151
|
Moyle LA, Davoudi S, Gilbert PM. Innovation in culture systems to study muscle complexity. Exp Cell Res 2021; 411:112966. [PMID: 34906582 DOI: 10.1016/j.yexcr.2021.112966] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/31/2021] [Accepted: 12/04/2021] [Indexed: 11/19/2022]
Abstract
Endogenous skeletal muscle development, regeneration, and pathology are extremely complex processes, influenced by local and systemic factors. Unpinning how these mechanisms function is crucial for fundamental biology and to develop therapeutic interventions for genetic disorders, but also conditions like sarcopenia and volumetric muscle loss. Ex vivo skeletal muscle models range from two- and three-dimensional primary cultures of satellite stem cell-derived myoblasts grown alone or in co-culture, to single muscle myofibers, myobundles, and whole tissues. Together, these systems provide the opportunity to gain mechanistic insights of stem cell behavior, cell-cell interactions, and mature muscle function in simplified systems, without confounding variables. Here, we highlight recent advances (published in the last 5 years) using in vitro primary cells and ex vivo skeletal muscle models, and summarize the new insights, tools, datasets, and screening methods they have provided. Finally, we highlight the opportunity for exponential advance of skeletal muscle knowledge, with spatiotemporal resolution, that is offered by guiding the study of muscle biology and physiology with in silico modelling and implementing high-content cell biology systems and ex vivo physiology platforms.
Collapse
Affiliation(s)
- Louise A Moyle
- Institute of Biomedical Engineering, Toronto, ON, M5S 3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, M5S 3E1, Canada
| | - Sadegh Davoudi
- Institute of Biomedical Engineering, Toronto, ON, M5S 3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, M5S 3E1, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, Toronto, ON, M5S 3G9, Canada; Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON, M5S 3E1, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
152
|
Zhang Y, Shi Y, Lin J, Li X, Yang B, Zhou J. Immune Cell Infiltration Analysis Demonstrates Excessive Mast Cell Activation in Psoriasis. Front Immunol 2021; 12:773280. [PMID: 34887864 PMCID: PMC8650163 DOI: 10.3389/fimmu.2021.773280] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/04/2021] [Indexed: 12/21/2022] Open
Abstract
Psoriasis represents multiple inflammatory processes and exaggerated physiological responses to epithelial damage by innate and adaptive immune components, thus it is critical to compare the immune cell niche in disease and healthy skin. Here, we inferred the proportions of different immune cell types in psoriatic and healthy skin using the CIBERSORT algorithm with expression profiles as input. As a result, we observed a dramatic change of immune cell profiles in psoriatic skin compared with healthy skin. Interestingly, the resting mast cells is almost eliminated in psoriatic skin. In contrast, the activated mast cells are enriched in psoriatic skin, indicating that mast cells activation may play an important role in psoriasis pathogenesis. In addition, we found that the proportion of the resting mast cells gradually come back to the normal level in lesioned skin upon etanercept treatment, suggesting that mast cells play a critical role in immune cell niche maintenance. Further experiments validated a significant decrease in mast cell population and an excessive mast cell activation in psoriatic skin compared with healthy skin. In conclusion, our integrative analyses of the immune cell profiles and the corresponding marker genes expression provide a better understanding of the inflammation response in psoriasis and important clues for clinical applications.
Collapse
Affiliation(s)
- Yongjun Zhang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yanqiang Shi
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jingxia Lin
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xuefei Li
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jiajian Zhou
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
153
|
Liu Q, Deng J, Qiu Y, Gao J, Li J, Guan L, Lee H, Zhou Q, Xiao J. Non-coding RNA basis of muscle atrophy. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:1066-1078. [PMID: 34786211 PMCID: PMC8569427 DOI: 10.1016/j.omtn.2021.10.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Muscle atrophy is a common complication of many chronic diseases including heart failure, cancer cachexia, aging, etc. Unhealthy habits and usage of hormones such as dexamethasone can also lead to muscle atrophy. However, the underlying mechanisms of muscle atrophy are not completely understood. Non-coding RNAs (ncRNAs), such as microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), play vital roles in muscle atrophy. This review mainly discusses the regulation of ncRNAs in muscle atrophy induced by various factors such as heart failure, cancer cachexia, aging, chronic obstructive pulmonary disease (COPD), peripheral nerve injury (PNI), chronic kidney disease (CKD), unhealthy habits, and usage of hormones; highlights the findings of ncRNAs as common regulators in multiple types of muscle atrophy; and summarizes current therapies and underlying mechanisms for muscle atrophy. This review will deepen the understanding of skeletal muscle biology and provide new strategies and insights into gene therapy for muscle atrophy.
Collapse
Affiliation(s)
- Qi Liu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Jiali Deng
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Yan Qiu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Juan Gao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Jin Li
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Longfei Guan
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Hangil Lee
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Qiulian Zhou
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
154
|
Molina T, Fabre P, Dumont NA. Fibro-adipogenic progenitors in skeletal muscle homeostasis, regeneration and diseases. Open Biol 2021; 11:210110. [PMID: 34875199 PMCID: PMC8651418 DOI: 10.1098/rsob.210110] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle possesses a remarkable regenerative capacity that relies on the activity of muscle stem cells, also known as satellite cells. The presence of non-myogenic cells also plays a key role in the coordination of skeletal muscle regeneration. Particularly, fibro-adipogenic progenitors (FAPs) emerged as master regulators of muscle stem cell function and skeletal muscle regeneration. This population of muscle resident mesenchymal stromal cells has been initially characterized based on its bi-potent ability to differentiate into fibroblasts or adipocytes. New technologies such as single-cell RNAseq revealed the cellular heterogeneity of FAPs and their complex regulatory network during muscle regeneration. In acute injury, FAPs rapidly enter the cell cycle and secrete trophic factors that support the myogenic activity of muscle stem cells. Conversely, deregulation of FAP cell activity is associated with the accumulation of fibrofatty tissue in pathological conditions such as muscular dystrophies and ageing. Considering their central role in skeletal muscle pathophysiology, the regulatory mechanisms of FAPs and their cellular and molecular crosstalk with muscle stem cells are highly investigated in the field. In this review, we summarize the current knowledge on FAP cell characteristics, heterogeneity and the cellular crosstalk during skeletal muscle homeostasis and regeneration. We further describe their role in muscular disorders, as well as different therapeutic strategies targeting these cells to restore muscle regeneration.
Collapse
Affiliation(s)
- Thomas Molina
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada,Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Paul Fabre
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada,Department of Pharmacology and Physiology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Nicolas A. Dumont
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada,School of Rehabilitation, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
155
|
Orchard P, Manickam N, Ventresca C, Vadlamudi S, Varshney A, Rai V, Kaplan J, Lalancette C, Mohlke KL, Gallagher K, Burant CF, Parker SCJ. Human and rat skeletal muscle single-nuclei multi-omic integrative analyses nominate causal cell types, regulatory elements, and SNPs for complex traits. Genome Res 2021; 31:2258-2275. [PMID: 34815310 PMCID: PMC8647829 DOI: 10.1101/gr.268482.120] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/16/2021] [Indexed: 12/12/2022]
Abstract
Skeletal muscle accounts for the largest proportion of human body mass, on average, and is a key tissue in complex diseases and mobility. It is composed of several different cell and muscle fiber types. Here, we optimize single-nucleus ATAC-seq (snATAC-seq) to map skeletal muscle cell-specific chromatin accessibility landscapes in frozen human and rat samples, and single-nucleus RNA-seq (snRNA-seq) to map cell-specific transcriptomes in human. We additionally perform multi-omics profiling (gene expression and chromatin accessibility) on human and rat muscle samples. We capture type I and type II muscle fiber signatures, which are generally missed by existing single-cell RNA-seq methods. We perform cross-modality and cross-species integrative analyses on 33,862 nuclei and identify seven cell types ranging in abundance from 59.6% to 1.0% of all nuclei. We introduce a regression-based approach to infer cell types by comparing transcription start site-distal ATAC-seq peaks to reference enhancer maps and show consistency with RNA-based marker gene cell type assignments. We find heterogeneity in enrichment of genetic variants linked to complex phenotypes from the UK Biobank and diabetes genome-wide association studies in cell-specific ATAC-seq peaks, with the most striking enrichment patterns in muscle mesenchymal stem cells (∼3.5% of nuclei). Finally, we overlay these chromatin accessibility maps on GWAS data to nominate causal cell types, SNPs, transcription factor motifs, and target genes for type 2 diabetes signals. These chromatin accessibility profiles for human and rat skeletal muscle cell types are a useful resource for nominating causal GWAS SNPs and cell types.
Collapse
Affiliation(s)
- Peter Orchard
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Nandini Manickam
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Christa Ventresca
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Swarooparani Vadlamudi
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Arushi Varshney
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Vivek Rai
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Jeremy Kaplan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Claudia Lalancette
- Epigenomics Core, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Katherine Gallagher
- Department of Surgery, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
156
|
Singh P, Chazaud B. Benefits and pathologies associated with the inflammatory response. Exp Cell Res 2021; 409:112905. [PMID: 34736921 DOI: 10.1016/j.yexcr.2021.112905] [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: 05/03/2021] [Revised: 10/14/2021] [Accepted: 10/29/2021] [Indexed: 10/20/2022]
Abstract
Adult skeletal muscle regenerates completely after a damage, thanks to the satellite cells, or muscle stem cells (MuSCs), that implement the adult myogenic program. This program is sustained by both robust intrinsic mechanisms and extrinsic cues coming from the close neighborhood of MuSCs during muscle regeneration. Among the various cell types present in the regenerating muscle, immune cells, and particularly macrophages, exert numerous functions and provide sequential transient niches to support the myogenic program. The adequate orchestration of the delivery of these cues ensures efficient muscle regeneration and full functional recovery. The situation is very different in muscular dystrophies where asynchronous and permanent microinjuries occur, triggering contradictory regenerating cues at the same time in a specific area, that lead to chronic inflammation and fibrogenesis. Here we review the beneficial effects that leukocytes, and particularly macrophages, exert on their neighboring cells during skeletal muscle regeneration after an acute injury. Then, the more complicated (and less beneficial) roles of leukocytes during muscular dystrophies are presented. Finally, we discuss how the inflammatory compartment may be a target to improve muscle regeneration in both acute muscle injury and muscle diseases.
Collapse
Affiliation(s)
- Pawandeep Singh
- Institut NeuroMyoGene, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université Lyon, Faculté de Médecine, 8 Avenue Rockefeller, 69008, Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGene, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université Lyon, Faculté de Médecine, 8 Avenue Rockefeller, 69008, Lyon, France.
| |
Collapse
|
157
|
McKellar DW, Walter LD, Song LT, Mantri M, Wang MFZ, De Vlaminck I, Cosgrove BD. Large-scale integration of single-cell transcriptomic data captures transitional progenitor states in mouse skeletal muscle regeneration. Commun Biol 2021; 4:1280. [PMID: 34773081 PMCID: PMC8589952 DOI: 10.1038/s42003-021-02810-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 10/19/2021] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle repair is driven by the coordinated self-renewal and fusion of myogenic stem and progenitor cells. Single-cell gene expression analyses of myogenesis have been hampered by the poor sampling of rare and transient cell states that are critical for muscle repair, and do not inform the spatial context that is important for myogenic differentiation. Here, we demonstrate how large-scale integration of single-cell and spatial transcriptomic data can overcome these limitations. We created a single-cell transcriptomic dataset of mouse skeletal muscle by integration, consensus annotation, and analysis of 23 newly collected scRNAseq datasets and 88 publicly available single-cell (scRNAseq) and single-nucleus (snRNAseq) RNA-sequencing datasets. The resulting dataset includes more than 365,000 cells and spans a wide range of ages, injury, and repair conditions. Together, these data enabled identification of the predominant cell types in skeletal muscle, and resolved cell subtypes, including endothelial subtypes distinguished by vessel-type of origin, fibro-adipogenic progenitors defined by functional roles, and many distinct immune populations. The representation of different experimental conditions and the depth of transcriptome coverage enabled robust profiling of sparsely expressed genes. We built a densely sampled transcriptomic model of myogenesis, from stem cell quiescence to myofiber maturation, and identified rare, transitional states of progenitor commitment and fusion that are poorly represented in individual datasets. We performed spatial RNA sequencing of mouse muscle at three time points after injury and used the integrated dataset as a reference to achieve a high-resolution, local deconvolution of cell subtypes. We also used the integrated dataset to explore ligand-receptor co-expression patterns and identify dynamic cell-cell interactions in muscle injury response. We provide a public web tool to enable interactive exploration and visualization of the data. Our work supports the utility of large-scale integration of single-cell transcriptomic data as a tool for biological discovery.
Collapse
Affiliation(s)
- David W McKellar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Lauren D Walter
- Department of Molecular Biology & Genetics, Cornell University, Ithaca, NY, 14853, USA
| | - Leo T Song
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Madhav Mantri
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Michael F Z Wang
- Department of Computational Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
158
|
Xie N, Chu SN, Azzag K, Schultz CB, Peifer LN, Kyba M, Perlingeiro RCR, Chan SSK. In vitro expanded skeletal myogenic progenitors from pluripotent stem cell-derived teratomas have high engraftment capacity. Stem Cell Reports 2021; 16:2900-2912. [PMID: 34798067 PMCID: PMC8693664 DOI: 10.1016/j.stemcr.2021.10.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/17/2022] Open
Abstract
One major challenge in realizing cell-based therapy for treating muscle-wasting disorders is the difficulty in obtaining therapeutically meaningful amounts of engraftable cells. We have previously described a method to generate skeletal myogenic progenitors with exceptional engraftability from pluripotent stem cells via teratoma formation. Here, we show that these cells are functionally expandable in vitro while retaining their in vivo regenerative potential. Within 37 days in culture, teratoma-derived skeletal myogenic progenitors were expandable to a billion-fold. Similar to their freshly sorted counterparts, the expanded cells expressed PAX7 and were capable of forming multinucleated myotubes in vitro. Importantly, these cells remained highly regenerative in vivo. Upon transplantation, the expanded cells formed new DYSTROPHIN+ fibers that reconstituted up to 40% of tibialis anterior muscle volume and repopulated the muscle stem cell pool. Our study thereby demonstrates the possibility of producing large quantities of engraftable skeletal myogenic cells for transplantation.
Collapse
Affiliation(s)
- Ning Xie
- Department of Pediatrics, University of Minnesota, 2231 6th Street SE, Cancer and Cardiovascular Research Building, Minneapolis, MN 55455 USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA; Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA
| | - Sabrina N Chu
- Department of Pediatrics, University of Minnesota, 2231 6th Street SE, Cancer and Cardiovascular Research Building, Minneapolis, MN 55455 USA
| | - Karim Azzag
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Cassandra B Schultz
- Department of Pediatrics, University of Minnesota, 2231 6th Street SE, Cancer and Cardiovascular Research Building, Minneapolis, MN 55455 USA
| | - Lindsay N Peifer
- Department of Pediatrics, University of Minnesota, 2231 6th Street SE, Cancer and Cardiovascular Research Building, Minneapolis, MN 55455 USA
| | - Michael Kyba
- Department of Pediatrics, University of Minnesota, 2231 6th Street SE, Cancer and Cardiovascular Research Building, Minneapolis, MN 55455 USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA; Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA
| | - Rita C R Perlingeiro
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA; Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA; Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Sunny S K Chan
- Department of Pediatrics, University of Minnesota, 2231 6th Street SE, Cancer and Cardiovascular Research Building, Minneapolis, MN 55455 USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA; Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
159
|
Farup J, Just J, de Paoli F, Lin L, Jensen JB, Billeskov T, Roman IS, Cömert C, Møller AB, Madaro L, Groppa E, Fred RG, Kampmann U, Gormsen LC, Pedersen SB, Bross P, Stevnsner T, Eldrup N, Pers TH, Rossi FMV, Puri PL, Jessen N. Human skeletal muscle CD90 + fibro-adipogenic progenitors are associated with muscle degeneration in type 2 diabetic patients. Cell Metab 2021; 33:2201-2214.e11. [PMID: 34678202 PMCID: PMC9165662 DOI: 10.1016/j.cmet.2021.10.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/29/2021] [Accepted: 10/01/2021] [Indexed: 01/12/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with impaired skeletal muscle function and degeneration of the skeletal muscles. However, the mechanisms underlying the degeneration are not well described in human skeletal muscle. Here we show that skeletal muscle of T2DM patients exhibit degenerative remodeling of the extracellular matrix that is associated with a selective increase of a subpopulation of fibro-adipogenic progenitors (FAPs) marked by expression of THY1 (CD90)-the FAPCD90+. We identify platelet-derived growth factor (PDGF) as a key FAP regulator, as it promotes proliferation and collagen production at the expense of adipogenesis. FAPsCD90+ display a PDGF-mimetic phenotype, with high proliferative activity, clonogenicity, and production of extracellular matrix. FAPCD90+ proliferation was reduced by in vitro treatment with metformin. Furthermore, metformin treatment reduced FAP content in T2DM patients. These data identify a PDGF-driven conversion of a subpopulation of FAPs as a key event in the fibrosis development in T2DM muscle.
Collapse
Affiliation(s)
- Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark.
| | - Jesper Just
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus 8200, Denmark; Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | - Frank de Paoli
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Lin Lin
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jonas Brorson Jensen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Tine Billeskov
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark; Diabetes and Hormonal Diseases, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Ines Sanchez Roman
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus 8000, Denmark; Department of Psychology, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid 28670, Spain
| | - Cagla Cömert
- Molecular Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | - Andreas Buch Møller
- Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Luca Madaro
- Department of AHFMO, University of Rome "la Sapienza," Rome 00185, Italy
| | - Elena Groppa
- The University of British Columbia, Vancouver BC CA V6T, Canada
| | - Rikard Göran Fred
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Ulla Kampmann
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Lars C Gormsen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Steen B Pedersen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Diabetes and Hormonal Diseases, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Peter Bross
- Molecular Research Unit, Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
| | - Tinna Stevnsner
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus 8000, Denmark
| | - Nikolaj Eldrup
- Department of Vascular Surgery, Rigshospitalet, Copenhagen 2100, Denmark
| | - Tune H Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Fabio M V Rossi
- The University of British Columbia, Vancouver BC CA V6T, Canada
| | - Pier Lorenzo Puri
- Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus 8200, Denmark.
| |
Collapse
|
160
|
Collins BC, Kardon G. It takes all kinds: heterogeneity among satellite cells and fibro-adipogenic progenitors during skeletal muscle regeneration. Development 2021; 148:dev199861. [PMID: 34739030 PMCID: PMC8602941 DOI: 10.1242/dev.199861] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Vertebrate skeletal muscle is composed of multinucleate myofibers that are surrounded by muscle connective tissue. Following injury, muscle is able to robustly regenerate because of tissue-resident muscle stem cells, called satellite cells. In addition, efficient and complete regeneration depends on other cells resident in muscle - including fibro-adipogenic progenitors (FAPs). Increasing evidence from single-cell analyses and genetic and transplantation experiments suggests that satellite cells and FAPs are heterogeneous cell populations. Here, we review our current understanding of the heterogeneity of satellite cells, their myogenic derivatives and FAPs in terms of gene expression, anatomical location, age and timing during the regenerative process - each of which have potentially important functional consequences.
Collapse
Affiliation(s)
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
161
|
Wei X, Nicoletti C, Puri PL. Fibro-Adipogenic Progenitors: Versatile keepers of skeletal muscle homeostasis, beyond the response to myotrauma. Semin Cell Dev Biol 2021; 119:23-31. [PMID: 34332886 PMCID: PMC8552908 DOI: 10.1016/j.semcdb.2021.07.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/12/2021] [Accepted: 07/17/2021] [Indexed: 10/20/2022]
Abstract
While Fibro-Adipogenic Progenitors (FAPs) have been originally identified as muscle-interstitial mesenchymal cells activated in response to muscle injury and endowed with inducible fibrogenic and adipogenic potential, subsequent studies have expanded their phenotypic and functional repertoire and revealed their contribution to skeletal muscle response to a vast range of perturbations. Here we review the emerging contribution of FAPs to skeletal muscle responses to motor neuron injuries and to systemic physiological (e.g., exercise) or pathological metabolic (e.g., diabetes) perturbations. We also provide an initial blueprint of discrete sub-clusters of FAPs that are activated by specific perturbations and discuss their role in muscle adaptation to these conditions.
Collapse
Affiliation(s)
- X Wei
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - C Nicoletti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - P L Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
162
|
Control of satellite cell function in muscle regeneration and its disruption in ageing. Nat Rev Mol Cell Biol 2021; 23:204-226. [PMID: 34663964 DOI: 10.1038/s41580-021-00421-2] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2021] [Indexed: 12/19/2022]
Abstract
Skeletal muscle contains a designated population of adult stem cells, called satellite cells, which are generally quiescent. In homeostasis, satellite cells proliferate only sporadically and usually by asymmetric cell division to replace myofibres damaged by daily activity and maintain the stem cell pool. However, satellite cells can also be robustly activated upon tissue injury, after which they undergo symmetric divisions to generate new stem cells and numerous proliferating myoblasts that later differentiate to muscle cells (myocytes) to rebuild the muscle fibre, thereby supporting skeletal muscle regeneration. Recent discoveries show that satellite cells have a great degree of population heterogeneity, and that their cell fate choices during the regeneration process are dictated by both intrinsic and extrinsic mechanisms. Extrinsic cues come largely from communication with the numerous distinct stromal cell types in their niche, creating a dynamically interactive microenvironment. This Review discusses the role and regulation of satellite cells in skeletal muscle homeostasis and regeneration. In particular, we highlight the cell-intrinsic control of quiescence versus activation, the importance of satellite cell-niche communication, and deregulation of these mechanisms associated with ageing. The increasing understanding of how satellite cells are regulated will help to advance muscle regeneration and rejuvenation therapies.
Collapse
|
163
|
Corso D, Chemello F, Alessio E, Urso I, Ferrarese G, Bazzega M, Romualdi C, Lanfranchi G, Sales G, Cagnin S. MyoData: An expression knowledgebase at single cell/nucleus level for the discovery of coding-noncoding RNA functional interactions in skeletal muscle. Comput Struct Biotechnol J 2021; 19:4142-4155. [PMID: 34527188 PMCID: PMC8342900 DOI: 10.1016/j.csbj.2021.07.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 12/22/2022] Open
Abstract
Regulation of gene expression through non-coding RNAs at single myofiber and nucleus resolution. Reinterpretation of KEGG pathways with microRNA and long non-coding RNA activities. miR-149, -214, and let-7e alter mitochondrial shape. The long non-coding RNA Pvt1 is a sponge for miR-27a. miR-208b regulates Sox6; miR-214 regulates both Sox6 and Slc16a3.
Non-coding RNAs represent the largest part of transcribed mammalian genomes and prevalently exert regulatory functions. Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) can modulate the activity of each other. Skeletal muscle is the most abundant tissue in mammals. It is composed of different cell types with myofibers that represent the smallest complete contractile system. Considering that lncRNAs and miRNAs are more cell type-specific than coding RNAs, to understand their function it is imperative to evaluate their expression and action within single myofibers. In this database, we collected gene expression data for coding and non-coding genes in single myofibers and used them to produce interaction networks based on expression correlations. Since biological pathways are more informative than networks based on gene expression correlation, to understand how altered genes participate in the studied phenotype, we integrated KEGG pathways with miRNAs and lncRNAs. The database also integrates single nucleus gene expression data on skeletal muscle in different patho-physiological conditions. We demonstrated that these networks can serve as a framework from which to dissect new miRNA and lncRNA functions to experimentally validate. Some interactions included in the database have been previously experimentally validated using high throughput methods. These can be the basis for further functional studies. Using database information, we demonstrate the involvement of miR-149, -214 and let-7e in mitochondria shaping; the ability of the lncRNA Pvt1 to mitigate the action of miR-27a via sponging; and the regulatory activity of miR-214 on Sox6 and Slc16a3. The MyoData is available at https://myodata.bio.unipd.it.
Collapse
Affiliation(s)
- Davide Corso
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Francesco Chemello
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Enrico Alessio
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Ilenia Urso
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Giulia Ferrarese
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Martina Bazzega
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Chiara Romualdi
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Gerolamo Lanfranchi
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy.,CRIBI Biotechnology Centre, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy.,CIR-Myo Myology Center, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Gabriele Sales
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Stefano Cagnin
- Department of Biology, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy.,CRIBI Biotechnology Centre, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy.,CIR-Myo Myology Center, University of Padova, Via Ugo Bassi 58/b, 35131 Padova, Italy
| |
Collapse
|
164
|
Partridge TA. Enhancing Interrogation of Skeletal Muscle Samples for Informative Quantitative Data. J Neuromuscul Dis 2021; 8:S257-S269. [PMID: 34511511 PMCID: PMC8673506 DOI: 10.3233/jnd-210736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Careful quantitative analysis of histological preparations of muscle samples is crucial to accurate investigation of myopathies in man and of interpretation of data from animals subjected to experimental or potentially therapeutic treatments. Protocols for measuring cell numbers are subject to problems arising from biases associated with preparative and analytical techniques. Prominent among these is the effect of polarized structure of skeletal muscle on sampling bias. It is also common in this tissue to collect data as ratios to convenient reference dominators, the fundamental bases of which are ill-defined, or unrecognized or not accurately assessable. Use of such 'floating' denominators raises a barrier to estimation of the absolute values that assume practical importance in medical research, where accurate comparison between different scenarios in different species is essential to the aim of translating preclinical research findings in animal models to clinical utility in Homo sapiens.This review identifies some of the underappreciated problems with current morphometric practice, some of which are exacerbated in skeletal muscle, and evaluates the extent of their intrusiveness into the of building an objective, accurate, picture of the structure of the muscle sample. It also contains recommendations for eliminating or at least minimizing these problems. Principal among these, would be the use of stereological procedures to avoid the substantial counting biases arising from inter-procedure differences in object size and section thickness.Attention is also drawn to the distortions of interpretation arising from use of undefined or inappropriate denominators.
Collapse
Affiliation(s)
- Terence A Partridge
- Professor of Integrative Systemic Biology, George Washington University, Washington DC.,Honorary Professor, Institute of Child Health, University College London
| |
Collapse
|
165
|
Function and regulation of muscle stem cells in skeletal muscle development and regeneration: a narrative review. JOURNAL OF BIO-X RESEARCH 2021. [DOI: 10.1097/jbr.0000000000000105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
166
|
Leuchtmann AB, Adak V, Dilbaz S, Handschin C. The Role of the Skeletal Muscle Secretome in Mediating Endurance and Resistance Training Adaptations. Front Physiol 2021; 12:709807. [PMID: 34456749 PMCID: PMC8387622 DOI: 10.3389/fphys.2021.709807] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
Exercise, in the form of endurance or resistance training, leads to specific molecular and cellular adaptions not only in skeletal muscles, but also in many other organs such as the brain, liver, fat or bone. In addition to direct effects of exercise on these organs, the production and release of a plethora of different signaling molecules from skeletal muscle are a centerpiece of systemic plasticity. Most studies have so far focused on the regulation and function of such myokines in acute exercise bouts. In contrast, the secretome of long-term training adaptation remains less well understood, and the contribution of non-myokine factors, including metabolites, enzymes, microRNAs or mitochondrial DNA transported in extracellular vesicles or by other means, is underappreciated. In this review, we therefore provide an overview on the current knowledge of endurance and resistance exercise-induced factors of the skeletal muscle secretome that mediate muscular and systemic adaptations to long-term training. Targeting these factors and leveraging their functions could not only have broad implications for athletic performance, but also for the prevention and therapy in diseased and elderly populations.
Collapse
|
167
|
Contreras O, Rossi FMV, Theret M. Origins, potency, and heterogeneity of skeletal muscle fibro-adipogenic progenitors-time for new definitions. Skelet Muscle 2021; 11:16. [PMID: 34210364 PMCID: PMC8247239 DOI: 10.1186/s13395-021-00265-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Striated muscle is a highly plastic and regenerative organ that regulates body movement, temperature, and metabolism-all the functions needed for an individual's health and well-being. The muscle connective tissue's main components are the extracellular matrix and its resident stromal cells, which continuously reshape it in embryonic development, homeostasis, and regeneration. Fibro-adipogenic progenitors are enigmatic and transformative muscle-resident interstitial cells with mesenchymal stem/stromal cell properties. They act as cellular sentinels and physiological hubs for adult muscle homeostasis and regeneration by shaping the microenvironment by secreting a complex cocktail of extracellular matrix components, diffusible cytokines, ligands, and immune-modulatory factors. Fibro-adipogenic progenitors are the lineage precursors of specialized cells, including activated fibroblasts, adipocytes, and osteogenic cells after injury. Here, we discuss current research gaps, potential druggable developments, and outstanding questions about fibro-adipogenic progenitor origins, potency, and heterogeneity. Finally, we took advantage of recent advances in single-cell technologies combined with lineage tracing to unify the diversity of stromal fibro-adipogenic progenitors. Thus, this compelling review provides new cellular and molecular insights in comprehending the origins, definitions, markers, fate, and plasticity of murine and human fibro-adipogenic progenitors in muscle development, homeostasis, regeneration, and repair.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, 2052, Australia.
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Fabio M V Rossi
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Marine Theret
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
168
|
Juban G. Transcriptional control of macrophage inflammatory shift during skeletal muscle regeneration. Semin Cell Dev Biol 2021; 119:82-88. [PMID: 34183241 DOI: 10.1016/j.semcdb.2021.06.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/13/2021] [Accepted: 06/18/2021] [Indexed: 01/05/2023]
Abstract
Skeletal muscle is a tissue able to fully regenerate after an acute injury. Macrophages play an essential role during skeletal muscle regeneration. Resolution of inflammation is a crucial step during the regeneration process, allowing to contain the inflammatory response to avoid damage of the healthy surrounding muscle and triggers the recovery phase during which the muscle regenerates. Resolution of inflammation is mainly mediated by macrophage phenotypic shift that is the transition from a pro-inflammatory damage associated profile towards an anti-inflammatory restorative phenotype, which is characterized by a major transcriptional rewiring. Failure of the resolution of inflammation is observed in chronic diseases such as degenerative myopathies where permanent asynchronous muscle injuries trigger contradictory inflammatory cues, leading to fibrosis and alteration of muscle function. This review will focus on the described molecular pathways that control macrophage inflammatory shift during skeletal muscle regeneration. First, we will highlight the transcriptional changes that characterize macrophage inflammatory shift during skeletal muscle regeneration. Then, we will describe how the signaling pathways and the metabolic changes associated with this shift are controlled. Finally, we will emphasize the transcription factors involved.
Collapse
Affiliation(s)
- Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, Lyon, France.
| |
Collapse
|
169
|
Camps J, Breuls N, Sifrim A, Giarratana N, Corvelyn M, Danti L, Grosemans H, Vanuytven S, Thiry I, Belicchi M, Meregalli M, Platko K, MacDonald ME, Austin RC, Gijsbers R, Cossu G, Torrente Y, Voet T, Sampaolesi M. Interstitial Cell Remodeling Promotes Aberrant Adipogenesis in Dystrophic Muscles. Cell Rep 2021; 31:107597. [PMID: 32375047 DOI: 10.1016/j.celrep.2020.107597] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 03/06/2020] [Accepted: 04/10/2020] [Indexed: 12/26/2022] Open
Abstract
Fibrosis and fat replacement in skeletal muscle are major complications that lead to a loss of mobility in chronic muscle disorders, such as muscular dystrophy. However, the in vivo properties of adipogenic stem and precursor cells remain unclear, mainly due to the high cell heterogeneity in skeletal muscles. Here, we use single-cell RNA sequencing to decomplexify interstitial cell populations in healthy and dystrophic skeletal muscles. We identify an interstitial CD142-positive cell population in mice and humans that is responsible for the inhibition of adipogenesis through GDF10 secretion. Furthermore, we show that the interstitial cell composition is completely altered in muscular dystrophy, with a near absence of CD142-positive cells. The identification of these adipo-regulatory cells in the skeletal muscle aids our understanding of the aberrant fat deposition in muscular dystrophy, paving the way for treatments that could counteract degeneration in patients with muscular dystrophy.
Collapse
Affiliation(s)
- Jordi Camps
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium; Bayer AG, Research & Development, Pharmaceuticals, 13353 Berlin, Germany
| | - Natacha Breuls
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Alejandro Sifrim
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; Wellcome Genome Campus, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Nefele Giarratana
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Marlies Corvelyn
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Laura Danti
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Hanne Grosemans
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Sebastiaan Vanuytven
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Irina Thiry
- Laboratory for Molecular Virology and Gene Therapy, and Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium
| | - Marzia Belicchi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122 Milan, Italy
| | - Mirella Meregalli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122 Milan, Italy
| | - Khrystyna Platko
- Department of Medicine, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada
| | - Melissa E MacDonald
- Department of Medicine, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada
| | - Richard C Austin
- Department of Medicine, The Research Institute of St. Joe's Hamilton, Hamilton Centre for Kidney Research, McMaster University, Hamilton, ON L8N 4A6, Canada
| | - Rik Gijsbers
- Laboratory for Molecular Virology and Gene Therapy, and Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium
| | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Yvan Torrente
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122 Milan, Italy
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; Wellcome Genome Campus, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Maurilio Sampaolesi
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium; Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|
170
|
Yao L, Tichy ED, Zhong L, Mohanty S, Wang L, Ai E, Yang S, Mourkioti F, Qin L. Gli1 Defines a Subset of Fibro-adipogenic Progenitors that Promote Skeletal Muscle Regeneration With Less Fat Accumulation. J Bone Miner Res 2021; 36:1159-1173. [PMID: 33529374 PMCID: PMC8633884 DOI: 10.1002/jbmr.4265] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/20/2021] [Accepted: 01/26/2021] [Indexed: 12/21/2022]
Abstract
Skeletal muscle has remarkable regenerative ability after injury. Mesenchymal fibro-adipogenic progenitors (FAPs) are necessary, active participants during this repair process, but the molecular signatures of these cells and their functional relevance remain largely unexplored. Here, using a lineage tracing mouse model (Gli1-CreER Tomato), we demonstrate that Gli1 marks a small subset of muscle-resident FAPs with elevated Hedgehog (Hh) signaling. Upon notexin muscle injury, these cells preferentially and rapidly expanded within FAPs. Ablation of Gli1+ cells using a DTA mouse model drastically reduced fibroblastic colony-forming unit (CFU-F) colonies generated by muscle cells and impaired muscle repair at 28 days. Pharmacologic manipulation revealed that Gli1+ FAPs rely on Hh signaling to increase the size of regenerating myofiber. Sorted Gli1+ FAPs displayed superior clonogenicity and reduced adipogenic differentiation ability in culture compared to sorted Gli1- FAPs. In a glycerol injury model, Gli1+ FAPs were less likely to give rise to muscle adipocytes compared to other FAPs. Further cell ablation and Hh activator/inhibitor treatments demonstrated their dual actions in enhancing myogenesis and reducing adipogenesis after injury. Examining single-cell RNA-sequencing dataset of FAPs from normal mice indicated that Gli1+ FAPs with increased Hh signaling provide trophic signals to myogenic cells while restrict their own adipogenic differentiation. Collectively, our findings identified a subpopulation of FAPs that play an essential role in skeletal muscle repair. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Orthopaedic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Elisia D Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Sarthak Mohanty
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Luqiang Wang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emily Ai
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shuying Yang
- Department of Basic & Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
171
|
Schüler SC, Kirkpatrick JM, Schmidt M, Santinha D, Koch P, Di Sanzo S, Cirri E, Hemberg M, Ori A, von Maltzahn J. Extensive remodeling of the extracellular matrix during aging contributes to age-dependent impairments of muscle stem cell functionality. Cell Rep 2021; 35:109223. [PMID: 34107247 DOI: 10.1016/j.celrep.2021.109223] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/25/2021] [Accepted: 05/14/2021] [Indexed: 12/19/2022] Open
Abstract
During aging, the regenerative capacity of skeletal muscle decreases due to intrinsic changes in muscle stem cells (MuSCs) and alterations in their niche. Here, we use quantitative mass spectrometry to characterize intrinsic changes in the MuSC proteome and remodeling of the MuSC niche during aging. We generate a network connecting age-affected ligands located in the niche and cell surface receptors on MuSCs. Thereby, we reveal signaling by integrins, Lrp1, Egfr, and Cd44 as the major cell communication axes perturbed through aging. We investigate the effect of Smoc2, a secreted protein that accumulates with aging, primarily originating from fibro-adipogenic progenitors. Increased levels of Smoc2 contribute to the aberrant Integrin beta-1 (Itgb1)/mitogen-activated protein kinase (MAPK) signaling observed during aging, thereby causing impaired MuSC functionality and muscle regeneration. By connecting changes in the proteome of MuSCs to alterations of their niche, our work will enable a better understanding of how MuSCs are affected during aging.
Collapse
Affiliation(s)
- Svenja C Schüler
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Joanna M Kirkpatrick
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Manuel Schmidt
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Deolinda Santinha
- Faculty of Medicine and Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
| | - Philipp Koch
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Simone Di Sanzo
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Emilio Cirri
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Martin Hemberg
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Alessandro Ori
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany.
| | - Julia von Maltzahn
- Leibniz Institute on Aging, Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany.
| |
Collapse
|
172
|
Li J, Knudsen JR, Henriquez-Olguin C, Li Z, Birk JB, Persson KW, Hellsten Y, Offergeld A, Jarassier W, Le Grand F, Schjerling P, Wojtaszewski JFP, Jensen TE. AXIN1 knockout does not alter AMPK/mTORC1 regulation and glucose metabolism in mouse skeletal muscle. J Physiol 2021; 599:3081-3100. [PMID: 33913171 DOI: 10.1113/jp281187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/20/2021] [Indexed: 01/15/2023] Open
Abstract
KEY POINTS Tamoxifen-inducible skeletal muscle-specific AXIN1 knockout (AXIN1 imKO) in mouse does not affect whole-body energy substrate metabolism. AXIN1 imKO does not affect AICAR or insulin-stimulated glucose uptake in adult skeletal muscle. AXIN1 imKO does not affect adult skeletal muscle AMPK or mTORC1 signalling during AICAR/insulin/amino acid incubation, contraction and exercise. During exercise, α2/β2/γ3AMPK and AMP/ATP ratio show greater increases in AXIN1 imKO than wild-type in gastrocnemius muscle. ABSTRACT AXIN1 is a scaffold protein known to interact with >20 proteins in signal transduction pathways regulating cellular development and function. Recently, AXIN1 was proposed to assemble a protein complex essential to catabolic-anabolic transition by coordinating AMPK activation and inactivation of mTORC1 and to regulate glucose uptake-stimulation by both AMPK and insulin. To investigate whether AXIN1 is permissive for adult skeletal muscle function, a phenotypic in vivo and ex vivo characterization of tamoxifen-inducible skeletal muscle-specific AXIN1 knockout (AXIN1 imKO) mice was conducted. AXIN1 imKO did not influence AMPK/mTORC1 signalling or glucose uptake stimulation at rest or in response to different exercise/contraction protocols, pharmacological AMPK activation, insulin or amino acids stimulation. The only genotypic difference observed was in exercising gastrocnemius muscle, where AXIN1 imKO displayed elevated α2/β2/γ3 AMPK activity and AMP/ATP ratio compared to wild-type mice. Our work shows that AXIN1 imKO generally does not affect skeletal muscle AMPK/mTORC1 signalling and glucose metabolism, probably due to functional redundancy of its homologue AXIN2.
Collapse
Affiliation(s)
- Jingwen Li
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jonas R Knudsen
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.,Microsystems Laboratory 2, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Carlos Henriquez-Olguin
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Zhencheng Li
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Jesper B Birk
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kaspar W Persson
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ylva Hellsten
- Section for Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Anika Offergeld
- School of Bioscience, Cardiff University, Cardiff, CF10 3AX, UK
| | - William Jarassier
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France
| | - Fabien Le Grand
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, 69008, France
| | - Peter Schjerling
- Institute of Sports Medicine Copenhagen, Department of Orthopedic Surgery M, Bispebjerg Hospital, Copenhagen, Denmark
| | - Jørgen F P Wojtaszewski
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Thomas E Jensen
- Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
173
|
Julien A, Kanagalingam A, Martínez-Sarrà E, Megret J, Luka M, Ménager M, Relaix F, Colnot C. Direct contribution of skeletal muscle mesenchymal progenitors to bone repair. Nat Commun 2021; 12:2860. [PMID: 34001878 PMCID: PMC8128920 DOI: 10.1038/s41467-021-22842-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/29/2021] [Indexed: 12/13/2022] Open
Abstract
Bone regenerates by activation of tissue resident stem/progenitor cells, formation of a fibrous callus followed by deposition of cartilage and bone matrices. Here, we show that mesenchymal progenitors residing in skeletal muscle adjacent to bone mediate the initial fibrotic response to bone injury and also participate in cartilage and bone formation. Combined lineage and single-cell RNA sequencing analyses reveal that skeletal muscle mesenchymal progenitors adopt a fibrogenic fate before they engage in chondrogenesis after fracture. In polytrauma, where bone and skeletal muscle are injured, skeletal muscle mesenchymal progenitors exhibit altered fibrogenesis and chondrogenesis. This leads to impaired bone healing, which is due to accumulation of fibrotic tissue originating from skeletal muscle and can be corrected by the anti-fibrotic agent Imatinib. These results elucidate the central role of skeletal muscle in bone regeneration and provide evidence that skeletal muscle can be targeted to prevent persistent callus fibrosis and improve bone healing after musculoskeletal trauma.
Collapse
Affiliation(s)
- Anais Julien
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France
| | | | | | - Jérome Megret
- Cytometry core facility, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris, France
| | - Marine Luka
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Université de Paris, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Mickaël Ménager
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Université de Paris, Imagine Institute, INSERM UMR 1163, Paris, France
| | | | - Céline Colnot
- Univ Paris Est Creteil, INSERM, IMRB, Creteil, France.
| |
Collapse
|
174
|
Verma RX, Kannan S, Lin BL, Fomchenko KM, Nieuwenhuis TO, Patil AH, Lukban C, Yang X, Fox-Talbot K, McCall MN, Kwon C, Kass DA, Rosenberg AZ, Halushka MK. Single cell RNA-seq analysis of the flexor digitorum brevis mouse myofibers. Skelet Muscle 2021; 11:13. [PMID: 34001262 PMCID: PMC8127317 DOI: 10.1186/s13395-021-00269-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/28/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Skeletal muscle myofibers can be separated into functionally distinct cell types that differ in gene and protein expression. Current single cell expression data is generally based upon single nucleus RNA, rather than whole myofiber material. We examined if a whole-cell flow sorting approach could be applied to perform single cell RNA-seq (scRNA-seq) in a single muscle type. METHODS We performed deep, whole cell, scRNA-seq on intact and fragmented skeletal myofibers from the mouse fast-twitch flexor digitorum brevis muscle utilizing a flow-gated method of large cell isolation. We performed deep sequencing of 763 intact and fragmented myofibers. RESULTS Quality control metrics across the different gates indicated only 171 of these cells were optimal, with a median read count of 239,252 and an average of 12,098 transcripts per cell. scRNA-seq identified three clusters of myofibers (a slow/fast 2A cluster and two fast 2X clusters). Comparison to a public skeletal nuclear RNA-seq dataset demonstrated a diversity in transcript abundance by method. RISH validated multiple genes across fast and slow twitch skeletal muscle types. CONCLUSION This study introduces and validates a method to isolate intact skeletal muscle myofibers to generate deep expression patterns and expands the known repertoire of fiber-type-specific genes.
Collapse
Affiliation(s)
- Rohan X. Verma
- Department of Pathology, Johns Hopkins University School of Medicine, Ross Bldg. Rm 632B, 720 Rutland Avenue, Baltimore, MD 21205 USA
| | - Suraj Kannan
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Brian L. Lin
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Katherine M. Fomchenko
- Department of Pathology, Johns Hopkins University School of Medicine, Ross Bldg. Rm 632B, 720 Rutland Avenue, Baltimore, MD 21205 USA
| | - Tim O. Nieuwenhuis
- Department of Pathology, Johns Hopkins University School of Medicine, Ross Bldg. Rm 632B, 720 Rutland Avenue, Baltimore, MD 21205 USA
| | - Arun H. Patil
- Department of Pathology, Johns Hopkins University School of Medicine, Ross Bldg. Rm 632B, 720 Rutland Avenue, Baltimore, MD 21205 USA
| | - Clarisse Lukban
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Xiaoping Yang
- Department of Pathology, Johns Hopkins University School of Medicine, Ross Bldg. Rm 632B, 720 Rutland Avenue, Baltimore, MD 21205 USA
| | - Karen Fox-Talbot
- Department of Pathology, Johns Hopkins University School of Medicine, Ross Bldg. Rm 632B, 720 Rutland Avenue, Baltimore, MD 21205 USA
| | - Matthew N. McCall
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - David A. Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine, Ross Bldg. Rm 632B, 720 Rutland Avenue, Baltimore, MD 21205 USA
| | - Marc K. Halushka
- Department of Pathology, Johns Hopkins University School of Medicine, Ross Bldg. Rm 632B, 720 Rutland Avenue, Baltimore, MD 21205 USA
| |
Collapse
|
175
|
Contreras O, Córdova-Casanova A, Brandan E. PDGF-PDGFR network differentially regulates the fate, migration, proliferation, and cell cycle progression of myogenic cells. Cell Signal 2021; 84:110036. [PMID: 33971280 DOI: 10.1016/j.cellsig.2021.110036] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/22/2022]
Abstract
Platelet-derived growth factors (PDGFs) regulate embryonic development, tissue regeneration, and wound healing through their binding to PDGF receptors, PDGFRα and PDGFRβ. However, the role of PDGF signaling in regulating muscle development and regeneration remains elusive, and the cellular and molecular responses of myogenic cells are understudied. Here, we explore the PDGF-PDGFR gene expression changes and their involvement in skeletal muscle myogenesis and myogenic fate. By surveying bulk RNA sequencing and single-cell profiling data of skeletal muscle stem cells, we show that myogenic progenitors and muscle stem cells differentially express PDGF ligands and PDGF receptors during myogenesis. Quiescent adult muscle stem cells and myoblasts preferentially express PDGFRβ over PDGFRα. Remarkably, cell culture- and injury-induced muscle stem cell activation altered PDGF family gene expression. In myoblasts, PDGF-AB and PDGF-BB treatments activate two pro-chemotactic and pro-mitogenic downstream transducers, RAS-ERK1/2 and PI3K-AKT. PDGFRs inhibitor AG1296 inhibited ERK1/2 and AKT activation, myoblast migration, proliferation, and cell cycle progression induced by PDGF-AB and PDGF-BB. We also found that AG1296 causes myoblast G0/G1 cell cycle arrest. Remarkably, PDGF-AA did not promote a noticeable ERK1/2 or AKT activation, myoblast migration, or expansion. Also, myogenic differentiation reduced the expression of both PDGFRα and PDGFRβ, whereas forced PDGFRα expression impaired myogenesis. Thus, our data highlight PDGF signaling pathway to stimulate satellite cell proliferation aiming to enhance skeletal muscle regeneration and provide a deeper understanding of the role of PDGF signaling in non-fibroblastic cells.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington 2052, Australia; Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile.
| | - Adriana Córdova-Casanova
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile
| | - Enrique Brandan
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150 Santiago, Chile; Fundación Ciencia & Vida, 7780272 Santiago, Chile
| |
Collapse
|
176
|
Kok HJ, Barton ER. Actions and interactions of IGF-I and MMPs during muscle regeneration. Semin Cell Dev Biol 2021; 119:11-22. [PMID: 33962867 DOI: 10.1016/j.semcdb.2021.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/04/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022]
Abstract
Muscle regeneration requires the coordination of several factors to mobilize satellite cells and macrophages, remodel the extracellular matrix surrounding muscle fibers, and repair existing and/or form new muscle fibers. In this review, we focus on insulin-like growth factor I and the matrix metalloproteinases, which are secreted proteins that act on cells and the matrix to resolve damage. While their actions appear independent, their interactions occur at the transcriptional and post-translational levels to promote feed-forward activation of each other. Together, these proteins assist at virtually every step of the repair process, and contribute significantly to muscle regenerative capacity.
Collapse
Affiliation(s)
- Hui Jean Kok
- Applied Physiology & Kinesiology, College of Health and Human Performance, University of Florida, 1864 Stadium Road, Gainesville, FL 32611, USA
| | - Elisabeth R Barton
- Applied Physiology & Kinesiology, College of Health and Human Performance, University of Florida, 1864 Stadium Road, Gainesville, FL 32611, USA.
| |
Collapse
|
177
|
Mittal A, Dua A, Gupta S, Injeti E. A research update: Significance of cytokine storm and diaphragm in COVID-19. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100031. [PMID: 34870147 PMCID: PMC8086263 DOI: 10.1016/j.crphar.2021.100031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/31/2021] [Accepted: 04/26/2021] [Indexed: 12/23/2022] Open
Abstract
Emerging research on severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) shows that it is spreading to multiple organs in addition to the respiratory system. Though the SARS-CoV2 enters the human body by binding to ACE2 receptors on pulmonary alveolar cells, recent studies indicate that it is spreading to the central nervous system, cardiac and skeletal muscles leading to various pathological conditions in these organs. In particular, the effects of SARS-CoV-2 on triggering the cytokine storm and its consequential effects on skeletal muscles has generated a lot of discussion. The effects of this virus on muscular function especially in susceptible elderly populations is still being explored. However, its effects on diaphragm, a respiratory muscle which plays an important role in determining lung capacity are not completely explored. Currently, as new evidence on using lung ultrasounds to confirm COVID-19 diagnosis is gaining traction, it is necessary to explore the role of diaphragm in treating COVID-19 patients. This article will review the effects of cytokine storm triggered by the SARS-CoV-2 and its resultant effects on skeletal muscle with a specific focus on the diaphragm in order to identify knowledge gaps in effectively treating COVID-19 patients, especially those who are on a mechanical ventilator.
Collapse
Affiliation(s)
- Ashwani Mittal
- Skeletal Muscle Laboratory, Institute of Integrated & Honors Studies, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Anita Dua
- Skeletal Muscle Laboratory, Institute of Integrated & Honors Studies, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Sanjeev Gupta
- Skeletal Muscle Laboratory, Institute of Integrated & Honors Studies, Kurukshetra University, Kurukshetra, Haryana, 136119, India
| | - Elisha Injeti
- Cedarville University School of Pharmacy, Cedarville, OH, 45314, USA
| |
Collapse
|
178
|
A new isolation and culture method for granulosa cells. Cell Tissue Bank 2021; 22:719-726. [PMID: 33914204 DOI: 10.1007/s10561-021-09929-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/19/2021] [Indexed: 11/25/2022]
Abstract
Granulosa cells are the cell population who have an increasing importance in the female genital system and reproduction. Thus, nowadays in vitro studies to address these cells are also gaining importance and attracts researcher's attention. The aim of our study is to develop a more feasible, low-cost granulosa cell isolation and culture method compared to methods defined so far. Granulosa cells were isolated from follicular fluids obtained from both healthy women donors (n = 19) and polycystic ovary syndrome (n = 15) applied to in vitro fertilization treatment process. Granulosa cells were isolated by using Lymphosep® separation fluid that was not used for this purpose before. The isolated cells were cultured in suitable culture dishes with a mixture of BIO-AMFTM-1 and DMEM/F12 in the first seeding and only complete DMEM/F12 in the following feeds. Complete medium contains only 5% fetal calf serum, 4% L-glutamine and 1% penicillin-streptomycin-amphotericin. The new methods we have developed in granulosa cell isolation and in vitro culture have been successful. Reduction in supplement types and amount; improved the proliferation rate of the granulosa cells in culture. Our new methods of isolation and cell culture for granulosa cells from healthy women, have been also successful in samples of polycystic ovarian patients. With these developed methods, granulosa cells, which belong to humans and have an important role in the ovary, could be isolated and subsequently be maintained to reproduce (proliferate) more easily and cheaper.
Collapse
|
179
|
Proietti D, Giordani L, De Bardi M, D’Ercole C, Lozanoska-Ochser B, Amadio S, Volonté C, Marinelli S, Muchir A, Bouché M, Borsellino G, Sacco A, Puri PL, Madaro L. Activation of skeletal muscle-resident glial cells upon nerve injury. JCI Insight 2021; 6:143469. [PMID: 33661767 PMCID: PMC8119188 DOI: 10.1172/jci.insight.143469] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/25/2021] [Indexed: 01/06/2023] Open
Abstract
Here, we report on the identification of Itga7-expressing muscle-resident glial cells activated by loss of neuromuscular junction (NMJ) integrity. Gene expression analysis at the bulk and single-cell level revealed that these cells are distinct from Itga7-expressing muscle satellite cells. We show that a selective activation and expansion of Itga7+ glial cells occur in response to muscle nerve lesion. Upon activation, muscle glial-derived progenies expressed neurotrophic genes, including nerve growth factor receptor, which enables their isolation by FACS. We show that activated muscle glial cells also expressed genes potentially implicated in extracellular matrix remodeling at NMJs. We found that tenascin C, which was highly expressed by muscle glial cells, activated upon nerve injury and preferentially localized to NMJ. Interestingly, we observed that the activation of muscle glial cells by acute nerve injury was reversible upon NMJ repair. By contrast, in a mouse model of ALS, in which NMJ degeneration is progressive, muscle glial cells steadily increased over the course of the disease. However, they exhibited an impaired neurotrophic activity, suggesting that pathogenic activation of glial cells may be implicated in ALS progression.
Collapse
Affiliation(s)
- Daisy Proietti
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | - Lorenzo Giordani
- Sorbonne Université, INSERM UMRS 974, Association Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | | | - Chiara D’Ercole
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | - Biliana Lozanoska-Ochser
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | | | - Cinzia Volonté
- IRCCS Fondazione Santa Lucia, Rome, Italy
- CNR, National Research Council, Institute for Systems Analysis and Computer Science, Rome, Italy
| | - Sara Marinelli
- CNR, National Research Council, Institute of Biochemistry and Cell Biology, Monterotondo Scalo, Rome, Italy
| | - Antoine Muchir
- Sorbonne Université, INSERM UMRS 974, Association Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Marina Bouché
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | | | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Luca Madaro
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| |
Collapse
|
180
|
Blackburn DM, Lazure F, Soleimani VD. SMART approaches for genome-wide analyses of skeletal muscle stem and niche cells. Crit Rev Biochem Mol Biol 2021; 56:284-300. [PMID: 33823731 DOI: 10.1080/10409238.2021.1908950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Muscle stem cells (MuSCs) also called satellite cells are the building blocks of skeletal muscle, the largest tissue in the human body which is formed primarily of myofibers. While MuSCs are the principal cells that directly contribute to the formation of the muscle fibers, their ability to do so depends on critical interactions with a vast array of nonmyogenic cells within their niche environment. Therefore, understanding the nature of communication between MuSCs and their niche is of key importance to understand how the skeletal muscle is maintained and regenerated after injury. MuSCs are rare and therefore difficult to study in vivo within the context of their niche environment. The advent of single-cell technologies, such as switching mechanism at 5' end of the RNA template (SMART) and tagmentation based technologies using hyperactive transposase, afford the unprecedented opportunity to perform whole transcriptome and epigenome studies on rare cells within their niche environment. In this review, we will delve into how single-cell technologies can be applied to the study of MuSCs and muscle-resident niche cells and the impact this can have on our understanding of MuSC biology and skeletal muscle regeneration.
Collapse
Affiliation(s)
- Darren M Blackburn
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| |
Collapse
|
181
|
Progressive and Coordinated Mobilization of the Skeletal Muscle Niche throughout Tissue Repair Revealed by Single-Cell Proteomic Analysis. Cells 2021; 10:cells10040744. [PMID: 33800595 PMCID: PMC8066646 DOI: 10.3390/cells10040744] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/02/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Skeletal muscle is one of the only mammalian tissues capable of rapid and efficient regeneration after trauma or in pathological conditions. Skeletal muscle regeneration is driven by the muscle satellite cells, the stem cell population in interaction with their niche. Upon injury, muscle fibers undergo necrosis and muscle stem cells activate, proliferate and fuse to form new myofibers. In addition to myogenic cell populations, interaction with other cell types such as inflammatory cells, mesenchymal (fibroadipogenic progenitors—FAPs, pericytes) and vascular (endothelial) lineages are important for efficient muscle repair. While the role of the distinct populations involved in skeletal muscle regeneration is well characterized, the quantitative changes in the muscle stem cell and niche during the regeneration process remain poorly characterized. Methods: We have used mass cytometry to follow the main muscle cell types (muscle stem cells, vascular, mesenchymal and immune cell lineages) during early activation and over the course of muscle regeneration at D0, D2, D5 and D7 compared with uninjured muscles. Results: Early activation induces a number of rapid changes in the proteome of multiple cell types. Following the induction of damage, we observe a drastic loss of myogenic, vascular and mesenchymal cell lineages while immune cells invade the damaged tissue to clear debris and promote muscle repair. Immune cells constitute up to 80% of the mononuclear cells 5 days post-injury. We show that muscle stem cells are quickly activated in order to form new myofibers and reconstitute the quiescent muscle stem cell pool. In addition, our study provides a quantitative analysis of the various myogenic populations during muscle repair. Conclusions: We have developed a mass cytometry panel to investigate the dynamic nature of muscle regeneration at a single-cell level. Using our panel, we have identified early changes in the proteome of stressed satellite and niche cells. We have also quantified changes in the major cell types of skeletal muscle during regeneration and analyzed myogenic transcription factor expression in satellite cells throughout this process. Our results highlight the progressive dynamic shifts in cell populations and the distinct states of muscle stem cells adopted during skeletal muscle regeneration. Our findings give a deeper understanding of the cellular and molecular aspects of muscle regeneration.
Collapse
|
182
|
De Micheli AJ, Laurilliard EJ, Heinke CL, Ravichandran H, Fraczek P, Soueid-Baumgarten S, De Vlaminck I, Elemento O, Cosgrove BD. Single-Cell Analysis of the Muscle Stem Cell Hierarchy Identifies Heterotypic Communication Signals Involved in Skeletal Muscle Regeneration. Cell Rep 2021; 30:3583-3595.e5. [PMID: 32160558 DOI: 10.1016/j.celrep.2020.02.067] [Citation(s) in RCA: 219] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/12/2019] [Accepted: 02/14/2020] [Indexed: 12/15/2022] Open
Abstract
Muscle regeneration relies on the regulation of muscle stem cells (MuSCs) through paracrine signaling interactions. We analyzed muscle regeneration in mice using single-cell RNA sequencing (scRNA-seq) and generated over 34,000 single-cell transcriptomes spanning four time-points. We identified 15 distinct cell types including heterogenous populations of muscle stem and progenitor cells. We resolved a hierarchical map of these myogenic cells by trajectory inference and observed stage-specific regulatory programs within this continuum. Through ligand-receptor interaction analysis, we identified over 100 candidate regeneration-associated paracrine communication pairs between MuSCs and non-myogenic cells. We show that myogenic stem/progenitor cells exhibit heterogeneous expression of multiple Syndecan proteins in cycling myogenic cells, suggesting that Syndecans may coordinate myogenic fate regulation. We performed ligand stimulation in vitro and confirmed that three paracrine factors (FGF2, TGFβ1, and RSPO3) regulate myogenic cell proliferation in a Syndecan-dependent manner. Our study provides a scRNA-seq reference resource to investigate cell communication interactions in muscle regeneration.
Collapse
Affiliation(s)
- Andrea J De Micheli
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Emily J Laurilliard
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Charles L Heinke
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Hiranmayi Ravichandran
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Paula Fraczek
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
183
|
Negative elongation factor regulates muscle progenitor expansion for efficient myofiber repair and stem cell pool repopulation. Dev Cell 2021; 56:1014-1029.e7. [PMID: 33735618 DOI: 10.1016/j.devcel.2021.02.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 01/08/2021] [Accepted: 02/19/2021] [Indexed: 11/24/2022]
Abstract
Negative elongation factor (NELF) is a critical transcriptional regulator that stabilizes paused RNA polymerase to permit rapid gene expression changes in response to environmental cues. Although NELF is essential for embryonic development, its role in adult stem cells remains unclear. In this study, through a muscle-stem-cell-specific deletion, we showed that NELF is required for efficient muscle regeneration and stem cell pool replenishment. In mechanistic studies using PRO-seq, single-cell trajectory analyses and myofiber cultures revealed that NELF works at a specific stage of regeneration whereby it modulates p53 signaling to permit massive expansion of muscle progenitors. Strikingly, transplantation experiments indicated that these progenitors are also necessary for stem cell pool repopulation, implying that they are able to return to quiescence. Thus, we identified a critical role for NELF in the expansion of muscle progenitors in response to injury and revealed that progenitors returning to quiescence are major contributors to the stem cell pool repopulation.
Collapse
|
184
|
Yartseva V, Goldstein LD, Rodman J, Kates L, Chen MZ, Chen YJJ, Foreman O, Siebel CW, Modrusan Z, Peterson AS, Jovičić A. Heterogeneity of Satellite Cells Implicates DELTA1/NOTCH2 Signaling in Self-Renewal. Cell Rep 2021; 30:1491-1503.e6. [PMID: 32023464 DOI: 10.1016/j.celrep.2019.12.100] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/27/2019] [Accepted: 12/30/2019] [Indexed: 12/20/2022] Open
Abstract
How satellite cells and their progenitors balance differentiation and self-renewal to achieve sustainable tissue regeneration is not well understood. A major roadblock to understanding satellite cell fate decisions has been the difficulty of studying this process in vivo. By visualizing expression dynamics of myogenic transcription factors during early regeneration in vivo, we identify the time point at which cells undergo decisions to differentiate or self-renew. Single-cell RNA sequencing reveals heterogeneity of satellite cells, including a subpopulation enriched in Notch2 receptor expression, during both muscle homeostasis and regeneration. Furthermore, we reveal that differentiating cells express the Dll1 ligand. Using antagonistic antibodies, we demonstrate that the DLL1 and NOTCH2 signaling pair is required for satellite cell self-renewal. Thus, differentiating cells provide the self-renewing signal during regeneration, enabling proportional regeneration in response to injury while maintaining the satellite cell pool. These findings have implications for therapeutic control of muscle regeneration.
Collapse
Affiliation(s)
- Valeria Yartseva
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA; Department of Neuroscience, Genentech Inc., South San Francisco, CA 94080, USA
| | - Leonard D Goldstein
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA; Department of Bioinformatics & Computational Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Julia Rodman
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Lance Kates
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Mark Z Chen
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Ying-Jiun J Chen
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA; Department of Protein Chemistry, Genentech Inc., South San Francisco, CA 94080, USA
| | - Oded Foreman
- Department of Pathology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Christian W Siebel
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Zora Modrusan
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA; Department of Protein Chemistry, Genentech Inc., South San Francisco, CA 94080, USA
| | - Andrew S Peterson
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA; Seven Rivers Genomic Medicines, MedGenome, Foster City, CA, USA
| | - Ana Jovičić
- Department of Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA; Department of Neuroscience, Genentech Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
185
|
Girard D, Torossian F, Oberlin E, Alexander KA, Gueguen J, Tseng HW, Genêt F, Lataillade JJ, Salga M, Levesque JP, Le Bousse-Kerdilès MC, Banzet S. Neurogenic Heterotopic Ossifications Recapitulate Hematopoietic Stem Cell Niche Development Within an Adult Osteogenic Muscle Environment. Front Cell Dev Biol 2021; 9:611842. [PMID: 33748104 PMCID: PMC7973025 DOI: 10.3389/fcell.2021.611842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Hematopoiesis and bone interact in various developmental and pathological processes. Neurogenic heterotopic ossifications (NHO) are the formation of ectopic hematopoietic bones in peri-articular muscles that develop following severe lesions of the central nervous system such as traumatic cerebral or spinal injuries or strokes. This review will focus on the hematopoietic facet of NHO. The characterization of NHO demonstrates the presence of hematopoietic marrow in which quiescent hematopoietic stem cells (HSC) are maintained by a functional stromal microenvironment, thus documenting that NHOs are neo-formed ectopic HSC niches. Similarly to adult bone marrow, the NHO permissive environment supports HSC maintenance, proliferation and differentiation through bidirectional signaling with mesenchymal stromal cells and endothelial cells, involving cell adhesion molecules, membrane-bound growth factors, hormones, and secreted matrix proteins. The participation of the nervous system, macrophages and inflammatory cytokines including oncostatin M and transforming growth factor (TGF)-β in this process, reveals how neural circuitry fine-tunes the inflammatory response to generate hematopoietic bones in injured muscles. The localization of NHOs in the peri-articular muscle environment also suggests a role of muscle mesenchymal cells and bone metabolism in development of hematopoiesis in adults. Little is known about the establishment of bone marrow niches and the regulation of HSC cycling during fetal development. Similarities between NHO and development of fetal bones make NHOs an interesting model to study the establishment of bone marrow hematopoiesis during development. Conversely, identification of stage-specific factors that specify HSC developmental state during fetal bone development will give more mechanistic insights into NHO.
Collapse
Affiliation(s)
- Dorothée Girard
- INSERM UMRS-MD 1197, Institut de Recherche Biomédicale des Armées (IRBA), Clamart, France
| | - Frédéric Torossian
- INSERM UMRS-MD 1197, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France
| | - Estelle Oberlin
- INSERM UMRS-MD 1197, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France
| | - Kylie A. Alexander
- Mater Research Institute—The University of Queensland, Woolloongabba, QLD, Australia
| | - Jules Gueguen
- INSERM UMRS-MD 1197, Institut de Recherche Biomédicale des Armées (IRBA), Clamart, France
| | - Hsu-Wen Tseng
- Mater Research Institute—The University of Queensland, Woolloongabba, QLD, Australia
| | - François Genêt
- INSERM U1179, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Versailles, France
| | | | - Marjorie Salga
- INSERM U1179, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Versailles, France
| | - Jean-Pierre Levesque
- Mater Research Institute—The University of Queensland, Woolloongabba, QLD, Australia
| | | | - Sébastien Banzet
- INSERM UMRS-MD 1197, Institut de Recherche Biomédicale des Armées (IRBA), Clamart, France
| |
Collapse
|
186
|
Muscle Diversity, Heterogeneity, and Gradients: Learning from Sarcoglycanopathies. Int J Mol Sci 2021; 22:ijms22052502. [PMID: 33801487 PMCID: PMC7958856 DOI: 10.3390/ijms22052502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle, the most abundant tissue in the body, is heterogeneous. This heterogeneity forms the basis of muscle diversity, which is reflected in the specialized functions of muscles in different parts of the body. However, these different parts are not always clearly delimitated, and this often gives rise to gradients within the same muscle and even across the body. During the last decade, several studies on muscular disorders both in mice and in humans have observed particular distribution patterns of muscle weakness during disease, indicating that the same mutation can affect muscles differently. Moreover, these phenotypical differences reveal gradients of severity, existing alongside other architectural gradients. These two factors are especially prominent in sarcoglycanopathies. Nevertheless, very little is known about the mechanism(s) driving the phenotypic diversity of the muscles affected by these diseases. Here, we will review the available literature on sarcoglycanopathies, focusing on phenotypic differences among affected muscles and gradients, characterization techniques, molecular signatures, and cell population heterogeneity, highlighting the possibilities opened up by new technologies. This review aims to revive research interest in the diverse disease phenotype affecting different muscles, in order to pave the way for new therapeutic interventions.
Collapse
|
187
|
Wong A, Garcia SM, Tamaki S, Striedinger K, Barruet E, Hansen SL, Young DM, Pomerantz JH. Satellite cell activation and retention of muscle regenerative potential after long-term denervation. Stem Cells 2021; 39:331-344. [PMID: 33326654 DOI: 10.1002/stem.3316] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
Irreversible denervation atrophy remains an unsolved clinical problem, and the role of skeletal muscle stem cell (MuSC, satellite cell) depletion in this process is unclear. We investigated the ability of MuSCs to regenerate muscle in the context of denervation. Three to 12 months following sciatic denervation in mice, MuSC number, size, EdU uptake, rate of division, and mitochondrial activity were increased. Following acute myotoxin injury, denervated muscles formed new muscle fibers in situ. MuSCs isolated via flow cytometry from denervated mouse muscle, or from atrophic denervated gluteus maximus muscles of humans with complete spinal cord injuries two decades prior, formed new muscle fibers and reoccupied the anatomic niche after transplantation into uninjured muscle. Our results show unequivocally that, even after prolonged denervation, MuSCs retain intrinsic regenerative potential similar to that of uninjured MuSCs. Treatment of denervation atrophy will require elucidating the non-MuSC environmental changes in muscle that prevent functional regeneration.
Collapse
Affiliation(s)
- Alvin Wong
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Steven M Garcia
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Stanley Tamaki
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Katharine Striedinger
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Emilie Barruet
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Scott L Hansen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - David M Young
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
| | - Jason H Pomerantz
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Program in Craniofacial Biology, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, California, USA
- Department of Orofacial Sciences, University of California, San Francisco, California, USA
| |
Collapse
|
188
|
Millward DJ. Interactions between Growth of Muscle and Stature: Mechanisms Involved and Their Nutritional Sensitivity to Dietary Protein: The Protein-Stat Revisited. Nutrients 2021; 13:729. [PMID: 33668846 PMCID: PMC7996181 DOI: 10.3390/nu13030729] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Childhood growth and its sensitivity to dietary protein is reviewed within a Protein-Stat model of growth regulation. The coordination of growth of muscle and stature is a combination of genetic programming, and of two-way mechanical interactions involving the mechanotransduction of muscle growth through stretching by bone length growth, the core Protein-Stat feature, and the strengthening of bone through muscle contraction via the mechanostat. Thus, growth in bone length is the initiating event and this is always observed. Endocrine and cellular mechanisms of growth in stature are reviewed in terms of the growth hormone-insulin like growth factor-1 (GH-IGF-1) and thyroid axes and the sex hormones, which together mediate endochondral ossification in the growth plate and bone lengthening. Cellular mechanisms of muscle growth during development are then reviewed identifying (a) the difficulties posed by the need to maintain its ultrastructure during myofibre hypertrophy within the extracellular matrix and the concept of muscle as concentric "bags" allowing growth to be conceived as bag enlargement and filling, (b) the cellular and molecular mechanisms involved in the mechanotransduction of satellite and mesenchymal stromal cells, to enable both connective tissue remodelling and provision of new myonuclei to aid myofibre hypertrophy and (c) the implications of myofibre hypertrophy for protein turnover within the myonuclear domain. Experimental data from rodent and avian animal models illustrate likely changes in DNA domain size and protein turnover during developmental and stretch-induced muscle growth and between different muscle fibre types. Growth of muscle in male rats during adulthood suggests that "bag enlargement" is achieved mainly through the action of mesenchymal stromal cells. Current understanding of the nutritional regulation of protein deposition in muscle, deriving from experimental studies in animals and human adults, is reviewed, identifying regulation by amino acids, insulin and myofibre volume changes acting to increase both ribosomal capacity and efficiency of muscle protein synthesis via the mechanistic target of rapamycin complex 1 (mTORC1) and the phenomenon of a "bag-full" inhibitory signal has been identified in human skeletal muscle. The final section deals with the nutritional sensitivity of growth of muscle and stature to dietary protein in children. Growth in length/height as a function of dietary protein intake is described in the context of the breastfed child as the normative growth model, and the "Early Protein Hypothesis" linking high protein intakes in infancy to later adiposity. The extensive paediatric studies on serum IGF-1 and child growth are reviewed but their clinical relevance is of limited value for understanding growth regulation; a role in energy metabolism and homeostasis, acting with insulin to mediate adiposity, is probably more important. Information on the influence of dietary protein on muscle mass per se as opposed to lean body mass is limited but suggests that increased protein intake in children is unable to promote muscle growth in excess of that linked to genotypic growth in length/height. One possible exception is milk protein intake, which cohort and cross-cultural studies suggest can increase height and associated muscle growth, although such effects have yet to be demonstrated by randomised controlled trials.
Collapse
Affiliation(s)
- D Joe Millward
- Department of Nutritional Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| |
Collapse
|
189
|
Machado L, Geara P, Camps J, Dos Santos M, Teixeira-Clerc F, Van Herck J, Varet H, Legendre R, Pawlotsky JM, Sampaolesi M, Voet T, Maire P, Relaix F, Mourikis P. Tissue damage induces a conserved stress response that initiates quiescent muscle stem cell activation. Cell Stem Cell 2021; 28:1125-1135.e7. [PMID: 33609440 DOI: 10.1016/j.stem.2021.01.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 10/30/2020] [Accepted: 01/22/2021] [Indexed: 12/27/2022]
Abstract
Tissue damage dramatically alters how cells interact with their microenvironment. These changes in turn dictate cellular responses, such as stem cell activation, yet early cellular responses in vivo remain ill defined. We generated single-cell and nucleus atlases from intact, dissociated, and injured muscle and liver and identified a common stress response signature shared by multiple cell types across these organs. This prevalent stress response was detected in published datasets across a range of tissues, demonstrating high conservation but also a significant degree of data distortion in single-cell reference atlases. Using quiescent muscle stem cells as a paradigm of cell activation following injury, we captured early cell activation following muscle injury and found that an essential ERK1/2 primary proliferation signal precedes initiation of the Notch-regulated myogenic program. This study defines initial events in response to tissue perturbation and identifies a broadly conserved transcriptional stress response that acts in parallel with cell-specific adaptive alterations.
Collapse
Affiliation(s)
- Léo Machado
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France
| | - Perla Geara
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France
| | - Jordi Camps
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium; Bayer AG, 13353 Berlin, Germany
| | | | | | - Jens Van Herck
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Hugo Varet
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 75015 Paris, France; Plate-forme Biomics - Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75015 Paris, France
| | - Rachel Legendre
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, 75015 Paris, France; Plate-forme Biomics - Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75015 Paris, France
| | - Jean-Michel Pawlotsky
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; Département de Virologie, Hôpital Henri Mondor, F-94010 Créteil, France
| | - Maurilio Sampaolesi
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium; Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Pascal Maire
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014 Paris, France
| | - Frederic Relaix
- Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France; EnvA, IMRB, 94700 Maisons-Alfort, France; EFS, IMRB, 94010 Creteil, France; AP-HP, Hopital Mondor, Service d'histologie, F-94010 Creteil, France.
| | | |
Collapse
|
190
|
Huang Z, Yin Z, Xu J, Fei Y, Heng BC, Jiang X, Chen W, Shen W. Tendon Stem/Progenitor Cell Subpopulations and Their Implications in Tendon Biology. Front Cell Dev Biol 2021; 9:631272. [PMID: 33681210 PMCID: PMC7930382 DOI: 10.3389/fcell.2021.631272] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/27/2021] [Indexed: 12/28/2022] Open
Abstract
Tendon harbors a cell population that possesses stem cell characteristics such as clonogenicity, multipotency and self-renewal capacity, commonly referred to as tendon stem/progenitor cells (TSPCs). Various techniques have been employed to study how TSPCs are implicated in tendon development, homeostasis and healing. Recent advances in single-cell analysis have enabled much progress in identifying and characterizing distinct subpopulations of TSPCs, which provides a more comprehensive view of TSPCs function in tendon biology. Understanding the mechanisms of physiological and pathological processes regulated by TSPCs, especially a particular subpopulation, would greatly benefit treatment of diseased tendons. Here, we summarize the current scientific literature on the various subpopulations of TSPCs, and discuss how TSPCs can contribute to tissue homeostasis and pathogenesis, as well as examine the key modulatory signaling pathways that determine stem/progenitor cell state. A better understanding of the roles that TSPCs play in tendon biology may facilitate the development of novel treatment strategies for tendon diseases.
Collapse
Affiliation(s)
- Zizhan Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China.,Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Zi Yin
- Institute of Sports Medicine, Zhejiang University, Hangzhou, China.,Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China.,China Orthopedic Regenerative Medicine (CORMed), Hangzhou, China
| | - Jialu Xu
- Department of Infectious Diseases, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yang Fei
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China.,Institute of Sports Medicine, Zhejiang University, Hangzhou, China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing, China
| | - Xuesheng Jiang
- Department of Orthopedic Surgery, Huzhou Hospital, Zhejiang University, Huzhou, China
| | - Weishan Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Weiliang Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute, Zhejiang University, Hangzhou, China.,Institute of Sports Medicine, Zhejiang University, Hangzhou, China.,Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China.,China Orthopedic Regenerative Medicine (CORMed), Hangzhou, China
| |
Collapse
|
191
|
Biressi S, Filareto A, Rando TA. Stem cell therapy for muscular dystrophies. J Clin Invest 2021; 130:5652-5664. [PMID: 32946430 DOI: 10.1172/jci142031] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Muscular dystrophies are a heterogeneous group of genetic diseases, characterized by progressive degeneration of skeletal and cardiac muscle. Despite the intense investigation of different therapeutic options, a definitive treatment has not been developed for this debilitating class of pathologies. Cell-based therapies in muscular dystrophies have been pursued experimentally for the last three decades. Several cell types with different characteristics and tissues of origin, including myogenic stem and progenitor cells, stromal cells, and pluripotent stem cells, have been investigated over the years and have recently entered in the clinical arena with mixed results. In this Review, we do a roundup of the past attempts and describe the updated status of cell-based therapies aimed at counteracting the skeletal and cardiac myopathy present in dystrophic patients. We present current challenges, summarize recent progress, and make recommendations for future research and clinical trials.
Collapse
Affiliation(s)
- Stefano Biressi
- Department of Cellular, Computational and Integrative Biology (CIBIO) and.,Dulbecco Telethon Institute, University of Trento, Povo, Italy
| | - Antonio Filareto
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Conneticut, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences and.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California, USA.,Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
192
|
Abstract
Quiescence is a cellular state in which a cell remains out of the cell cycle but retains the capacity to divide. The unique ability of adult stem cells to maintain quiescence is crucial for life-long tissue homeostasis and regenerative capacity. Quiescence has long been viewed as an inactive state but recent studies have shown that it is in fact an actively regulated process and that adult stem cells are highly reactive to extrinsic stimuli. This has fuelled hopes of boosting the reactivation potential of adult stem cells to improve tissue function during ageing. In this Review, we provide a perspective of the quiescent state and discuss how quiescent adult stem cells transition into the cell cycle. We also discuss current challenges in the field, highlighting recent technical advances that could help overcome some of these challenges.
Collapse
Affiliation(s)
- Noelia Urbán
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter Campus (VBC), Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, The Hong Kong University of Science and Technology Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China
| |
Collapse
|
193
|
Xu Z, You W, Chen W, Zhou Y, Nong Q, Valencak TG, Wang Y, Shan T. Single-cell RNA sequencing and lipidomics reveal cell and lipid dynamics of fat infiltration in skeletal muscle. J Cachexia Sarcopenia Muscle 2021; 12:109-129. [PMID: 33244879 PMCID: PMC7890272 DOI: 10.1002/jcsm.12643] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/23/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ageing is accompanied by sarcopenia and intramuscular fat (IMAT) infiltration. In skeletal muscle, fat infiltration is a common feature in several myopathies and is associated with muscular dysfunction and insulin resistance. However, the cellular origin and lipidomic and transcriptomic changes during fat infiltration in skeletal muscle remain unclear. METHODS In the current study, we generated a high IMAT-infiltrated skeletal muscle model by glycerol (GLY) injection. Single-cell RNA sequencing and lineage tracing were performed on GLY-injured skeletal muscle at 5 days post-injection (DPI) to identify the cell origins and dynamics. Lipidomics and RNA sequencing were performed on IMAT-infiltrated skeletal muscle at 14 DPI (or 17 DPI for the cold treatment) to analyse alterations of lipid compositions and gene expression levels. RESULTS We identified nine distinct major clusters including myeloid-derived cells (52.13%), fibroblast/fibro/adipogenic progenitors (FAPs) (23.24%), and skeletal muscle stem cells (2.02%) in GLY-injured skeletal muscle. Clustering and pseudotemporal trajectories revealed six subpopulations in fibroblast/FAPs and 10 subclusters in myeloid-derived cells. A subpopulation of myeloid-derived cells expressing adipocyte-enriched genes and Pdgfra- /Cd68+ cells displayed lipid droplets upon adipogenic induction, indicating their adipogenic potential. Lipidomic analysis revealed the changes of overall lipid classes composition (e.g. triglycerides (TAGs) increased by 19.3 times, P = 0.0098; sulfoquinovosyl diacylglycerol decreased by 83%, P = 0.0056) and in the distribution of lipids [e.g. TAGs (18:2/18:2/22:6) increased by 181.6 times, P = 0.021] between GLY-group and saline control. RNA-seq revealed 1847 up-regulated genes and 321 down-regulated genes and significant changes in lipid metabolism-related pathways (e.g. glycerolipid pathway and glycerophospholipid pathway) in our model of GLY-injured skeletal muscle. Notably, short-term cold exposure altered fatty acid composition (e.g. saturated fatty acid decreased by 6.4%, P = 0.058) in fat-infiltrated muscles through directly affecting lipid metabolism pathways including PI3K-AKT and MAPK signalling pathway. CONCLUSIONS Our results showed that a subpopulation of myeloid-derived cells may contribute to IMAT infiltration. GLY-induced IMAT infiltration changed the lipid composition and gene expression profiles. Short-term cold exposure might regulate lipid metabolism and its related signalling pathways in fat-infiltrated muscle. Our study provides a comprehensive resource describing the molecular signature of fat infiltration in skeletal muscle.
Collapse
Affiliation(s)
- Ziye Xu
- College of Animal SciencesZhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Laboratory of Animal Feed and Nutrition of Zhejiang ProvinceHangzhouChina
| | - Wenjing You
- College of Animal SciencesZhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Laboratory of Animal Feed and Nutrition of Zhejiang ProvinceHangzhouChina
| | - Wentao Chen
- College of Animal SciencesZhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Laboratory of Animal Feed and Nutrition of Zhejiang ProvinceHangzhouChina
| | - Yanbing Zhou
- College of Animal SciencesZhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Laboratory of Animal Feed and Nutrition of Zhejiang ProvinceHangzhouChina
| | - Qiuyun Nong
- College of Animal SciencesZhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Laboratory of Animal Feed and Nutrition of Zhejiang ProvinceHangzhouChina
| | | | - Yizhen Wang
- College of Animal SciencesZhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Laboratory of Animal Feed and Nutrition of Zhejiang ProvinceHangzhouChina
| | - Tizhong Shan
- College of Animal SciencesZhejiang UniversityHangzhouZhejiangChina
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University)Ministry of EducationHangzhouChina
- Laboratory of Animal Feed and Nutrition of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
194
|
Relaix F, Bencze M, Borok MJ, Der Vartanian A, Gattazzo F, Mademtzoglou D, Perez-Diaz S, Prola A, Reyes-Fernandez PC, Rotini A, Taglietti. Perspectives on skeletal muscle stem cells. Nat Commun 2021; 12:692. [PMID: 33514709 PMCID: PMC7846784 DOI: 10.1038/s41467-020-20760-6] [Citation(s) in RCA: 198] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 12/17/2020] [Indexed: 01/30/2023] Open
Abstract
Skeletal muscle has remarkable regeneration capabilities, mainly due to its resident muscle stem cells (MuSCs). In this review, we introduce recently developed technologies and the mechanistic insights they provide to the understanding of MuSC biology, including the re-definition of quiescence and Galert states. Additionally, we present recent studies that link MuSC function with cellular heterogeneity, highlighting the complex regulation of self-renewal in regeneration, muscle disorders and aging. Finally, we discuss MuSC metabolism and its role, as well as the multifaceted regulation of MuSCs by their niche. The presented conceptual advances in the MuSC field impact on our general understanding of stem cells and their therapeutic use in regenerative medicine.
Collapse
Affiliation(s)
- F. Relaix
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France ,EnvA, IMRB, 94700 Maisons-Alfort, France ,grid.462410.50000 0004 0386 3258EFS, IMRB, 94010 Creteil, France ,grid.50550.350000 0001 2175 4109AP-HP, Hopital Mondor, Service d’histologie, 94010 Creteil, France
| | - M. Bencze
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France
| | - M. J. Borok
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France
| | - A. Der Vartanian
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France
| | - F. Gattazzo
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France ,grid.462410.50000 0004 0386 3258EFS, IMRB, 94010 Creteil, France
| | - D. Mademtzoglou
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France
| | - S. Perez-Diaz
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France
| | - A. Prola
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France ,EnvA, IMRB, 94700 Maisons-Alfort, France
| | - P. C. Reyes-Fernandez
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France
| | - A. Rotini
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France
| | - Taglietti
- grid.462410.50000 0004 0386 3258Univ Paris Est Creteil, INSERM, IMRB, 94010 Creteil, France
| |
Collapse
|
195
|
Feige P, Tsai EC, Rudnicki MA. Analysis of human satellite cell dynamics on cultured adult skeletal muscle myofibers. Skelet Muscle 2021; 11:1. [PMID: 33397479 PMCID: PMC7780694 DOI: 10.1186/s13395-020-00256-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/06/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Maintaining stem cells in physiologically relevant states is necessary to understand cell and context-specific signalling paradigms and to understand complex interfaces between cells in situ. Understanding human stem cell function is largely based on tissue biopsies, cell culture, and transplantation into model organisms. METHODS Here, we describe a method to isolate post-mortem intact human muscle myofibers and culture muscle stem cells within the niche microenvironment to assay cellular dynamics, stem cell identity, stem cell hierarchy, and differentiation potential. RESULTS We show human myofiber culture maintains complex cell-cell contacts and extracellular niche composition during culture. Human satellite cells can be cultured at least 8 days, which represents a timepoint of activation, differentiation, and de novo human myofiber formation. We demonstrate that adult human muscle stem cells undergo apicobasal and planar cell divisions and express polarized dystrophin and EGFR. Furthermore, we validate that stimulation of the EGFR pathway stimulates the generation of myogenic progenitors and myogenic differentiation. CONCLUSIONS This method provides proof of principle evidence for the use of human muscle to evaluate satellite cell dynamics and has applications in pre-clinical evaluation of therapeutics targeting muscle repair.
Collapse
Affiliation(s)
- Peter Feige
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Eve C Tsai
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Surgery, Division of Neurosurgery, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
196
|
Abstract
The resident stem cell for skeletal muscle is the satellite cell. On the 50th anniversary of its discovery in 1961, we described the history of skeletal muscle research and the seminal findings made during the first 20 years in the life of the satellite cell (Scharner and Zammit 2011, doi: 10.1186/2044-5040-1-28). These studies established the satellite cell as the source of myoblasts for growth and regeneration of skeletal muscle. Now on the 60th anniversary, we highlight breakthroughs in the second phase of satellite cell research from 1980 to 2000. These include technical innovations such as isolation of primary satellite cells and viable muscle fibres complete with satellite cells in their niche, together with generation of many useful reagents including genetically modified organisms and antibodies still in use today. New methodologies were combined with description of endogenous satellite cells markers, notably Pax7. Discovery of the muscle regulatory factors Myf5, MyoD, myogenin, and MRF4 in the late 1980s revolutionized understanding of the control of both developmental and regerenative myogenesis. Emergence of genetic lineage markers facilitated identification of satellite cells in situ, and also empowered transplantation studies to examine satellite cell function. Finally, satellite cell heterogeneity and the supportive role of non-satellite cell types in muscle regeneration were described. These major advances in methodology and in understanding satellite cell biology provided further foundations for the dramatic escalation of work on muscle stem cells in the 21st century.
Collapse
Affiliation(s)
- Elise N Engquist
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, SE1 1UL, UK
| |
Collapse
|
197
|
Wisdom AJ, Mowery YM, Hong CS, Himes JE, Nabet BY, Qin X, Zhang D, Chen L, Fradin H, Patel R, Bassil AM, Muise ES, King DA, Xu ES, Carpenter DJ, Kent CL, Smythe KS, Williams NT, Luo L, Ma Y, Alizadeh AA, Owzar K, Diehn M, Bradley T, Kirsch DG. Single cell analysis reveals distinct immune landscapes in transplant and primary sarcomas that determine response or resistance to immunotherapy. Nat Commun 2020; 11:6410. [PMID: 33335088 PMCID: PMC7746723 DOI: 10.1038/s41467-020-19917-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy fails to cure most cancer patients. Preclinical studies indicate that radiotherapy synergizes with immunotherapy, promoting radiation-induced antitumor immunity. Most preclinical immunotherapy studies utilize transplant tumor models, which overestimate patient responses. Here, we show that transplant sarcomas are cured by PD-1 blockade and radiotherapy, but identical treatment fails in autochthonous sarcomas, which demonstrate immunoediting, decreased neoantigen expression, and tumor-specific immune tolerance. We characterize tumor-infiltrating immune cells from transplant and primary tumors, revealing striking differences in their immune landscapes. Although radiotherapy remodels myeloid cells in both models, only transplant tumors are enriched for activated CD8+ T cells. The immune microenvironment of primary murine sarcomas resembles most human sarcomas, while transplant sarcomas resemble the most inflamed human sarcomas. These results identify distinct microenvironments in murine sarcomas that coevolve with the immune system and suggest that patients with a sarcoma immune phenotype similar to transplant tumors may benefit most from PD-1 blockade and radiotherapy.
Collapse
Affiliation(s)
- Amy J Wisdom
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Yvonne M Mowery
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA.
- Duke Cancer Institute, Durham, NC, 27708, USA.
| | - Cierra S Hong
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Jonathon E Himes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Barzin Y Nabet
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Department of Oncology Biomarker Development, Genentech, South San Francisco, CA, 94080, USA
| | - Xiaodi Qin
- Duke Cancer Institute, Durham, NC, 27708, USA
| | | | - Lan Chen
- Merck & Co., Inc, Kenilworth, NJ, 07033, USA
| | - Hélène Fradin
- Duke Center for Genomic and Computational Biology, Durham, NC, 27708, USA
| | - Rutulkumar Patel
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Alex M Bassil
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | | | - Daniel A King
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Eric S Xu
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - David J Carpenter
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Collin L Kent
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | | | - Nerissa T Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Kouros Owzar
- Duke Cancer Institute, Durham, NC, 27708, USA
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, 27710, USA
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA, 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Radiation Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Todd Bradley
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, Kansas City, MO, 64108, USA
| | - David G Kirsch
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27708, USA.
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27708, USA.
- Duke Cancer Institute, Durham, NC, 27708, USA.
| |
Collapse
|
198
|
Kim M, Franke V, Brandt B, Lowenstein ED, Schöwel V, Spuler S, Akalin A, Birchmeier C. Single-nucleus transcriptomics reveals functional compartmentalization in syncytial skeletal muscle cells. Nat Commun 2020; 11:6375. [PMID: 33311457 PMCID: PMC7732842 DOI: 10.1038/s41467-020-20064-9] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/09/2020] [Indexed: 11/25/2022] Open
Abstract
Syncytial skeletal muscle cells contain hundreds of nuclei in a shared cytoplasm. We investigated nuclear heterogeneity and transcriptional dynamics in the uninjured and regenerating muscle using single-nucleus RNA-sequencing (snRNAseq) of isolated nuclei from muscle fibers. This revealed distinct nuclear subtypes unrelated to fiber type diversity, previously unknown subtypes as well as the expected ones at the neuromuscular and myotendinous junctions. In fibers of the Mdx dystrophy mouse model, distinct subtypes emerged, among them nuclei expressing a repair signature that were also abundant in the muscle of dystrophy patients, and a nuclear population associated with necrotic fibers. Finally, modifications of our approach revealed the compartmentalization in the rare and specialized muscle spindle. Our data identifies nuclear compartments of the myofiber and defines a molecular roadmap for their functional analyses; the data can be freely explored on the MyoExplorer server ( https://shiny.mdc-berlin.de/MyoExplorer/ ).
Collapse
Affiliation(s)
- Minchul Kim
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Vedran Franke
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Bettina Brandt
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Elijah D Lowenstein
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Verena Schöwel
- Muscle Research Unit, Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Altuna Akalin
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center for Molecular Medicine, Berlin, Germany.
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
199
|
Petrany MJ, Swoboda CO, Sun C, Chetal K, Chen X, Weirauch MT, Salomonis N, Millay DP. Single-nucleus RNA-seq identifies transcriptional heterogeneity in multinucleated skeletal myofibers. Nat Commun 2020; 11:6374. [PMID: 33311464 PMCID: PMC7733460 DOI: 10.1038/s41467-020-20063-w] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
While the majority of cells contain a single nucleus, cell types such as trophoblasts, osteoclasts, and skeletal myofibers require multinucleation. One advantage of multinucleation can be the assignment of distinct functions to different nuclei, but comprehensive interrogation of transcriptional heterogeneity within multinucleated tissues has been challenging due to the presence of a shared cytoplasm. Here, we utilized single-nucleus RNA-sequencing (snRNA-seq) to determine the extent of transcriptional diversity within multinucleated skeletal myofibers. Nuclei from mouse skeletal muscle were profiled across the lifespan, which revealed the presence of distinct myonuclear populations emerging in postnatal development as well as aging muscle. Our datasets also provided a platform for discovery of genes associated with rare specialized regions of the muscle cell, including markers of the myotendinous junction and functionally validated factors expressed at the neuromuscular junction. These findings reveal that myonuclei within syncytial muscle fibers possess distinct transcriptional profiles that regulate muscle biology.
Collapse
Affiliation(s)
- Michael J Petrany
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Casey O Swoboda
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
200
|
Leung C, Murad KBA, Tan ALT, Yada S, Sagiraju S, Bode PK, Barker N. Lgr5 Marks Adult Progenitor Cells Contributing to Skeletal Muscle Regeneration and Sarcoma Formation. Cell Rep 2020; 33:108535. [PMID: 33357435 DOI: 10.1016/j.celrep.2020.108535] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/15/2020] [Accepted: 11/25/2020] [Indexed: 12/21/2022] Open
Abstract
Regeneration of adult skeletal muscle is driven largely by resident satellite cells, a stem cell population increasingly considered to display a high degree of molecular heterogeneity. In this study, we find that Lgr5, a receptor for Rspo and a potent mediator of Wnt/β-catenin signaling, marks a subset of activated satellite cells that contribute to muscle regeneration. Lgr5 is found to be rapidly upregulated in purified myogenic progenitors following acute cardiotoxin-induced injury. In vivo lineage tracing using our Lgr5-2ACreERT2R26tdTomatoLSL reporter mouse model shows that Lgr5+ cells can reconstitute damaged muscle fibers following muscle injury, as well as replenish the quiescent satellite cell pool. Moreover, conditional mutation in Lgr52ACreERT2;KrasG12D;Trp53flox/flox mice drives undifferentiated pleomorphic sarcoma formation in adult mice, thereby substantiating Lgr5+ cells as a cell of origin of sarcomas. Our findings provide the groundwork for developing Rspo/Wnt-signaling-based therapeutics to potentially enhance regenerative outcomes of skeletal muscles in degenerative muscle diseases.
Collapse
Affiliation(s)
- Carly Leung
- A(∗)STAR Institute of Medical Biology, Singapore 138648, Singapore; A(∗)STAR Institute of Molecular and Cellular Biology, Singapore 138648, Singapore
| | - Katzrin Bte Ahmad Murad
- A(∗)STAR Institute of Medical Biology, Singapore 138648, Singapore; A(∗)STAR Institute of Molecular and Cellular Biology, Singapore 138648, Singapore
| | - Adelyn Liang Thing Tan
- A(∗)STAR Institute of Medical Biology, Singapore 138648, Singapore; A(∗)STAR Institute of Molecular and Cellular Biology, Singapore 138648, Singapore
| | - Swathi Yada
- A(∗)STAR Institute of Medical Biology, Singapore 138648, Singapore; A(∗)STAR Institute of Molecular and Cellular Biology, Singapore 138648, Singapore
| | - Sowmya Sagiraju
- A(∗)STAR Institute of Medical Biology, Singapore 138648, Singapore; A(∗)STAR Institute of Molecular and Cellular Biology, Singapore 138648, Singapore
| | - Peter Karl Bode
- Department of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Nick Barker
- A(∗)STAR Institute of Medical Biology, Singapore 138648, Singapore; A(∗)STAR Institute of Molecular and Cellular Biology, Singapore 138648, Singapore; Cancer Research Institute, Kanazawa University, Kakuma-machi Kanazawa 920-1192, Japan; School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore.
| |
Collapse
|