Ressearch Unit in Cell Biology (URBC), Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), 5000 Namur, Belgium; (I.A.); (S.M.)
| | - Sébastien Meurant
- Ressearch Unit in Cell Biology (URBC), Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), 5000 Namur, Belgium; (I.A.); (S.M.)
| | - Patricia Renard
- Ressearch Unit in Cell Biology (URBC), Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), 5000 Namur, Belgium; (I.A.); (S.M.)
- Mass Spectrometry Platform (MaSUN), Namur Research Institute for Life Sciences (Narilis), University of Namur (UNamur), 5000 Namur, Belgium
| |
Collapse
|
22
|
Sharma T, Olea-Flores M, Imbalzano AN. Regulation of the Wnt signaling pathway during myogenesis by the mammalian SWI/SNF ATPase BRG1. Front Cell Dev Biol 2023; 11:1160227. [PMID: 37484913 PMCID: PMC10360407 DOI: 10.3389/fcell.2023.1160227] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Skeletal muscle differentiation is a tightly regulated process, and the importance of the mammalian SWI/SNF (mSWI/SNF) chromatin remodeling family for regulation of genes involved in skeletal myogenesis is well-established. Our prior work showed that bromodomains of mSWI/SNF ATPases BRG1 and BRM contribute to myogenesis by facilitating the binding of mSWI/SNF enzymes to regulatory regions of myogenic and other target genes. Here, we report that pathway analyses of differentially expressed genes from that study identified an additional role for mSWI/SNF enzymes via the regulation of the Wnt signaling pathway. The Wnt pathway has been previously shown to be important for skeletal muscle development. To investigate the importance of mSWI/SNF enzymes for the regulation of the Wnt pathway, individual and dual knockdowns were performed for BRG1 and BRM followed by RNA-sequencing. The results show that BRG1, but not BRM, is a regulator of Wnt pathway components and downstream genes. Reactivation of Wnt pathway by stabilization of β-catenin could rescue the defect in myogenic gene expression and differentiation due to BRG1 knockdown or bromodomain inhibition using a specific small molecule inhibitor, PFI-3. These results demonstrate that BRG1 is required upstream of β-catenin function. Chromatin immunoprecipitation of BRG1, BRM and β-catenin at promoters of Wnt pathway component genes showed binding of BRG1 and β-catenin, which provides further mechanistic insight to the transcriptional regulation of these genes.
Collapse
Affiliation(s)
| | | | - Anthony N. Imbalzano
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, United States
| |
Collapse
|
23
|
Skoupá K, Bátik A, Št'astný K, Sládek Z. Structural Changes in the Skeletal Muscle of Pigs after Long-Term Administration of Testosterone, Nandrolone and a Combination of the Two. Animals (Basel) 2023; 13:2141. [PMID: 37443939 DOI: 10.3390/ani13132141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Anabolic steroid hormones (AASs) are used in most countries of the world to accelerate the growth of animals, increase the volume of their muscles and thereby increase meat production. However, there is a strict ban on the use of AASs in the fattening of all animals in all countries of the European Union, and there must therefore be effective methods of detection and control of these substances. Methods based on chromatography and mass spectrometry may no longer be completely effective when faced with new synthetic steroids of unknown chemical structures and low concentrations. Therefore, there is an effort to develop new methods of AAS detection, based primarily on the monitoring of biological changes at the level of gene expression or changes in metabolism or structure at the cellular level. More detailed knowledge of the mechanisms of action of AASs on tissues is essential for these methods, and histological changes are one of them. In this study, we report histological changes in muscle structure after AAS application, specifically in the size of muscle fibers, the amount of endomysium and the number of nuclei and satellite cells in muscle fibers. A pig model was also intentionally used for the study, as no such study has been carried out on this species, and at the same time, pork is one of the most consumed meats across Europe. The results of histology and fluorescent antibody labeling showed that AASs increased the diameter and surface area of muscle fibers and also significantly increased the number of satellite cells on the fiber surface. The evident correlations between the number of satellite cells, all nuclei and the diameters of muscle fibers between some experimental groups provide evidence that the selected histological parameters could be additional detection mechanisms for screening a large number of samples and indicate the possibility of the presence of AASs in pork meat in the future.
Collapse
Affiliation(s)
- Kristýna Skoupá
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgrSciences, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic
| | - Andrej Bátik
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgrSciences, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic
| | - Kamil Št'astný
- Veterinary Research Institute in Brno, Hudcova 296/70, 621 00 Brno, Czech Republic
| | - Zbyšek Sládek
- Department of Animal Morphology, Physiology and Genetics, Faculty of AgrSciences, Mendel University in Brno, Zemedelska 1, 613 00 Brno, Czech Republic
| |
Collapse
|
24
|
Cabezas F, Cabello-Verrugio C, González N, Salas J, Ramírez MJ, de la Vega E, Olguín HC. NEDD4-1 deficiency impairs satellite cell function during skeletal muscle regeneration. Biol Res 2023; 56:21. [PMID: 37147738 PMCID: PMC10161651 DOI: 10.1186/s40659-023-00432-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/17/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Satellite cells are tissue-specific stem cells primarily responsible for the regenerative capacity of skeletal muscle. Satellite cell function and maintenance are regulated by extrinsic and intrinsic mechanisms, including the ubiquitin-proteasome system, which is key for maintaining protein homeostasis. In this context, it has been shown that ubiquitin-ligase NEDD4-1 targets the transcription factor PAX7 for proteasome-dependent degradation, promoting muscle differentiation in vitro. Nonetheless, whether NEDD4-1 is required for satellite cell function in regenerating muscle remains to be determined. RESULTS Using conditional gene ablation, we show that NEDD4-1 loss, specifically in the satellite cell population, impairs muscle regeneration resulting in a significant reduction of whole-muscle size. At the cellular level, NEDD4-1-null muscle progenitors exhibit a significant decrease in the ability to proliferate and differentiate, contributing to the formation of myofibers with reduced diameter. CONCLUSIONS These results indicate that NEDD4-1 expression is critical for proper muscle regeneration in vivo and suggest that it may control satellite cell function at multiple levels.
Collapse
Affiliation(s)
- Felipe Cabezas
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, 7510157, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Natalia González
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jeremy Salas
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel J Ramírez
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo de la Vega
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hugo C Olguín
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
25
|
Kotsaris G, Qazi TH, Bucher CH, Zahid H, Pöhle-Kronawitter S, Ugorets V, Jarassier W, Börno S, Timmermann B, Giesecke-Thiel C, Economides AN, Le Grand F, Vallecillo-García P, Knaus P, Geissler S, Stricker S. Odd skipped-related 1 controls the pro-regenerative response of fibro-adipogenic progenitors. NPJ Regen Med 2023; 8:19. [PMID: 37019910 PMCID: PMC10076435 DOI: 10.1038/s41536-023-00291-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 03/17/2023] [Indexed: 04/07/2023] Open
Abstract
Skeletal muscle regeneration requires the coordinated interplay of diverse tissue-resident- and infiltrating cells. Fibro-adipogenic progenitors (FAPs) are an interstitial cell population that provides a beneficial microenvironment for muscle stem cells (MuSCs) during muscle regeneration. Here we show that the transcription factor Osr1 is essential for FAPs to communicate with MuSCs and infiltrating macrophages, thus coordinating muscle regeneration. Conditional inactivation of Osr1 impaired muscle regeneration with reduced myofiber growth and formation of excessive fibrotic tissue with reduced stiffness. Osr1-deficient FAPs acquired a fibrogenic identity with altered matrix secretion and cytokine expression resulting in impaired MuSC viability, expansion and differentiation. Immune cell profiling suggested a novel role for Osr1-FAPs in macrophage polarization. In vitro analysis suggested that increased TGFβ signaling and altered matrix deposition by Osr1-deficient FAPs actively suppressed regenerative myogenesis. In conclusion, we show that Osr1 is central to FAP function orchestrating key regenerative events such as inflammation, matrix secretion and myogenesis.
Collapse
Affiliation(s)
- Georgios Kotsaris
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Taimoor H Qazi
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Bioengineering, University of Pennsylvania, 19104, Philadelphia, USA
- Weldon School of Biomedical Engineering, Purdue University, 47907, West Lafayette, IN, USA
| | - Christian H Bucher
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117, Berlin, Germany
| | - Hafsa Zahid
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
- International Max Planck Research School for Biology and Computing IMPRS-BAC, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Ihnestrasse 73, 14195, Berlin, Germany
| | - Sophie Pöhle-Kronawitter
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Vladimir Ugorets
- Institute of Chemistry and Biochemistry, Cell Signaling Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - William Jarassier
- Institut NeuroMyoGène, CNRS UMR 5261, Inserm U1315, Université Claude Bernard Lyon 1, 69008, Lyon, France
| | - Stefan Börno
- Max Planck Institute for Molecular Genetics, Ihnestrasse 73, 14195, Berlin, Germany
| | - Bernd Timmermann
- Max Planck Institute for Molecular Genetics, Ihnestrasse 73, 14195, Berlin, Germany
| | | | | | - Fabien Le Grand
- Institut NeuroMyoGène, CNRS UMR 5261, Inserm U1315, Université Claude Bernard Lyon 1, 69008, Lyon, France
| | - Pedro Vallecillo-García
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Petra Knaus
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Institute of Chemistry and Biochemistry, Cell Signaling Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Sven Geissler
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117, Berlin, Germany
- Berlin Center for Advanced Therapies (BECAT), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin, Germany
| | - Sigmar Stricker
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany.
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
| |
Collapse
|
26
|
Pedrosa MB, Barbosa S, Vitorino R, Ferreira R, Moreira-Gonçalves D, Santos LL. Chemotherapy-Induced Molecular Changes in Skeletal Muscle. Biomedicines 2023; 11:biomedicines11030905. [PMID: 36979884 PMCID: PMC10045751 DOI: 10.3390/biomedicines11030905] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Paraneoplastic conditions such as cancer cachexia are often exacerbated by chemotherapy, which affects the patient’s quality of life as well as the response to therapy. The aim of this narrative review was to overview the body-composition-related changes and molecular effects of different chemotherapy agents used in cancer treatment on skeletal-muscle remodeling. A literature search was performed using the Web of Science, Scopus, and Science Direct databases and a total of 77 papers was retrieved. In general, the literature survey showed that the molecular changes induced by chemotherapy in skeletal muscle have been studied mainly in animal models and mostly in non-tumor-bearing rodents, whereas clinical studies have essentially assessed changes in body composition by computerized tomography. Data from preclinical studies showed that chemotherapy modulates several molecular pathways in skeletal muscle, including the ubiquitin–proteasome pathway, autophagy, IGF-1/PI3K/Akt/mTOR, IL-6/JAK/STAT, and NF-κB pathway; however, the newest chemotherapy agents are underexplored. In conclusion, chemotherapy exacerbates skeletal-muscle wasting in cancer patients; however, the incomplete characterization of the chemotherapy-related molecular effects on skeletal muscle makes the development of new preventive anti-wasting strategies difficult. Therefore, further investigation on molecular mechanisms and clinical studies are necessary.
Collapse
Affiliation(s)
- Mafalda Barbosa Pedrosa
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV-REQUIMTE), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Experimental Pathology and Therapeutics Group, Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal
- Correspondence: (M.B.P.); (L.L.S.)
| | - Samuel Barbosa
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV-REQUIMTE), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Experimental Pathology and Therapeutics Group, Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal
| | - Rui Vitorino
- Department of Medical Sciences, Institute of Biomedicine—iBiMED, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rita Ferreira
- Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology (LAQV-REQUIMTE), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Daniel Moreira-Gonçalves
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto, 4200-450 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600 Porto, Portugal
| | - Lúcio Lara Santos
- Experimental Pathology and Therapeutics Group, Research Center (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal
- Correspondence: (M.B.P.); (L.L.S.)
| |
Collapse
|
27
|
Pan Y, Zhou T, Dong X, Wu L, Wang P, Wang S, Zhang A. Urotensin II can Induce Skeletal Muscle Atrophy Associated with Upregulating Ubiquitin-Proteasome System and Inhibiting the Differentiation of Satellite Cells in CRF Mice. Calcif Tissue Int 2023; 112:603-612. [PMID: 36892588 DOI: 10.1007/s00223-023-01073-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/21/2023] [Indexed: 03/10/2023]
Abstract
Skeletal muscle wasting and atrophy is highly prevalent in chronic renal failure (CRF) and increases the risk of mortality. According to our previous study, we speculate that urotensin II (UII) can induce skeletal muscle atrophy by upregulating ubiquitin-proteasome system(UPS) in CRF. C2C12 mouse myoblast cells were differentiated into myotubes, and myotubes were exposed to different concentrations of UII. Myotube diameters, myosin heavy chain(MHC), p-Fxo03A, skeletal muscle-specific E3 ubiquitin ligases such as muscle RING finger 1 (MuRF1) and muscle atrophy F-box (MAFbx/atrogin1) were detected. Three animal models (the sham operation mice as normal control (NC) group, wild-type C57BL/6 mice with 5/6 nephrectomy (WT CRF) group, UII receptor gene knock out (UT KO) mice with 5/6 nephrectomy (UT KO CRF) group) were designed. Cross-sectional area (CSA) of skeletal muscle tissues in three animal models were measured, and western blot detected protein of UII, p-Fxo03A, MAFbx and MuRF1, and immunofluorescence assays explored the satellite cell marker of Myod1 and Pax7, and PCR arrays detected the muscle protein degradation genes, protein synthesis genes and the genes which were involved in muscle components. UII could decrease mouse myotube diameters, and upregulate dephosphorylated Fxo03A protein. MAFbx and MuRF1 were higher in WT CRF group than that in NC group, but after UII receptor gene was knocked out (UT KO CRF), their expressions were downregulated. UII could inhibit the expression of Myod1 but not Pax7 in animal study. We first demonstrate that skeletal muscle atrophy induced by UII associated with upregulating ubiquitin-proteasome system and inhibiting the differentiation of satellite cells in CRF mice.
Collapse
Affiliation(s)
- Yajing Pan
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Ting Zhou
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Xingtong Dong
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Leiyun Wu
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Peiwen Wang
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Shiyuan Wang
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China
| | - Aihua Zhang
- Department of Nephrology, Xuan Wu Hospital, Capital Medical University, No. 45, Chang-Chun Street, Xicheng District, Beijing, People's Republic of China.
| |
Collapse
|
28
|
Abbas H, Olivere LA, Padgett ME, Schmidt CA, Gilmore BF, McCord TJ, Southerland KW, McClung JM, Kontos CD. Muscle progenitor cells are required for skeletal muscle regeneration and prevention of adipogenesis after limb ischemia. Front Cardiovasc Med 2023; 10:1118738. [PMID: 36937923 PMCID: PMC10017542 DOI: 10.3389/fcvm.2023.1118738] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/08/2023] [Indexed: 03/06/2023] Open
Abstract
Skeletal muscle injury in peripheral artery disease (PAD) has been attributed to vascular insufficiency, however evidence has demonstrated that muscle cell responses play a role in determining outcomes in limb ischemia. Here, we demonstrate that genetic ablation of Pax7+ muscle progenitor cells (MPCs) in a model of hindlimb ischemia (HLI) inhibited muscle regeneration following ischemic injury, despite a lack of morphological or physiological changes in resting muscle. Compared to control mice (Pax7WT), the ischemic limb of Pax7-deficient mice (Pax7Δ) was unable to generate significant force 7 or 28 days after HLI. A significant increase in adipose was observed in the ischemic limb 28 days after HLI in Pax7Δ mice, which replaced functional muscle. Adipogenesis in Pax7Δ mice corresponded with a significant increase in PDGFRα+ fibro/adipogenic progenitors (FAPs). Inhibition of FAPs with batimastat decreased muscle adipose but increased fibrosis. In vitro, Pax7Δ MPCs failed to form myotubes but displayed increased adipogenesis. Skeletal muscle from patients with critical limb threatening ischemia displayed increased adipose in more ischemic regions of muscle, which corresponded with fewer satellite cells. Collectively, these data demonstrate that Pax7+ MPCs are required for muscle regeneration after ischemia and suggest that muscle regeneration may be an important therapeutic target in PAD.
Collapse
Affiliation(s)
- Hasan Abbas
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
- Duke-NUS Medical School, Singapore, Singapore
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
| | | | - Michael E. Padgett
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
| | - Cameron A. Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| | - Brian F. Gilmore
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Timothy J. McCord
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Kevin W. Southerland
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Joseph M. McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Heart Institute, East Carolina University, Greenville, NC, United States
| | - Christopher D. Kontos
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
- Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
29
|
CPNE1 regulates myogenesis through the PERK-eIF2α pathway mediated by endoplasmic reticulum stress. Cell Tissue Res 2023; 391:545-560. [PMID: 36525128 PMCID: PMC9974702 DOI: 10.1007/s00441-022-03720-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022]
Abstract
Sarcopenia is characterized by a progressive reduction in muscle mass or muscle physiological function associated with aging, but the relevant molecular mechanisms are not clear. Here, we identify the role of the myogenesis modifier CPNE1 in sarcopenia. CPNE1 is upregulated in aged skeletal muscles and young skeletal muscle satellite cells with palmitate-induced atrophy. The overexpression of CPNE1 hinders proliferation and differentiation and increases muscle atrophy characteristics in young skeletal muscle-derived satellite cells. In addition, CPNE1 overexpression disrupts the balance of mitochondrial fusion and division and causes endoplasmic reticulum stress. We found that the effects of CPNE1 on mitochondrial function are dependent on the PERK/eIF2α/ATF4 pathway. The overexpression of CPNE1 in young muscles alters membrane lipid composition, reduces skeletal muscle fibrosis regeneration, and exercise capacity in mice. These effects were reversed by PERK inhibitor GSK2606414. Moreover, immunoprecipitation indicates that CPNE1 overexpression greatly increased the acetylation of PERK. Therefore, CPNE1 is an important modifier that drives mitochondrial homeostasis to regulate myogenic cell proliferation and differentiation via the PERK-eIF2α pathway, which could be a valuable target for age-related sarcopenia.
Collapse
|
30
|
Detection of multiple biomarkers associated with satellite cell fate in the contused skeletal muscle of rats for wound age estimation. Int J Legal Med 2023; 137:875-886. [PMID: 36797435 DOI: 10.1007/s00414-023-02971-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 02/08/2023] [Indexed: 02/18/2023]
Abstract
From the perspective of forensic wound age estimation, experiments related to skeletal muscle regeneration after injury have rarely been reported. Here, we examined the time-dependent expression patterns of multiple biomarkers associated with satellite cell fate, including the transcription factor paired box 7 (Pax7), myoblast determination protein (MyoD), myogenin, and insulin-like growth factor (IGF-1), using immunohistochemistry, western blotting, and quantitative real-time PCR in contused skeletal muscle. An animal model of skeletal muscle contusion was established in 30 Sprague-Dawley male rats, and another five rats were employed as non-contused controls. Morphometrically, the data obtained from the numbers of Pax7 + , MyoD + , and myogenin + cells were highly correlated with the wound age. Pax7, MyoD, myogenin, and IGF-1 expression patterns were upregulated after injury at both the mRNA and protein levels. Pax7, MyoD, and myogenin protein expression levels confirmed the results of the morphometrical analysis. Additionally, the relative quantity of IGF-1 protein > 0.92 suggested a wound age of 3 to 7 days. The relative quantity of Pax7 mRNA > 2.44 also suggested a wound age of 3 to 7 days. Relative quantities of Myod1, Myog, and Igf1 mRNA expression > 2.78, > 7.80, or > 3.13, respectively, indicated a wound age of approximately 3 days. In conclusion, the expression levels of Pax7, MyoD, myogenin, and IGF-1 were upregulated in a time-dependent manner during skeletal muscle wound healing, suggesting the potential for using them as candidate biomarkers for wound age estimation in skeletal muscle.
Collapse
|
31
|
Motohashi N, Minegishi K, Imamura M, Aoki Y. Techniques for Injury, Cell Transplantation, and Histological Analysis in Skeletal Muscle. Methods Mol Biol 2023; 2640:193-205. [PMID: 36995596 DOI: 10.1007/978-1-0716-3036-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Skeletal muscle can adjust to changes in physiological and pathological environments by regenerating using myogenic progenitor cells or adapting muscle fiber sizes and types, metabolism, and contraction ability. To study these changes, muscle samples should be appropriately prepared. Therefore, reliable techniques to accurately analyze and evaluate skeletal muscle phenotypes are required. However, although technical approaches to genetically investigating skeletal muscle are improving, the fundamental strategies for capturing muscle pathology are the same over the decades. Hematoxylin and eosin (H&E) staining or antibodies are the simplest and standard methodologies for assessing skeletal muscle phenotypes. In this chapter, we describe fundamental techniques and protocols for inducing skeletal muscle regeneration by using chemicals and cell transplantation, in addition to methods of preparing and evaluating skeletal muscle samples.
Collapse
Affiliation(s)
- Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan.
| | - Katsura Minegishi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Michihiro Imamura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| |
Collapse
|
32
|
Li P, Zhao Y, Liu Y, Zhao Y, Yan Y, Li S, Li S, Tong H. Cyanocobalamin promotes muscle development through the TGF-β signaling pathway. Food Funct 2022; 13:12721-12732. [PMID: 36408829 DOI: 10.1039/d2fo00315e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cyanocobalamin (CNCbl, the compound name of Vitamin B12) is the only mineral vitamin that is essential for growth and development and cannot be produced by animals. Some studies have found that CNCbl can promote the proliferation and migration of C2C12 cells, but the mechanism by which it affects muscle development is still unknown. In this study, we elucidated the effect of CNCbl on muscle development and studied its underlying mechanism. CNCbl could promote the differentiation of C2C12 cells and upregulate Acvr1, p-Smad2 and p-Smad3 in the TGF-β signaling pathway in vitro. CD320 (the receptor in cell surface for binding with CNCbl transporter transcobalamin II) inhibition could reduce the uptake of CNCbl and significantly downregulate the expression of differentiation marker proteins MyoG and MYH2. Furthermore, the levels of p-Smad2 and p-Smad3 were also reduced with the inhibition of CD320, even though CNCbl was added to the C2C12 culture medium. In addition, the injection of CNCbl could accelerate the process of mouse muscle injury repair, enlarge the diameter of newly formed myofibers and upregulate the expression of MYH2, PAX7, CD320, Acvr1, p-Smad2 and p-Smad3 in vivo. These results suggest that CNCbl can promote muscle development and may play its role by regulating the expression of Acvr1, p-Smad2 and p-Smad3 related to the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Ping Li
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China.
| | - Yahao Zhao
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China.
| | - Yongze Liu
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China.
| | - Yuelei Zhao
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China.
| | - Yunqin Yan
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China.
| | - Shuang Li
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China.
| | - Shufeng Li
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China.
| | - Huili Tong
- Laboratory of Cell and Developmental Biology, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
33
|
Saleh KK, Xi H, Switzler C, Skuratovsky E, Romero MA, Chien P, Gibbs D, Gane L, Hicks MR, Spencer MJ, Pyle AD. Single cell sequencing maps skeletal muscle cellular diversity as disease severity increases in dystrophic mouse models. iScience 2022; 25:105415. [PMID: 36388984 PMCID: PMC9646951 DOI: 10.1016/j.isci.2022.105415] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/01/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by out-of-frame mutations in the DMD gene resulting in the absence of a functional dystrophin protein, leading to a devastating and progressive lethal muscle-wasting disease. Little is known about cellular heterogeneity as disease severity increases. Advances in single-cell RNA sequencing (scRNA-seq) enabled us to explore skeletal muscle-resident cell populations in healthy, dystrophic, and severely dystrophic mouse models. We found increased frequencies of activated fibroblasts, fibro-adipogenic progenitor cells, and pro-inflammatory macrophages in dystrophic gastrocnemius muscles and an upregulation of extracellular matrix genes on endothelial cells in dystrophic and severely dystrophic muscles. We observed a pronounced risk of clotting, especially in the severely dystrophic mice with increased expression of plasminogen activator inhibitor-1 in endothelial cells, indicating endothelial cell impairment as disease severity increases. This work extends our understanding of the severe nature of DMD which should be considered when developing single or combinatorial approaches for DMD.
Collapse
Affiliation(s)
- Kholoud K. Saleh
- Department of Molecular, Cellular and Integrative Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Haibin Xi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Corey Switzler
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Emily Skuratovsky
- CIRM Bridges Program, California State University, Northridge, CA 91330, USA
| | - Matthew A. Romero
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Peggie Chien
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Devin Gibbs
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Lily Gane
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Michael R. Hicks
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Melissa J. Spencer
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, University of California Los Angeles, CA 90095, USA
| | - April D. Pyle
- Department of Molecular, Cellular and Integrative Physiology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
34
|
Koike H, Manabe I, Oishi Y. Mechanisms of cooperative cell-cell interactions in skeletal muscle regeneration. Inflamm Regen 2022; 42:48. [DOI: 10.1186/s41232-022-00234-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022] Open
Abstract
AbstractSkeletal muscles have an extraordinary capacity to regenerate themselves when injured. Skeletal muscle stem cells, called satellite cells, play a central role in muscle regeneration via three major steps: activation, proliferation, and differentiation. These steps are affected by multiple types of cells, such as immune cells, fibro-adipogenic progenitor cells, and vascular endothelial cells. The widespread use of single-cell sequencing technologies has enabled the identification of novel cell subpopulations associated with muscle regeneration and their regulatory mechanisms. This review summarizes the dynamism of the cellular community that controls and promotes muscle regeneration, with a particular focus on skeletal muscle stem cells.
Collapse
|
35
|
McCann C, Quinteros M, Adelugba I, Morgada MN, Castelblanco AR, Davis EJ, Lanzirotti A, Hainer SJ, Vila AJ, Navea JG, Padilla-Benavides T. The mitochondrial Cu+ transporter PiC2 (SLC25A3) is a target of MTF1 and contributes to the development of skeletal muscle in vitro. Front Mol Biosci 2022; 9:1037941. [PMID: 36438658 PMCID: PMC9682256 DOI: 10.3389/fmolb.2022.1037941] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
The loading of copper (Cu) into cytochrome c oxidase (COX) in mitochondria is essential for energy production in cells. Extensive studies have been performed to characterize mitochondrial cuproenzymes that contribute to the metallation of COX, such as Sco1, Sco2, and Cox17. However, limited information is available on the upstream mechanism of Cu transport and delivery to mitochondria, especially through Cu-impermeable membranes, in mammalian cells. The mitochondrial phosphate transporter SLC25A3, also known as PiC2, binds Cu+ and transports the ion through these membranes in eukaryotic cells, ultimately aiding in the metallation of COX. We used the well-established differentiation model of primary myoblasts derived from mouse satellite cells, wherein Cu availability is necessary for growth and maturation, and showed that PiC2 is a target of MTF1, and its expression is both induced during myogenesis and favored by Cu supplementation. PiC2 deletion using CRISPR/Cas9 showed that the transporter is required for proliferation and differentiation of primary myoblasts, as both processes are delayed upon PiC2 knock-out. The effects of PiC2 deletion were rescued by the addition of Cu to the growth medium, implying the deleterious effects of PiC2 knockout in myoblasts may be in part due to a failure to deliver sufficient Cu to the mitochondria, which can be compensated by other mitochondrial cuproproteins. Co-localization and co-immunoprecipitation of PiC2 and COX also suggest that PiC2 may participate upstream in the copper delivery chain into COX, as verified by in vitro Cu+-transfer experiments. These data indicate an important role for PiC2 in both the delivery of Cu to the mitochondria and COX, favoring the differentiation of primary myoblasts.
Collapse
|
36
|
Yamanouchi K, Tanaka Y, Ikeda M, Kato S, Okino R, Nishi H, Hakuno F, Takahashi SI, Chambers J, Matsuwaki T, Uchida K. Macroglossia and less advanced dystrophic change in the tongue muscle of the Duchenne muscular dystrophy rat. Skelet Muscle 2022; 12:24. [PMID: 36258243 PMCID: PMC9580129 DOI: 10.1186/s13395-022-00307-7] [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: 02/21/2022] [Accepted: 10/06/2022] [Indexed: 11/25/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an X-linked muscle disease caused by a complete lack of dystrophin, which stabilizes the plasma membrane of myofibers. The orofacial function is affected in an advanced stage of DMD and this often leads to an eating disorder such as dysphagia. Dysphagia is caused by multiple etiologies including decreased mastication and swallowing. Therefore, preventing the functional declines of mastication and swallowing in DMD is important to improve the patient’s quality of life. In the present study, using a rat model of DMD we generated previously, we performed analyses on the masseter and tongue muscles, both are required for proper eating function. Methods Age-related changes of the masseter and tongue muscle of DMD rats were analyzed morphometrically, histologically, and immunohistochemically. Also, transcription of cellular senescent markers, and utrophin (Utrn), a functional analog of dystrophin, was examined. Results The masseter muscle of DMD rats showed progressive dystrophic changes as observed in their hindlimb muscle, accompanied by increased transcription of p16 and p19. On the other hand, the tongue of DMD rats showed macroglossia due to hypertrophy of myofibers with less dystrophic changes. Proliferative activity was preserved in the satellite cells from the tongue muscle but was perturbed severely in those from the masseter muscle. While Utrn transcription was increased in the masseter muscle of DMD rats compared to WT rats, probably due to a compensatory mechanism, its level in the tongue muscle was comparable between WT and DMD rats and was similar to that in the masseter muscle of DMD rats. Conclusions Muscular dystrophy is less advanced in the tongue muscle compared to the masseter muscle in the DMD rat. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-022-00307-7.
Collapse
Affiliation(s)
- Keitaro Yamanouchi
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan.
| | - Yukie Tanaka
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Masanari Ikeda
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Shizuka Kato
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Ryosuke Okino
- Laboratory of Animal Cell Regulation, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Hiroki Nishi
- Laboratory of Animal Cell Regulation, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Fumihiko Hakuno
- Laboratory of Animal Cell Regulation, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Shin-Ichiro Takahashi
- Laboratory of Animal Cell Regulation, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - James Chambers
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Takashi Matsuwaki
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Kazuyuki Uchida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-8657, Japan
| |
Collapse
|
37
|
Chen YF, Lee CW, Wu HH, Lin WT, Lee OK. Immunometabolism of macrophages regulates skeletal muscle regeneration. Front Cell Dev Biol 2022; 10:948819. [PMID: 36147742 PMCID: PMC9485946 DOI: 10.3389/fcell.2022.948819] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Sarcopenia is an age-related progressive loss of skeletal muscle mass, quality, and strength disease. In addition, sarcopenia is tightly correlated with age-associated pathologies, such as sarcopenic obesity and osteoporosis. Further understanding of disease mechanisms and the therapeutic strategies in muscle regeneration requires a deeper knowledge of the interaction of skeletal muscle and other cells in the muscle tissue. Skeletal muscle regeneration is a complex process that requires a series of highly coordinated events involving communication between muscle stem cells and niche cells, such as muscle fibro/adipogenic progenitors and macrophages. Macrophages play a critical role in tissue regeneration and the maintenance of muscle homeostasis by producing growth factors and cytokines that regulate muscle stem cells and myofibroblast activation. Furthermore, the aging-related immune dysregulation associated with the release of trophic factors and the polarization in macrophages transiently affect the inflammatory phase and impair muscle regeneration. In this review, we focus on the role and regulation of macrophages in skeletal muscle regeneration and homeostasis. The aim of this review is to highlight the important roles of macrophages as a therapeutic target in age-related sarcopenia and the increasing understanding of how macrophages are regulated will help to advance skeletal muscle regeneration.
Collapse
Affiliation(s)
- Yu-Fan Chen
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, Taiwan
| | - Chien-Wei Lee
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, Taiwan
| | - Hao-Hsiang Wu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Ting Lin
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Doctoral Degree Program of Translational Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Oscar K. Lee
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan
- *Correspondence: Oscar K. Lee,
| |
Collapse
|
38
|
Ramírez de Acuña F, Hernandez-Torres F, Rodriguez-Outeiriño L, Dominguez JN, Matias-Valiente L, Sanchez-Fernandez C, Franco D, Aranega AE. Pitx2 Differentially Regulates the Distinct Phases of Myogenic Program and Delineates Satellite Cell Lineages During Muscle Development. Front Cell Dev Biol 2022; 10:940622. [PMID: 35874842 PMCID: PMC9298408 DOI: 10.3389/fcell.2022.940622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
The knowledge of the molecular mechanisms that regulate embryonic myogenesis from early myogenic progenitors to myoblasts, as well as the emergence of adult satellite stem cells (SCs) during development, are key concepts to understanding the genesis and regenerative abilities of the skeletal muscle. Several previous pieces of evidence have revealed that the transcription factor Pitx2 might be a player within the molecular pathways controlling somite-derived muscle progenitors’ fate and SC behavior. However, the role exerted by Pitx2 in the progression from myogenic progenitors to myoblasts including SC precursors remains unsolved. Here, we show that Pitx2 inactivation in uncommitted early myogenic precursors diminished cell proliferation and migration leading to muscle hypotrophy and a low number of SCs with decreased myogenic differentiation potential. However, the loss of Pitx2 in committed myogenic precursors gave rise to normal muscles with standard amounts of SCs exhibiting high levels of Pax7 expression. This SC population includes few MYF5+ SC-primed but increased amount of less proliferative miR-106b+cells, and display myogenic differentiation defects failing to undergo proper muscle regeneration. Overall our results demonstrate that Pitx2 is required in uncommitted myogenic progenitors but it is dispensable in committed precursors for proper myogenesis and reveal a role for this transcription factor in the generation of diverse SC subpopulations.
Collapse
Affiliation(s)
- Felícitas Ramírez de Acuña
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaen, Jaén, Spain
- Cardiac and Skeletal Myogenesis Group, MEDINA Foundation, Center for Excellence in Research of Innovative Medicines in Andalusia, Granada, Spain
| | - Francisco Hernandez-Torres
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaen, Jaén, Spain
- Cardiac and Skeletal Myogenesis Group, MEDINA Foundation, Center for Excellence in Research of Innovative Medicines in Andalusia, Granada, Spain
- Department of Biochemistry, Molecular Biology III and Immunology, School of Medicine, University of Granada, Granada, Spain
| | - Lara Rodriguez-Outeiriño
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaen, Jaén, Spain
- Cardiac and Skeletal Myogenesis Group, MEDINA Foundation, Center for Excellence in Research of Innovative Medicines in Andalusia, Granada, Spain
| | - Jorge N. Dominguez
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaen, Jaén, Spain
- Cardiac and Skeletal Myogenesis Group, MEDINA Foundation, Center for Excellence in Research of Innovative Medicines in Andalusia, Granada, Spain
| | - Lidia Matias-Valiente
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaen, Jaén, Spain
- Cardiac and Skeletal Myogenesis Group, MEDINA Foundation, Center for Excellence in Research of Innovative Medicines in Andalusia, Granada, Spain
| | - Cristina Sanchez-Fernandez
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaen, Jaén, Spain
- Cardiac and Skeletal Myogenesis Group, MEDINA Foundation, Center for Excellence in Research of Innovative Medicines in Andalusia, Granada, Spain
| | - Diego Franco
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaen, Jaén, Spain
- Cardiac and Skeletal Myogenesis Group, MEDINA Foundation, Center for Excellence in Research of Innovative Medicines in Andalusia, Granada, Spain
| | - Amelia E. Aranega
- Cardiac and Skeletal Myogenesis Group, Department of Experimental Biology, University of Jaen, Jaén, Spain
- Cardiac and Skeletal Myogenesis Group, MEDINA Foundation, Center for Excellence in Research of Innovative Medicines in Andalusia, Granada, Spain
- *Correspondence: Amelia E. Aranega,
| |
Collapse
|
39
|
Alheib O, da Silva LP, Youn YH, Kwon IK, Reis RL, Correlo VM. 3D bioprinting of gellan gum-based hydrogels tethered with laminin-derived peptides for improved cellular behavior. J Biomed Mater Res A 2022; 110:1655-1668. [PMID: 35678701 DOI: 10.1002/jbm.a.37415] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 11/05/2022]
Abstract
The treatment of skeletal muscle defects is still a topic of noteworthy concern since surgical intervention is not capable of recovering muscle function. Herein, we propose myoblasts laden in laminin-inspired biofunctionalized gellan gum hydrogels as promising tissue-engineered skeletal muscle surrogates. Gellan gum-based hydrogels were developed by combining native gellan gum (GG) and GG tethered with laminin-derived peptides (CIKVAVS (V), KNRLTIELEVRTC (T) or RKRLQVQLSIRTC (Q)), using different polymer content (0.75%-1.875%). Hydrogels were characterized in terms of compressive modulus, molecules trafficking, and C2C12 adhesion. Hydrogels with higher polymeric content (1.125%-1.875%) showed higher stiffness whereas hydrogels with lower polymer content (0.75%-1.125%) showed higher fluorescein isothiocyanate-dextran molecules diffusion. Cell spreading was achieved regardless of the laminin-derived peptide but preferred in hydrogels with higher polymer content (1.125%-1.875%). Taken together, hydrogels with 1.125% of polymer content were selected for printability analysis. GG-based inks showed a non-newtonian, shear-thinning, and thixotropic behavior suitable for printing. Accordingly, all inks were printable, but inks tethered with T and Q peptides presented some signs of clogging. Cell viability was affected after printing but increased after 7 days of culture. After 7 days, cells were spreading but not showing significant signs of cell-cell communications. Therefore, cell density was increased, thus, myocytes loaded in V-tethered GG-based inks showed higher cell-cell communication, spreading morphology, and alignment 7, 14 days post-printing. Overall, myoblasts laden in laminin-inspired biofunctionalized GG-based hydrogels are a promising skeletal muscle surrogate with the potential to be used as in vitro model or explored for further in vivo applications.
Collapse
Affiliation(s)
- Omar Alheib
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Lucilia P da Silva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| | - Yun Hee Youn
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal.,Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal.,Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Vitor M Correlo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| |
Collapse
|
40
|
Shen D, Sugiyama Y, Ishida K, Fuseya S, Ishida T, Kawamata M, Tanaka S. Subfascial infiltration of 0.5% ropivacaine, but not 0.25% ropivacaine, exacerbates damage and inflammation in surgically incised abdominal muscles of rats. Sci Rep 2022; 12:9409. [PMID: 35672375 PMCID: PMC9174254 DOI: 10.1038/s41598-022-13628-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/26/2022] [Indexed: 11/08/2022] Open
Abstract
Ropivacaine-induced myotoxicity in surgically incised muscles has not been fully investigated. We evaluated the effects of infiltration anesthesia with ropivacaine on damage, inflammation and regeneration in the incised muscles of rats undergoing laparotomy. Ropivacaine or saline was infiltrated below the muscle fascia over the incised muscles. Pain-related behaviors and histological muscle damage were assessed. Macrophage infiltration at days 2 and 5 and proliferation of satellite cells at day 5 were detected by CD68 and MyoD immunostaining, respectively. Pain-related behaviors were inhibited by 0.25% and 0.5% of ropivacaine for 2 h after surgery. Single infiltration of 0.5% ropivacaine did not induce injury in intact muscles without incision, but single and repeated infiltration of 0.5% ropivacaine significantly augmented laparotomy-induced muscle injury and increased the numbers of CD68-positve macrophages and MyoD-positive cells compared to those in rats with infiltration of saline or 0.25% ropivacaine. In contrast, there were no significant differences in them between rats with saline infusion and rats with 0.25% ropivacaine infiltration. In conclusion, single or repeated subfascial infiltration of 0.25% ropivacaine can be used without exacerbating the damage and inflammation in surgically incised muscles, but the use of 0.5% ropivacaine may be a concern because of potentially increased muscle damage.
Collapse
Affiliation(s)
- Dandan Shen
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto City, Nagano, 390-8621, Japan
| | - Yuki Sugiyama
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto City, Nagano, 390-8621, Japan
| | - Kumiko Ishida
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto City, Nagano, 390-8621, Japan
| | - Satoshi Fuseya
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto City, Nagano, 390-8621, Japan
| | - Takashi Ishida
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto City, Nagano, 390-8621, Japan
| | - Mikito Kawamata
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto City, Nagano, 390-8621, Japan
| | - Satoshi Tanaka
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto City, Nagano, 390-8621, Japan.
| |
Collapse
|
41
|
Wang J, Broer T, Chavez T, Zhou CJ, Tran S, Xiang Y, Khodabukus A, Diao Y, Bursac N. Myoblast deactivation within engineered human skeletal muscle creates a transcriptionally heterogeneous population of quiescent satellite-like cells. Biomaterials 2022; 284:121508. [PMID: 35421801 PMCID: PMC9289780 DOI: 10.1016/j.biomaterials.2022.121508] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 03/18/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022]
Abstract
Satellite cells (SCs), the adult Pax7-expressing stem cells of skeletal muscle, are essential for muscle repair. However, in vitro investigations of SC function are challenging due to isolation-induced SC activation, loss of native quiescent state, and differentiation to myoblasts. In the present study, we optimized methods to deactivate in vitro expanded human myoblasts within a 3D culture environment of engineered human skeletal muscle tissues ("myobundles"). Immunostaining and gene expression analyses revealed that a fraction of myoblasts within myobundles adopted a quiescent phenotype (3D-SCs) characterized by increased Pax7 expression, cell cycle exit, and activation of Notch signaling. Similar to native SCs, 3D-SC quiescence is regulated by Notch and Wnt signaling while loss of quiescence and reactivation of 3D-SCs can be induced by growth factors including bFGF. Myobundle injury with a bee toxin, melittin, induces robust myofiber fragmentation, functional decline, and 3D-SC proliferation. By applying single cell RNA-sequencing (scRNA-seq), we discover the existence of two 3D-SC subpopulations (quiescent and activated), identify deactivation-associated gene signature using trajectory inference between 2D myoblasts and 3D-SCs, and characterize the transcriptomic changes within reactivated 3D-SCs in response to melittin-induced injury. These results demonstrate the ability of an in vitro engineered 3D human skeletal muscle environment to support the formation of a quiescent and heterogeneous SC population recapitulating several aspects of the native SC phenotype, and provide a platform for future studies of human muscle regeneration and disease-associated SC dysfunction.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Torie Broer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Taylor Chavez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Chris J Zhou
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sabrina Tran
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yu Xiang
- Department of Cell Biology, Duke University, Durham, NC, USA
| | | | - Yarui Diao
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
42
|
Vaes RDW, van Dijk DPJ, Farshadi EA, Olde Damink SWM, Rensen SS, Langen RC. Human pancreatic tumour organoid-derived factors enhance myogenic differentiation. J Cachexia Sarcopenia Muscle 2022; 13:1302-1313. [PMID: 35146962 PMCID: PMC8977981 DOI: 10.1002/jcsm.12917] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/26/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Most patients with pancreatic cancer develop cachexia, which is characterized by progressive muscle loss. The mechanisms underlying muscle loss in cancer cachexia remain elusive. Pancreatic tumour organoids are 3D cell culture models that retain key characteristics of the parent tumour. We aimed to investigate the effect of pancreatic tumour organoid-derived factors on processes that determine skeletal muscle mass, including the regulation of muscle protein turnover and myogenesis. METHODS Conditioned medium (CM) was collected from human pancreatic cancer cell lines (PK-45H, PANC-1, PK-1, and KLM-1), pancreatic tumour organoid cultures from a severely cachectic (PANCO-9a) and a non-cachectic patient (PANCO-12a), and a normal pancreas organoid culture. Differentiating C2C12 myoblasts and mature C2C12 myotubes were exposed to CM for 24 h or maintained in control medium. In myotubes, NF-kB activation was monitored using a NF-κB luciferase reporter construct, and mRNA expression of E3-ubiquitin ligases and REDD1 was analysed by RT-qPCR. C2C12 myoblast proliferation and differentiation were monitored by live cell imaging and myogenic markers and myosin heavy chain (MyHC) isoforms were assessed by RT-qPCR. RESULTS Whereas CM from PK-1 and KLM-1 cells significantly induced NF-κB activation in C2C12 myotubes (PK-1: 3.1-fold, P < 0.001; KLM-1: 2.1-fold, P = 0.01), Atrogin-1/MAFbx and MuRF1 mRNA were only minimally and inconsistently upregulated by the CM of pancreatic cancer cell lines. Similarly, E3-ubiquitin ligases and REDD1 mRNA expression in myotubes were not altered by exposure to pancreatic tumour organoid CM. Compared with the control condition, CM from both PANCO-9a and PANCO-12a tumour organoids increased proliferation of myoblasts, which was accompanied by significant downregulation of the satellite cell marker paired-box 7 (PAX7) (PANCO-9a: -2.1-fold, P < 0.001; PANCO-12a: -2.0-fold, P < 0.001) and myogenic factor 5 (MYF5) (PANCO-9a: -2.1-fold, P < 0.001; PANCO-12a: -1.8-fold, P < 0.001) after 48 h of differentiation. Live cell imaging revealed accelerated alignment and fusion of myoblasts exposed to CM from PANCO-9a and PANCO-12a, which was in line with significantly increased Myomaker mRNA expression levels (PANCO-9a: 2.4-fold, P = 0.001; PANCO-12a: 2.2-fold, P = 0.004). These morphological and transcriptional alterations were accompanied by increased expression of muscle differentiation markers such as MyHC-IIB (PANCO-9a: 2.5-fold, P = 0.04; PANCO-12a: 3.1-fold, P = 0.006). Although the impact of organoid CM on myogenesis was not associated with the cachexia phenotype of the donor patients, it was specific for tumour organoids, as CM of control pancreas organoids did not modulate myogenic fusion. CONCLUSIONS These data show that pancreatic tumour organoid-derived factors alter the kinetics of myogenesis, which may eventually contribute to impaired muscle mass maintenance in cancer cachexia.
Collapse
Affiliation(s)
- Rianne D W Vaes
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - David P J van Dijk
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Elham Aïda Farshadi
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Steven W M Olde Damink
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands.,Department of General, Visceral and Transplantation Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Sander S Rensen
- Department of Surgery and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Ramon C Langen
- Department of Respiratory Medicine and NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
43
|
Floriano JF, Emanueli C, Vega S, Barbosa AMP, Oliveira RGD, Floriano EAF, Graeff CFDO, Abbade JF, Herculano RD, Sobrevia L, Rudge MVC. Pro-angiogenic approach for skeletal muscle regeneration. Biochim Biophys Acta Gen Subj 2022; 1866:130059. [PMID: 34793875 DOI: 10.1016/j.bbagen.2021.130059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022]
Abstract
The angiogenesis process is a phenomenon in which numerous molecules participate in the stimulation of the new vessels' formation from pre-existing vessels. Angiogenesis is a crucial step in tissue regeneration and recovery of organ and tissue function. Muscle diseases affect millions of people worldwide overcome the ability of skeletal muscle to self-repair. Pro-angiogenic therapies are key in skeletal muscle regeneration where both myogenesis and angiogenesis occur. These therapies have been based on mesenchymal stem cells (MSCs), exosomes, microRNAs (miRs) and delivery of biological factors. The use of different calls of biomaterials is another approach, including ceramics, composites, and polymers. Natural polymers are use due its bioactivity and biocompatibility in addition to its use as scaffolds and in drug delivery systems. One of these polymers is the natural rubber latex (NRL) which is biocompatible, bioactive, versatile, low-costing, and capable of promoting tissue regeneration and angiogenesis. In this review, the advances in the field of pro-angiogenic therapies are discussed.
Collapse
Affiliation(s)
- Juliana Ferreira Floriano
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; National Heart and Lung Institute, Imperial College London, London, UK.
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sofia Vega
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | | | | | | | | | - Joelcio Francisco Abbade
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil
| | | | - Luis Sobrevia
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland, Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD, 4029, Queensland, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands.
| | | |
Collapse
|
44
|
Arjona M, Goshayeshi A, Rodriguez-Mateo C, Brett JO, Both P, Ishak H, Rando TA. Tubastatin A maintains adult skeletal muscle stem cells in a quiescent state ex vivo and improves their engraftment ability in vivo. Stem Cell Reports 2022; 17:82-95. [PMID: 35021050 PMCID: PMC8758944 DOI: 10.1016/j.stemcr.2021.11.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 01/11/2023] Open
Abstract
Adult skeletal muscle stem cells (MuSCs) are important for muscle regeneration and constitute a potential source of cell therapy. However, upon isolation, MuSCs rapidly exit quiescence and lose transplantation potency. Maintenance of the quiescent state in vitro preserves MuSC transplantation efficiency and provides an opportunity to study the biology of quiescence. Here we show that Tubastatin A (TubA), an Hdac6 inhibitor, prevents primary cilium resorption, maintains quiescence, and enhances MuSC survival ex vivo. Phenotypic characterization and transcriptomic analysis of TubA-treated cells revealed that TubA maintains most of the biological features and molecular signatures of quiescence. Furthermore, TubA-treated MuSCs showed improved engraftment ability upon transplantation. TubA also induced a return to quiescence and improved engraftment of cycling MuSCs, revealing a potentially expanded application for MuSC therapeutics. Altogether, these studies demonstrate the ability of TubA to maintain MuSC quiescence ex vivo and to enhance the therapeutic potential of MuSCs and their progeny.
Collapse
Affiliation(s)
- Marina Arjona
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Armon Goshayeshi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Cristina Rodriguez-Mateo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Jamie O Brett
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Stem Cell Biology and Regenerative Medicine Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Pieter Both
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Stem Cell Biology and Regenerative Medicine Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Heather Ishak
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Neurology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
45
|
Abstract
The avian pectoralis muscle demonstrates plasticity with regard to size, so that temperate birds facing winter conditions or birds enduring a migration bout tend to have significant increases in the size and mass of this tissue due to muscular hypertrophy. Myonuclear domain (MND), the volume of cytoplasm a myonuclei services, in the pectoralis muscle of birds seems to be altered during thermal stress or changing seasons. However, there is no information available regarding muscle DNA content or ploidy level within the avian pectoralis. Changes in muscle DNA content can be used in this tissue to aid in size and mass changes. Here, we hypothesized that long-distance migrants or temperate residents would use the process of endoreduplication to aid in altering muscle size. Mostly contradictory to our hypotheses, we found no differences in the mean muscle DNA content in any of the 62 species of birds examined in this study. We also found no correlations between mean muscle DNA content and other muscle structural measurements, such as the number of nuclei per millimeter of fiber, myonuclear domain, and fiber cross-sectional area. Thus, while avian muscle seems more phenotypically plastic than mammalian muscle, the biological processes surrounding myonuclear function may be more closely related to those seen in mammals.
Collapse
Affiliation(s)
- Ana Gabriela Jimenez
- Department of Biology, Colgate University, Hamilton, NY 13346, USA.,Department of Biology, Colgate University, Hamilton, NY 13346, USA
| | - Emily Gray Lencyk
- Department of Biology, Colgate University, Hamilton, NY 13346, USA.,Department of Biology, Colgate University, Hamilton, NY 13346, USA
| |
Collapse
|
46
|
Fujii T, Maehara K, Fujita M, Ohkawa Y. Discriminative feature of cells characterizes cell populations of interest by a small subset of genes. PLoS Comput Biol 2021; 17:e1009579. [PMID: 34797848 PMCID: PMC8641884 DOI: 10.1371/journal.pcbi.1009579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 12/03/2021] [Accepted: 10/19/2021] [Indexed: 12/13/2022] Open
Abstract
Organisms are composed of various cell types with specific states. To obtain a comprehensive understanding of the functions of organs and tissues, cell types have been classified and defined by identifying specific marker genes. Statistical tests are critical for identifying marker genes, which often involve evaluating differences in the mean expression levels of genes. Differentially expressed gene (DEG)-based analysis has been the most frequently used method of this kind. However, in association with increases in sample size such as in single-cell analysis, DEG-based analysis has faced difficulties associated with the inflation of P-values. Here, we propose the concept of discriminative feature of cells (DFC), an alternative to using DEG-based approaches. We implemented DFC using logistic regression with an adaptive LASSO penalty to perform binary classification for discriminating a population of interest and variable selection to obtain a small subset of defining genes. We demonstrated that DFC prioritized gene pairs with non-independent expression using artificial data and that DFC enabled characterization of the muscle satellite/progenitor cell population. The results revealed that DFC well captured cell-type-specific markers, specific gene expression patterns, and subcategories of this cell population. DFC may complement DEG-based methods for interpreting large data sets. DEG-based analysis uses lists of genes with differences in expression between groups, while DFC, which can be termed a discriminative approach, has potential applications in the task of cell characterization. Upon recent advances in the high-throughput analysis of single cells, methods of cell characterization such as scRNA-seq can be effectively subjected to the discriminative methods. Statistical methods for detecting differences in individual gene expression are indispensable for understanding cell types. However, conventional statistical methods, such as differentially expressed gene (DEG)-based analysis, have faced difficulties associated with the inflation of P-values because of both the large sample size and selection bias introduced by exploratory data analysis such as single-cell transcriptomics. Here, we propose the concept of discriminative feature of cells (DFC), an alternative to using DEG-based approaches. We implemented DFC using logistic regression with an adaptive LASSO penalty to perform binary classification for the discrimination of a population of interest and variable selection to obtain a small subset of defining genes. We demonstrated that DFC prioritized gene pairs with non-independent expression using artificial data, and that it enabled characterization of the muscle satellite/progenitor cell population. The results revealed that DFC well captured cell-type-specific markers, specific gene expression patterns, and subcategories of this cell population. DFC may complement differentially expressed gene-based methods for interpreting large data sets.
Collapse
Affiliation(s)
- Takeru Fujii
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazumitsu Maehara
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- * E-mail: (KM); (YO)
| | - Masatoshi Fujita
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- * E-mail: (KM); (YO)
| |
Collapse
|
47
|
Rodriguez-Outeiriño L, Hernandez-Torres F, Ramírez-de Acuña F, Matías-Valiente L, Sanchez-Fernandez C, Franco D, Aranega AE. Muscle Satellite Cell Heterogeneity: Does Embryonic Origin Matter? Front Cell Dev Biol 2021; 9:750534. [PMID: 34722534 PMCID: PMC8554119 DOI: 10.3389/fcell.2021.750534] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/27/2021] [Indexed: 12/25/2022] Open
Abstract
Muscle regeneration is an important homeostatic process of adult skeletal muscle that recapitulates many aspects of embryonic myogenesis. Satellite cells (SCs) are the main muscle stem cells responsible for skeletal muscle regeneration. SCs reside between the myofiber basal lamina and the sarcolemma of the muscle fiber in a quiescent state. However, in response to physiological stimuli or muscle trauma, activated SCs transiently re-enter the cell cycle to proliferate and subsequently exit the cell cycle to differentiate or self-renew. Recent evidence has stated that SCs display functional heterogeneity linked to regenerative capability with an undifferentiated subgroup that is more prone to self-renewal, as well as committed progenitor cells ready for myogenic differentiation. Several lineage tracing studies suggest that such SC heterogeneity could be associated with different embryonic origins. Although it has been established that SCs are derived from the central dermomyotome, how a small subpopulation of the SCs progeny maintain their stem cell identity while most progress through the myogenic program to construct myofibers is not well understood. In this review, we synthesize the works supporting the different developmental origins of SCs as the genesis of their functional heterogeneity.
Collapse
Affiliation(s)
- Lara Rodriguez-Outeiriño
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Francisco Hernandez-Torres
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Granada, Spain
| | - F. Ramírez-de Acuña
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Lidia Matías-Valiente
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Cristina Sanchez-Fernandez
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| | - Amelia Eva Aranega
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
- Medina Foundation, Technology Park of Health Sciences, Granada, Spain
| |
Collapse
|
48
|
Shi DL, Grifone R. RNA-Binding Proteins in the Post-transcriptional Control of Skeletal Muscle Development, Regeneration and Disease. Front Cell Dev Biol 2021; 9:738978. [PMID: 34616743 PMCID: PMC8488162 DOI: 10.3389/fcell.2021.738978] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
Embryonic myogenesis is a temporally and spatially regulated process that generates skeletal muscle of the trunk and limbs. During this process, mononucleated myoblasts derived from myogenic progenitor cells within the somites undergo proliferation, migration and differentiation to elongate and fuse into multinucleated functional myofibers. Skeletal muscle is the most abundant tissue of the body and has the remarkable ability to self-repair by re-activating the myogenic program in muscle stem cells, known as satellite cells. Post-transcriptional regulation of gene expression mediated by RNA-binding proteins is critically required for muscle development during embryogenesis and for muscle homeostasis in the adult. Differential subcellular localization and activity of RNA-binding proteins orchestrates target gene expression at multiple levels to regulate different steps of myogenesis. Dysfunctions of these post-transcriptional regulators impair muscle development and homeostasis, but also cause defects in motor neurons or the neuromuscular junction, resulting in muscle degeneration and neuromuscular disease. Many RNA-binding proteins, such as members of the muscle blind-like (MBNL) and CUG-BP and ETR-3-like factors (CELF) families, display both overlapping and distinct targets in muscle cells. Thus they function either cooperatively or antagonistically to coordinate myoblast proliferation and differentiation. Evidence is accumulating that the dynamic interplay of their regulatory activity may control the progression of myogenic program as well as stem cell quiescence and activation. Moreover, the role of RNA-binding proteins that regulate post-transcriptional modification in the myogenic program is far less understood as compared with transcription factors involved in myogenic specification and differentiation. Here we review past achievements and recent advances in understanding the functions of RNA-binding proteins during skeletal muscle development, regeneration and disease, with the aim to identify the fundamental questions that are still open for further investigations.
Collapse
Affiliation(s)
- De-Li Shi
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Developmental Biology Laboratory, CNRS-UMR 7622, Institut de Biologie de Paris-Seine, Sorbonne University, Paris, France
| | - Raphaëlle Grifone
- Developmental Biology Laboratory, CNRS-UMR 7622, Institut de Biologie de Paris-Seine, Sorbonne University, Paris, France
| |
Collapse
|
49
|
Rebboah E, Reese F, Williams K, Balderrama-Gutierrez G, McGill C, Trout D, Rodriguez I, Liang H, Wold BJ, Mortazavi A. Mapping and modeling the genomic basis of differential RNA isoform expression at single-cell resolution with LR-Split-seq. Genome Biol 2021; 22:286. [PMID: 34620214 PMCID: PMC8495978 DOI: 10.1186/s13059-021-02505-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/20/2021] [Indexed: 11/24/2022] Open
Abstract
The rise in throughput and quality of long-read sequencing should allow unambiguous identification of full-length transcript isoforms. However, its application to single-cell RNA-seq has been limited by throughput and expense. Here we develop and characterize long-read Split-seq (LR-Split-seq), which uses combinatorial barcoding to sequence single cells with long reads. Applied to the C2C12 myogenic system, LR-split-seq associates isoforms to cell types with relative economy and design flexibility. We find widespread evidence of changing isoform expression during differentiation including alternative transcription start sites (TSS) and/or alternative internal exon usage. LR-Split-seq provides an affordable method for identifying cluster-specific isoforms in single cells.
Collapse
Affiliation(s)
- Elisabeth Rebboah
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA
| | - Fairlie Reese
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA
| | - Katherine Williams
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA
| | - Gabriela Balderrama-Gutierrez
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA
| | - Cassandra McGill
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA
| | - Diane Trout
- Division of Biology, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Isaryhia Rodriguez
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA
| | - Heidi Liang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA
| | - Barbara J Wold
- Division of Biology, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA.
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
50
|
Świątkowska‐Flis B, Zdolińska‐Malinowska I, Sługocka D, Boruczkowski D. The use of umbilical cord-derived mesenchymal stem cells in patients with muscular dystrophies: Results from compassionate use in real-life settings. Stem Cells Transl Med 2021; 10:1372-1383. [PMID: 34313400 PMCID: PMC8459640 DOI: 10.1002/sctm.21-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/10/2021] [Accepted: 06/16/2021] [Indexed: 12/15/2022] Open
Abstract
Muscular dystrophies are genetically determined progressive diseases with no cause-related treatment and limited supportive treatment. Although stem cells cannot resolve the underlying genetic conditions, their wide-ranging therapeutic properties may ameliorate the consequences of the involved mutations (oxidative stress, inflammation, mitochondrial dysfunction, necrosis). In this study, we administered advanced therapy medicinal product containing umbilical cord-derived mesenchymal stem cells (UC-MSCs) to 22 patients with muscular dystrophies. Patients received one to five intravenous and/or intrathecal injections per treatment course in up to two courses every 2 months. Four standard doses of 10, 20, 30, or 40 × 106 UC-MSCs per injection were used; the approximate dose per kilogram was 1 × 106 UC-MSCs. Muscle strength was measured with a set of CQ Dynamometer computerized force meters (CQ Elektronik System, Czernica, Poland). Statistical analysis of muscle strength in the whole group showed significant improvement in the right upper limb (+4.0 N); left hip straightening (+4.5 N) and adduction (+0.5 N); right hip straightening (+1.0 N), bending (+7.5 N), and adduction (+2.5 N); right knee straightening (+8.5 N); left shoulder revocation (+13.0 N), straightening (+5.5 N), and bending (+6.5 N); right shoulder adduction (+3.0 N), revocation (+10.5 N), and bending (+5 N); and right elbow straightening (+9.5 N); all these differences were statistically significant. In six patients (27.3%) these changes led to improvement in gait analysis or movement scale result. Only one patient experienced transient headache and lower back pain after the last administration. In conclusion, UC-MSC therapy may be considered as a therapeutic option for these patients.
Collapse
Affiliation(s)
- Beata Świątkowska‐Flis
- Polish Center of Cell Therapy and Immunotherapy in Częstochowa, CM KlaraCzęstochowaPoland
- Faculty of Health SciencesJan Długosz University of Humanities and Life SciencesCzęstochowaPoland
| | | | - Dominika Sługocka
- Polish Center of Cell Therapy and Immunotherapy in Częstochowa, CM KlaraCzęstochowaPoland
| | | |
Collapse
|