201
|
CPEB1 directs muscle stem cell activation by reprogramming the translational landscape. Nat Commun 2022; 13:947. [PMID: 35177647 PMCID: PMC8854658 DOI: 10.1038/s41467-022-28612-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/14/2022] [Indexed: 11/08/2022] Open
Abstract
Skeletal muscle stem cells, also called Satellite Cells (SCs), are actively maintained in quiescence but can activate quickly upon extrinsic stimuli. However, the mechanisms of how quiescent SCs (QSCs) activate swiftly remain elusive. Here, using a whole mouse perfusion fixation approach to obtain bona fide QSCs, we identify massive proteomic changes during the quiescence-to-activation transition in pathways such as chromatin maintenance, metabolism, transcription, and translation. Discordant correlation of transcriptomic and proteomic changes reveals potential translational regulation upon SC activation. Importantly, we show Cytoplasmic Polyadenylation Element Binding protein 1 (CPEB1), post-transcriptionally affects protein translation during SC activation by binding to the 3' UTRs of different transcripts. We demonstrate phosphorylation-dependent CPEB1 promoted Myod1 protein synthesis by binding to the cytoplasmic polyadenylation elements (CPEs) within its 3' UTRs to regulate SC activation and muscle regeneration. Our study characterizes CPEB1 as a key regulator to reprogram the translational landscape directing SC activation and subsequent proliferation.
Collapse
|
202
|
Dominici C, Richard S. Muscle stem cell polarity requires QKI-mediated alternative splicing of Integrin Alpha-7 (Itga7). Life Sci Alliance 2022; 5:5/5/e202101192. [PMID: 35165120 PMCID: PMC8860092 DOI: 10.26508/lsa.202101192] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/30/2022] Open
Abstract
The RNA-binding protein Quaking (QKI) is a post-transcriptional regulator of genes encoding polarity proteins in muscle stem cells. Loss of QKI in MuSCs results in reduced myogenic progenitors and a striking muscle regeneration defect. Muscle stem cells (MuSCs) have the ability to carry out the specialized function of cell polarization, which is required for the production of one repopulating cell and one myogenic progenitor cell with muscle regeneration capabilities. The mechanisms which regulate proteins involved in establishing MuSC polarity such as Dmd and Itga7 are currently not well understood. Herein, we define the RNA-binding protein Quaking (QKI) as a major regulator alternative splicing of key MuSC polarity factors including Dmd, Itga7, Mark2, and Numb. We generate a conditional QKI knockout mouse, and for the first time it is shown in vivo that deficiency of QKI in MuSCs results in reduced asymmetric cell divisions, leading to a loss of the myogenic progenitor cell population and striking muscle regeneration defects. Transcriptomic analysis of QKI-deficient MuSCs identifies QKI as a regulator of the splicing events which give rise to the mutually exclusive Itga7-X1 and -X2 isoforms. We observe increased X1 expression in QKI-deficient MuSCs and recapitulate this splicing event using antisense oligonucleotide directed against a quaking binding site within the Itga7 mRNA. Interestingly, recreating this single splicing event is detrimental to the polarization of Itga7 and Dmd proteins, and leads to a drastic reduction of the myogenic progenitor population, highlighting the significance of QKI-mediated alternative splicing of Itga7 in maintaining MuSC polarity. Altogether, these findings define a novel role for QKI as a post-transcriptional regulator of MuSC polarity.
Collapse
Affiliation(s)
- Claudia Dominici
- Segal Cancer Center, Lady Davis Institute for Medical Research and Gerald Bronfman Department of Oncology and Departments of Medicine, Human Genetics and Biochemistry, McGill University, Montréal, Québec, Canada
| | - Stéphane Richard
- Segal Cancer Center, Lady Davis Institute for Medical Research and Gerald Bronfman Department of Oncology and Departments of Medicine, Human Genetics and Biochemistry, McGill University, Montréal, Québec, Canada
| |
Collapse
|
203
|
Gugliuzza MV, Crist C. Muscle stem cell adaptations to cellular and environmental stress. Skelet Muscle 2022; 12:5. [PMID: 35151369 PMCID: PMC8840228 DOI: 10.1186/s13395-022-00289-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/30/2022] [Indexed: 12/21/2022] Open
Abstract
Background Lifelong regeneration of the skeletal muscle is dependent on a rare population of resident skeletal muscle stem cells, also named ‘satellite cells’ for their anatomical position on the outside of the myofibre and underneath the basal lamina. Muscle stem cells maintain prolonged quiescence, but activate the myogenic programme and the cell cycle in response to injury to expand a population of myogenic progenitors required to regenerate muscle. The skeletal muscle does not regenerate in the absence of muscle stem cells. Main body The notion that lifelong regeneration of the muscle is dependent on a rare, non-redundant population of stem cells seems contradictory to accumulating evidence that muscle stem cells have activated multiple stress response pathways. For example, muscle stem cell quiescence is mediated in part by the eIF2α arm of the integrated stress response and by negative regulators of mTORC1, two translational control pathways that downregulate protein synthesis in response to stress. Muscle stem cells also activate pathways to protect against DNA damage, heat shock, and environmental stress. Here, we review accumulating evidence that muscle stem cells encounter stress during their prolonged quiescence and their activation. While stress response pathways are classically described to be bimodal whereby a threshold dictates cell survival versus cell death responses to stress, we review evidence that muscle stem cells additionally respond to stress by spontaneous activation and fusion to myofibres. Conclusion We propose a cellular stress test model whereby the prolonged state of quiescence and the microenvironment serve as selective pressures to maintain muscle stem cell fitness, to safeguard the lifelong regeneration of the muscle. Fit muscle stem cells that maintain robust stress responses are permitted to maintain the muscle stem cell pool. Unfit muscle stem cells are depleted from the pool first by spontaneous activation, or in the case of severe stress, by activating cell death or senescence pathways.
Collapse
|
204
|
Soulez M, Tanguay PL, Dô F, Dort J, Crist C, Kotlyarov A, Gaestel M, Ferron M, Dumont NA, Meloche S. ERK3-MK5 signaling regulates myogenic differentiation and muscle regeneration by promoting FoxO3 degradation. J Cell Physiol 2022; 237:2271-2287. [PMID: 35141958 DOI: 10.1002/jcp.30695] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 12/07/2021] [Accepted: 01/24/2022] [Indexed: 12/25/2022]
Abstract
The physiological functions and downstream effectors of the atypical mitogen-activated protein kinase extracellular signal-regulated kinase 3 (ERK3) remain to be characterized. We recently reported that mice expressing catalytically-inactive ERK3 (Mapk6KD/KD ) exhibit a reduced postnatal growth rate as compared to control mice. Here, we show that genetic inactivation of ERK3 impairs postnatal skeletal muscle growth and adult muscle regeneration after injury. Loss of MAPK-activated protein kinase 5 (MK5) phenocopies the muscle phenotypes of Mapk6KD/KD mice. At the cellular level, genetic or pharmacological inactivation of ERK3 or MK5 induces precocious differentiation of C2C12 or primary myoblasts, concomitant with MyoD activation. Reciprocally, ectopic expression of activated MK5 inhibits myogenic differentiation. Mechanistically, we show that MK5 directly phosphorylates FoxO3, promoting its degradation and reducing its association with MyoD. Depletion of FoxO3 rescues in part the premature differentiation of C2C12 myoblasts observed upon inactivation of ERK3 or MK5. Our findings reveal that ERK3 and its substrate MK5 act in a linear signaling pathway to control postnatal myogenic differentiation.
Collapse
Affiliation(s)
- Mathilde Soulez
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Pierre-Luc Tanguay
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada.,Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Ipsen Biopharmaceuticals Canada, Mississauga, Ontario, Canada
| | - Florence Dô
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Junio Dort
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,School of Rehabilitation, Université de Montréal, Montreal, Quebec, Canada
| | - Colin Crist
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Alexey Kotlyarov
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Mathieu Ferron
- Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Department of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Nicolas A Dumont
- CHU Sainte-Justine Research Center, Montreal, Quebec, Canada.,School of Rehabilitation, Université de Montréal, Montreal, Quebec, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada.,Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
205
|
A Long Journey before Cycling: Regulation of Quiescence Exit in Adult Muscle Satellite Cells. Int J Mol Sci 2022; 23:ijms23031748. [PMID: 35163665 PMCID: PMC8836154 DOI: 10.3390/ijms23031748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
Skeletal muscle harbors a pool of stem cells called muscle satellite cells (MuSCs) that are mainly responsible for its robust regenerative capacities. Adult satellite cells are mitotically quiescent in uninjured muscles under homeostasis, but they exit quiescence upon injury to re-enter the cell cycle to proliferate. While most of the expanded satellites cells differentiate and fuse to form new myofibers, some undergo self-renewal to replenish the stem cell pool. Specifically, quiescence exit describes the initial transition of MuSCs from quiescence to the first cell cycle, which takes much longer than the time required for subsequent cell cycles and involves drastic changes in cell size, epigenetic and transcriptomic profiles, and metabolic status. It is, therefore, an essential period indispensable for the success of muscle regeneration. Diverse mechanisms exist in MuSCs to regulate quiescence exit. In this review, we summarize key events that occur during quiescence exit in MuSCs and discuss the molecular regulation of this process with an emphasis on multiple levels of intrinsic regulatory mechanisms. A comprehensive understanding of how quiescence exit is regulated will facilitate satellite cell-based muscle regenerative therapies and advance their applications in various disease and aging conditions.
Collapse
|
206
|
Zhu P, Hamlish NX, Thakkar AV, Steffeck AWT, Rendleman EJ, Khan NH, Waldeck NJ, DeVilbiss AW, Martin-Sandoval MS, Mathews TP, Chandel NS, Peek CB. BMAL1 drives muscle repair through control of hypoxic NAD + regeneration in satellite cells. Genes Dev 2022; 36:149-166. [PMID: 35115380 PMCID: PMC8887128 DOI: 10.1101/gad.349066.121] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 01/05/2022] [Indexed: 01/07/2023]
Abstract
The process of tissue regeneration occurs in a developmentally timed manner, yet the role of circadian timing is not understood. Here, we identify a role for the adult muscle stem cell (MuSC)-autonomous clock in the control of muscle regeneration following acute ischemic injury. We observed greater muscle repair capacity following injury during the active/wake period as compared with the inactive/rest period in mice, and loss of Bmal1 within MuSCs leads to impaired muscle regeneration. We demonstrate that Bmal1 loss in MuSCs leads to reduced activated MuSC number at day 3 postinjury, indicating a failure to properly expand the myogenic precursor pool. In cultured primary myoblasts, we observed that loss of Bmal1 impairs cell proliferation in hypoxia (a condition that occurs in the first 1-3 d following tissue injury in vivo), as well as subsequent myofiber differentiation. Loss of Bmal1 in both cultured myoblasts and in vivo activated MuSCs leads to reduced glycolysis and premature activation of prodifferentiation gene transcription and epigenetic remodeling. Finally, hypoxic cell proliferation and myofiber formation in Bmal1-deficient myoblasts are restored by increasing cytosolic NAD+ Together, we identify the MuSC clock as a pivotal regulator of oxygen-dependent myoblast cell fate and muscle repair through the control of the NAD+-driven response to injury.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Noah X Hamlish
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Abhishek Vijay Thakkar
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Adam W T Steffeck
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Emily J Rendleman
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Nabiha H Khan
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Nathan J Waldeck
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Andrew W DeVilbiss
- Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Misty S Martin-Sandoval
- Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Thomas P Mathews
- Children's Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | - Navdeep S Chandel
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Clara B Peek
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
207
|
Takada N, Takasugi M, Nonaka Y, Kamiya T, Takemura K, Satoh J, Ito S, Fujimoto K, Uematsu S, Yoshida K, Morita T, Nakamura H, Uezumi A, Ohtani N. Galectin-3 promotes the adipogenic differentiation of PDGFRα+ cells and ectopic fat formation in regenerating muscle. Development 2022; 149:274217. [DOI: 10.1242/dev.199443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 12/16/2021] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Worldwide prevalence of obesity is associated with the increase of lifestyle-related diseases. The accumulation of intermuscular adipose tissue (IMAT) is considered a major problem whereby obesity leads to sarcopenia and metabolic disorders and thus is a promising target for treating these pathological conditions. However, whereas obesity-associated IMAT is suggested to originate from PDGFRα+ mesenchymal progenitors, the processes underlying this adipogenesis remain largely unexplored. Here, we comprehensively investigated intra- and extracellular changes associated with these processes using single-cell RNA sequencing and mass spectrometry. Our single-cell RNA sequencing analysis identified a small PDGFRα+ cell population in obese mice directed strongly toward adipogenesis. Proteomic analysis showed that the appearance of this cell population is accompanied by an increase in galectin-3 in interstitial environments, which was found to activate adipogenic PPARγ signals in PDGFRα+ cells. Moreover, IMAT formation during muscle regeneration was significantly suppressed in galectin-3 knockout mice. Our findings, together with these multi-omics datasets, could unravel microenvironmental networks during muscle regeneration highlighting possible therapeutic targets against IMAT formation in obesity.
Collapse
Affiliation(s)
- Naoki Takada
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
| | - Masaki Takasugi
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
| | - Yoshiki Nonaka
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
| | - Tomonori Kamiya
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
| | - Kazuaki Takemura
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
| | - Junko Satoh
- Division for Mass Spectrometry, Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Shinji Ito
- Division for Mass Spectrometry, Medical Research Support Center, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Kosuke Fujimoto
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
- Division of Metagenome Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
- Division of Metagenome Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Kayo Yoshida
- Department of Laboratory Animal Science, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
- Facility of Laboratory Animals, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
| | - Takashi Morita
- Facility of Laboratory Animals, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
| | - Hiroaki Nakamura
- Department of Orthopedic Surgery, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
| | - Akiyoshi Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, 173-0015, Japan
| | - Naoko Ohtani
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Osaka, 545-8585, Japan
- AMED-CREST, AMED, Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| |
Collapse
|
208
|
Philips C, Terrie L, Thorrez L. Decellularized skeletal muscle: A versatile biomaterial in tissue engineering and regenerative medicine. Biomaterials 2022; 283:121436. [DOI: 10.1016/j.biomaterials.2022.121436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 01/27/2022] [Accepted: 02/17/2022] [Indexed: 12/31/2022]
|
209
|
Agarwal M, Bharadwaj A, Mathew SJ. TLE4 regulates muscle stem cell quiescence and skeletal muscle differentiation. J Cell Sci 2022; 135:274455. [PMID: 35099008 DOI: 10.1242/jcs.256008] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/18/2022] [Indexed: 10/19/2022] Open
Abstract
Muscle stem (satellite) cells express Pax7, a key transcription factor essential for satellite cell maintenance and adult muscle regeneration. We identify the corepressor Transducin-Like Enhancer of Split-4 (TLE4) as a Pax7 interaction partner expressed in quiescent satellite cells under homeostasis. A subset of satellite cells transiently downregulate TLE4 during early time points following injury. We identify these to be activated satellite cells where TLE4 downregulation is required for Myf5 activation and myogenic commitment. Our results indicate that TLE4 represses Pax7-mediated Myf5 transcriptional activation by occupying the -111 kb Myf5 enhancer to maintain quiescence. Loss of TLE4 function causes Myf5 upregulation, increase in satellite cell numbers, and altered differentiation dynamics during regeneration. Thus, we have uncovered a novel mechanism to maintain satellite cell quiescence and regulating muscle differentiation mediated by the corepressor TLE4.
Collapse
Affiliation(s)
- Megha Agarwal
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, Haryana, India.,Manipal University, Manipal, Karnataka, 576104, India
| | - Anushree Bharadwaj
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, Haryana, India
| | - Sam J Mathew
- Developmental Genetics Laboratory, Regional Centre for Biotechnology (RCB), NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, Haryana, India.,Manipal University, Manipal, Karnataka, 576104, India
| |
Collapse
|
210
|
Wang X, Shelton SD, Bordieanu B, Frank AR, Yi Y, Venigalla SSK, Gu Z, Lenser NP, Glogauer M, Chandel NS, Zhao H, Zhao Z, McFadden DG, Mishra P. Scinderin promotes fusion of electron transport chain dysfunctional muscle stem cells with myofibers. NATURE AGING 2022; 2:155-169. [PMID: 35342888 PMCID: PMC8954567 DOI: 10.1038/s43587-021-00164-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
Muscle stem cells (MuSCs) experience age-associated declines in number and function, accompanied by mitochondrial electron transport chain (ETC) dysfunction and increased reactive oxygen species (ROS). The source of these changes, and how MuSCs respond to mitochondrial dysfunction, is unknown. We report here that in response to mitochondrial ROS, murine MuSCs directly fuse with neighboring myofibers; this phenomenon removes ETC-dysfunctional MuSCs from the stem cell compartment. MuSC-myofiber fusion is dependent on the induction of Scinderin, which promotes formation of actin-dependent protrusions required for membrane fusion. During aging, we find that the declining MuSC population accumulates mutations in the mitochondrial genome, but selects against dysfunctional variants. In the absence of clearance by Scinderin, the decline in MuSC numbers during aging is repressed; however, ETC-dysfunctional MuSCs are retained and can regenerate dysfunctional myofibers. We propose a model in which ETC-dysfunctional MuSCs are removed from the stem cell compartment by fusing with differentiated tissue.
Collapse
Affiliation(s)
- Xun Wang
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Spencer D Shelton
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bogdan Bordieanu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Present Address: Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Anderson R Frank
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Internal Medicine, Division of Endocrinology, Program in Molecular Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Yating Yi
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
- Present address: State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041 China
| | - Siva Sai Krishna Venigalla
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhimin Gu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Nicholas P Lenser
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Present address: Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Navdeep S Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Department of Biochemistry & Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hu Zhao
- Department of Comprehensive Dentistry, College of Dentistry, Texas A&M University, Dallas, TX 75246, USA
- Present address: The Chinese Institute for Brain Research, Beijing, China
| | - Zhiyu Zhao
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David G McFadden
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Internal Medicine, Division of Endocrinology, Program in Molecular Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Prashant Mishra
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
211
|
Bianconi V, Mozzetta C. Epigenetic control of muscle stem cells: time for a new dimension. Trends Genet 2022; 38:501-513. [DOI: 10.1016/j.tig.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022]
|
212
|
Ritso M, Tung LW, Rossi FMV. Emerging skeletal muscle stromal cell diversity: Functional divergence in fibro/adipogenic progenitor and mural cell populations. Exp Cell Res 2022; 410:112947. [PMID: 34822813 DOI: 10.1016/j.yexcr.2021.112947] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 12/27/2022]
Abstract
While the majority of healthy skeletal muscle consists of multinucleated syncytial repetitive contractile myofibers, repaired by skeletal muscle stem cells when damaged, the maintenance of muscle function also requires a range of tissue-resident stromal populations. In fact, the careful orchestration of damage response processes upon muscle injury relies heavily on stromal cell contribution for effective repair. The two main types of muscle-resident stromal cells are fibro/adipogenic progenitors and mural cells. The latter is comprised of pericytes and vascular smooth muscle cells. Recent publications identifying common markers for stromal cell populations have allowed investigating population dynamics throughout the regenerative process at a higher resolution. Mounting evidence now suggests that subpopulations with distinct roles may exist among stromal cells. In various degenerative muscle wasting conditions, critical cross-talk and spatial signalling amongst various cell populations become dysregulated. This can result in the failure to curb pathological fibro/adipogenic progenitor proliferation and propensity for laying down excessive extracellular matrix, which in turn leads to fibrotic infiltration, reduced contractile units and gradual decline in muscle function. Restoration of physiologically appropriate stromal cell function is therefore just as crucial for therapeutic targeting as the homeostatic maintenance of muscle function.
Collapse
Affiliation(s)
- Morten Ritso
- School of Biomedical Engineering, 2222 Health Sciences Mall, The University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, 2222 Health Sciences Mall, The University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Fabio M V Rossi
- School of Biomedical Engineering, 2222 Health Sciences Mall, The University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
213
|
Jacques E, Kuang Y, Kann AP, Le Grand F, Krauss RS, Gilbert PM. The mini-IDLE 3D biomimetic culture assay enables interrogation of mechanisms governing muscle stem cell quiescence and niche repopulation. eLife 2022; 11:81738. [PMID: 36537758 PMCID: PMC9904761 DOI: 10.7554/elife.81738] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Adult skeletal muscle harbours a population of muscle stem cells (MuSCs) that are required for repair after tissue injury. In youth, MuSCs return to a reversible state of cell-cycle arrest termed 'quiescence' after injury resolution. Conversely, some MuSCs in aged muscle remain semi-activated, causing a premature response to injuries that results in incomplete repair and eventual stem cell depletion. Regulating this balance between MuSC quiescence and activation may hold the key to restoring tissue homeostasis with age, but is incompletely understood. To fill this gap, we developed a simple and tractable in vitro method, to rapidly inactivate MuSCs freshly isolated from young murine skeletal muscle, and return them to a quiescent-like state for at least 1-week, which we name mini-IDLE (Inactivation and Dormancy LEveraged in vitro). This was achieved by introducing MuSCs into a 3D bioartificial niche comprised of a thin sheet of mouse myotubes, which we demonstrate provides the minimal cues necessary to induce quiescence. With different starting numbers of MuSCs, the assay revealed cellular heterogeneity and population-level adaptations that converged on a common niche repopulation density; behaviours previously observed only in vivo. Quiescence-associated hallmarks included a Pax7+CalcR+DDX6+MyoD-c-FOS- signature, quiescent-like morphologies, and polarized niche markers. Leveraging high-content bioimaging pipelines, we demonstrate a relationship between morphology and cell fate signatures for possible real-time morphology-based screening. When using MuSCs from aged muscle, they displayed aberrant proliferative activities and delayed inactivation kinetics, among other quiescence-associated defects that we show are partially rescued by wortmannin treatment. Thus, the assay offers an unprecedented opportunity to systematically investigate long-standing queries in areas such as regulation of pool size and functional heterogeneity within the MuSC population, and to uncover quiescence regulators in youth and age.
Collapse
Affiliation(s)
- Erik Jacques
- Institute of Biomedical Engineering, University of TorontoTorontoCanada,Donnelly Centre, University of TorontoTorontoCanada
| | - Yinni Kuang
- Donnelly Centre, University of TorontoTorontoCanada,Department of Cell and Systems Biology, University of TorontoTorontoCanada
| | - Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Black Family Stem Cell Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Fabien Le Grand
- Université Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and MuscleLyonFrance
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Black Family Stem Cell Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of TorontoTorontoCanada,Donnelly Centre, University of TorontoTorontoCanada,Department of Cell and Systems Biology, University of TorontoTorontoCanada
| |
Collapse
|
214
|
Arneson-Wissink PC, Doles JD. Quantification of Muscle Stem Cell Differentiation Using Live-Cell Imaging and Eccentricity Measures. Methods Mol Biol 2022; 2429:455-471. [PMID: 35507181 PMCID: PMC11476825 DOI: 10.1007/978-1-0716-1979-7_31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Culturing primary muscle stem cells ex vivo is a useful method for studying this cell population in controlled environments. Primary muscle stem cells respond to external stimuli differently than immortalized myoblasts (C2C12 cells), making ex vivo culture of muscle stem cells an important tool in understanding cell responses to stimuli. Primary muscle stem cells cultured ex vivo retain a majority of the characteristics they possess in vivo such as the abilities to differentiate into multinucleated structures, and self-renew a stem cell-like population. In this chapter, we describe methods for isolating primary muscle stem cells, controlled differentiation into myotubes, and quantification of differentiation using IncuCyte live cell imaging and analysis software.
Collapse
Affiliation(s)
| | - Jason D Doles
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
215
|
Domenig SA, Bundschuh N, Lenardič A, Ghosh A, Kim I, Qabrati X, D'Hulst G, Bar-Nur O. CRISPR/Cas9 editing of directly reprogrammed myogenic progenitors restores dystrophin expression in a mouse model of muscular dystrophy. Stem Cell Reports 2021; 17:321-336. [PMID: 34995499 PMCID: PMC8828535 DOI: 10.1016/j.stemcr.2021.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 01/09/2023] Open
Abstract
Genetic mutations in dystrophin manifest in Duchenne muscular dystrophy (DMD), the most commonly inherited muscle disease. Here, we report on reprogramming of fibroblasts from two DMD mouse models into induced myogenic progenitor cells (iMPCs) by MyoD overexpression in concert with small molecule treatment. DMD iMPCs proliferate extensively, while expressing myogenic stem cell markers including Pax7 and Myf5. Additionally, DMD iMPCs readily give rise to multinucleated myofibers that express mature skeletal muscle markers; however, they lack DYSTROPHIN expression. Utilizing an exon skipping-based approach with CRISPR/Cas9, we report on genetic correction of the dystrophin mutation in DMD iMPCs and restoration of protein expression in vitro. Furthermore, engraftment of corrected DMD iMPCs into the muscles of dystrophic mice restored DYSTROPHIN expression and contributed to the muscle stem cell reservoir. Collectively, our findings report on a novel in vitro cellular model for DMD and utilize it in conjunction with gene editing to restore DYSTROPHIN expression in vivo. iMPCs generated from DMD mouse models DMD iMPCs are expandable and express satellite cell and differentiation markers Correction of the dystrophin mutation in DMD iMPCs utilizing CRISPR/Cas9 Engraftment of corrected DMD iMPCs restores DYSTROPHIN expression in vivo
Collapse
Affiliation(s)
- Seraina A Domenig
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Nicola Bundschuh
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Ajda Lenardič
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Adhideb Ghosh
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland; Functional Genomics Center Zurich, Swiss Federal Institute of Technology (ETH) Zurich and University of Zurich, Zurich, Switzerland
| | - Inseon Kim
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Xhem Qabrati
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Gommaar D'Hulst
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland
| | - Ori Bar-Nur
- Laboratory of Regenerative and Movement Biology, Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
216
|
Eigler T, Zarfati G, Amzallag E, Sinha S, Segev N, Zabary Y, Zaritsky A, Shakked A, Umansky KB, Schejter ED, Millay DP, Tzahor E, Avinoam O. ERK1/2 inhibition promotes robust myotube growth via CaMKII activation resulting in myoblast-to-myotube fusion. Dev Cell 2021; 56:3349-3363.e6. [PMID: 34932950 PMCID: PMC8693863 DOI: 10.1016/j.devcel.2021.11.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 07/28/2021] [Accepted: 11/21/2021] [Indexed: 11/19/2022]
Abstract
Myoblast fusion is essential for muscle development and regeneration. Yet, it remains poorly understood how mononucleated myoblasts fuse with preexisting fibers. We demonstrate that ERK1/2 inhibition (ERKi) induces robust differentiation and fusion of primary mouse myoblasts through a linear pathway involving RXR, ryanodine receptors, and calcium-dependent activation of CaMKII in nascent myotubes. CaMKII activation results in myotube growth via fusion with mononucleated myoblasts at a fusogenic synapse. Mechanistically, CaMKII interacts with and regulates MYMK and Rac1, and CaMKIIδ/γ knockout mice exhibit smaller regenerated myofibers following injury. In addition, the expression of a dominant negative CaMKII inhibits the formation of large multinucleated myotubes. Finally, we demonstrate the evolutionary conservation of the pathway in chicken myoblasts. We conclude that ERK1/2 represses a signaling cascade leading to CaMKII-mediated fusion of myoblasts to myotubes, providing an attractive target for the cultivated meat industry and regenerative medicine.
Collapse
Affiliation(s)
- Tamar Eigler
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Giulia Zarfati
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Emmanuel Amzallag
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sansrity Sinha
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Nadav Segev
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yishaia Zabary
- Department of Software & Information Systems Engineering, Ben Gurion University, Be'er Sheva, Israel
| | - Assaf Zaritsky
- Department of Software & Information Systems Engineering, Ben Gurion University, Be'er Sheva, Israel
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kfir-Baruch Umansky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal D Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - 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
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
217
|
Zofkie W, Southard SM, Braun T, Lepper C. Fibroblast growth factor 6 regulates sizing of the muscle stem cell pool. Stem Cell Reports 2021; 16:2913-2927. [PMID: 34739848 PMCID: PMC8693628 DOI: 10.1016/j.stemcr.2021.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 11/25/2022] Open
Abstract
Skeletal muscle stem cells, i.e., satellite cells (SCs), are the essential source of new myonuclei for skeletal muscle regeneration following injury or chronic degenerative myopathies. Both SC number and regenerative capacity diminish during aging. However, molecular regulators that govern sizing of the initial SC pool are unknown. We demonstrate that fibroblast growth factor 6 (FGF6) is critical for SC pool scaling. Mice lacking FGF6 have reduced SCs of early postnatal origin and impaired regeneration. By contrast, increasing FGF6 during the early postnatal period is sufficient for SC expansion. Together, these data support that FGF6 is necessary and sufficient to modulate SC numbers during a critical postnatal period to establish the quiescent adult muscle stem cell pool. Our work highlights postnatal development as a time window receptive for scaling a somatic stem cell population via growth factor signaling, which might be relevant for designing new biomedical strategies to enhance tissue regeneration.
Collapse
Affiliation(s)
- William Zofkie
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | | | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Christoph Lepper
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
218
|
Englund DA, Zhang X, Aversa Z, LeBrasseur NK. Skeletal muscle aging, cellular senescence, and senotherapeutics: Current knowledge and future directions. Mech Ageing Dev 2021; 200:111595. [PMID: 34742751 PMCID: PMC8627455 DOI: 10.1016/j.mad.2021.111595] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/16/2022]
Abstract
Cellular senescence is a state of cell cycle arrest induced by several forms of metabolic stress. Senescent cells accumulate with advancing age and have a distinctive phenotype, characterized by profound chromatin alterations and a robust senescence-associated secretory phenotype (SASP) that exerts negative effects on tissue health, both locally and systemically. In preclinical models, pharmacological agents that eliminate senescent cells (senotherapeutics) restore health and youthful properties in multiple tissues. To date, however, very little is understood about the vulnerability of terminally-differentiated skeletal muscle fibers and the resident mononuclear cells that populate the interstitial microenvironment of skeletal muscle to senescence, and their contribution to the onset and progression of skeletal muscle loss and dysfunction with aging. Scientific advances in these areas have the potential to highlight new therapeutic approaches to optimize late-life muscle health. To this end, this review highlights the current evidence and the key questions that need to be addressed to advance the field's understanding of cellular senescence as a mediator of skeletal muscle aging and the potential for emerging senescent cell-targeting therapies to counter age-related deficits in muscle mass, strength, and function. This article is part of the Special Issue - Senolytics - Edited by Joao Passos and Diana Jurk.
Collapse
Affiliation(s)
- Davis A Englund
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Zaira Aversa
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Nathan K LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA; Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
219
|
Valentino T, Figueiredo VC, Mobley CB, McCarthy JJ, Vechetti IJ. Evidence of myomiR regulation of the pentose phosphate pathway during mechanical load-induced hypertrophy. Physiol Rep 2021; 9:e15137. [PMID: 34889054 PMCID: PMC8661100 DOI: 10.14814/phy2.15137] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/13/2021] [Accepted: 11/10/2021] [Indexed: 12/29/2022] Open
Abstract
Many of the molecular and cellular mechanisms discovered to regulate skeletal muscle hypertrophy were first identified using the rodent synergist ablation model. This model reveals the intrinsic capability and necessary pathways of skeletal muscle growth in response to mechanical overload (MOV). Reminiscent of the rapid cellular growth observed with cancer, we hypothesized that in response to MOV, skeletal muscle would undergo metabolic programming to sustain increased demands to support hypertrophy. To test this hypothesis, we analyzed the gene expression of specific metabolic pathways taken from transcriptomic microarray data of a MOV time course. We found an upregulation of genes involved in the oxidative branch of the pentose phosphate pathways (PPP) and mitochondrial branch of the folate cycle suggesting an increase in the production of NADPH. In addition, we sought to determine the potential role of skeletal muscle-enriched microRNA (myomiRs) and satellite cells in the regulation of the metabolic pathways that changed during MOV. We observed an inverse pattern in gene expression between muscle-enriched myomiR-1 and its known target gene glucose-6-phosphate dehydrogenase, G6pdx, suggesting myomiR regulation of PPP activation in response to MOV. Satellite cell fusion had a significant but modest impact on PPP gene expression. These transcriptomic findings suggest the robust muscle hypertrophy induced by MOV requires enhanced redox metabolism via PPP production of NADPH which is potentially regulated by a myomiR network.
Collapse
Affiliation(s)
- Taylor Valentino
- Department of PhysiologyCollege of MedicineLexingtonKentuckyUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Vandre C. Figueiredo
- Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
- Department of Physical TherapyCollege of Health SciencesUniversity of KentuckyLexingtonKentuckyUSA
| | | | - John J. McCarthy
- Department of PhysiologyCollege of MedicineLexingtonKentuckyUSA
- Center for Muscle BiologyUniversity of KentuckyLexingtonKentuckyUSA
| | - Ivan J. Vechetti
- Department of Nutrition and Health SciencesCollege of Education and Human SciencesUniversity of Nebraska‐LincolnLincolnNebraskaUSA
| |
Collapse
|
220
|
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: 80] [Impact Index Per Article: 20.0] [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
|
221
|
Tian L, Jabbari JS, Thijssen R, Gouil Q, Amarasinghe SL, Voogd O, Kariyawasam H, Du MRM, Schuster J, Wang C, Su S, Dong X, Law CW, Lucattini A, Prawer YDJ, Collar-Fernández C, Chung JD, Naim T, Chan A, Ly CH, Lynch GS, Ryall JG, Anttila CJA, Peng H, Anderson MA, Flensburg C, Majewski I, Roberts AW, Huang DCS, Clark MB, Ritchie ME. Comprehensive characterization of single-cell full-length isoforms in human and mouse with long-read sequencing. Genome Biol 2021; 22:310. [PMID: 34763716 PMCID: PMC8582192 DOI: 10.1186/s13059-021-02525-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022] Open
Abstract
A modified Chromium 10x droplet-based protocol that subsamples cells for both short-read and long-read (nanopore) sequencing together with a new computational pipeline (FLAMES) is developed to enable isoform discovery, splicing analysis, and mutation detection in single cells. We identify thousands of unannotated isoforms and find conserved functional modules that are enriched for alternative transcript usage in different cell types and species, including ribosome biogenesis and mRNA splicing. Analysis at the transcript level allows data integration with scATAC-seq on individual promoters, improved correlation with protein expression data, and linked mutations known to confer drug resistance to transcriptome heterogeneity.
Collapse
Affiliation(s)
- Luyi Tian
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| | - Jafar S Jabbari
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Australian Genome Research Facility, Victorian Comprehensive Cancer Centre, Melbourne, VIC, Australia
| | - Rachel Thijssen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Quentin Gouil
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Shanika L Amarasinghe
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Oliver Voogd
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Hasaru Kariyawasam
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Mei R M Du
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jakob Schuster
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Changqing Wang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Shian Su
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Xueyi Dong
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Charity W Law
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Alexis Lucattini
- Australian Genome Research Facility, Victorian Comprehensive Cancer Centre, Melbourne, VIC, Australia
| | - Yair David Joseph Prawer
- Centre for Stem Cell Systems, Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | | | - Jin D Chung
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Timur Naim
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Audrey Chan
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Chi Hai Ly
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia
- Present address: Department of Neurology, Stanford University, Stanford, CA, USA
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - James G Ryall
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia
- Present address: VOW, North Parramatta, NSW, Australia
| | - Casey J A Anttila
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Hongke Peng
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Mary Ann Anderson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Christoffer Flensburg
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Ian Majewski
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Andrew W Roberts
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
- Victorian Comprehensive Cancer Centre, Melbourne, VIC, Australia
| | - David C S Huang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Michael B Clark
- Centre for Stem Cell Systems, Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Matthew E Ritchie
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
222
|
Blum JE, Gheller BJ, Benvie A, Field MS, Panizza E, Vacanti NM, Berry D, Thalacker-Mercer A. Pyruvate Kinase M2 Supports Muscle Progenitor Cell Proliferation but Is Dispensable for Skeletal Muscle Regeneration after Injury. J Nutr 2021; 151:3313-3328. [PMID: 34383048 PMCID: PMC8562082 DOI: 10.1093/jn/nxab251] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/21/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Skeletal muscle progenitor cells (MPCs) repair damaged muscle postinjury. Pyruvate kinase M2 (PKM2) is a glycolytic enzyme (canonical activity) that can also interact with other proteins (noncanonical activity) to modify diverse cellular processes. Recent evidence links PKM2 to MPC proliferation. OBJECTIVES This study aimed to understand cellular roles for PKM2 in MPCs and the necessity of PKM2 in MPCs for muscle regeneration postinjury. METHODS Cultured, proliferating MPCs (C2C12 cells) were treated with a short hairpin RNA targeting PKM2 or small molecules that selectively affect canonical and noncanonical PKM2 activity (shikonin and TEPP-46). Cell number was measured, and RNA-sequencing and metabolic assays were used in follow-up experiments. Immunoprecipitation coupled to proteomics was used to identify binding partners of PKM2. Lastly, an MPC-specific PKM2 knockout mouse was generated and challenged with a muscle injury to determine the impact of PKM2 on regeneration. RESULTS When the noncanonical activity of PKM2 was blocked or impaired, there was an increase in reactive oxygen species concentrations (1.6-2.0-fold, P < 0.01). Blocking noncanonical PKM2 activity also increased lactate excretion (1.2-1.6-fold, P < 0.05) and suppressed mitochondrial oxygen consumption (1.3-1.6-fold, P < 0.01). Glutamate dehydrogenase 1 (GLUD1) was identified as a PKM2 binding partner and blocking noncanonical PKM2 activity increased GLUD activity (1.5-1.6-fold, P < 0.05). Mice with an MPC-specific PKM2 deletion did not demonstrate impaired muscle regeneration. CONCLUSIONS The results suggest that the noncanonical activity of PKM2 is important for MPC proliferation in vitro and demonstrate GLUD1 as a PKM2 binding partner. Because no impairments in muscle regeneration were detected in a mouse model, the endogenous environment may compensate for loss of PKM2.
Collapse
Affiliation(s)
- Jamie E Blum
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Brandon J Gheller
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Abby Benvie
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Elena Panizza
- Department of Molecular Medicine, Cornell University, Ithaca, NY, USA
| | | | - Daniel Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Anna Thalacker-Mercer
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
223
|
Fu C, Huang AH, Galatz LM, Han WM. Cellular and molecular modulation of rotator cuff muscle pathophysiology. J Orthop Res 2021; 39:2310-2322. [PMID: 34553789 DOI: 10.1002/jor.25179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/04/2021] [Accepted: 09/07/2021] [Indexed: 02/04/2023]
Abstract
Rotator cuff (RC) tendon tears are common shoulder injuries that result in irreversible and persistent degeneration of the associated muscles, which is characterized by severe inflammation, atrophy, fibrosis, and fatty infiltration. Although RC muscle degeneration strongly dictates the overall clinical outcomes, strategies to stimulate RC muscle regeneration have largely been overlooked to date. In this review, we highlight the current understanding of the cellular processes that coordinate muscle regeneration, and the roles of muscle resident cells, including immune cells, fibroadipogenic progenitors, and muscle satellite cells in the pathophysiologic regulation of RC muscles following injury. This review also provides perspectives for potential therapies to alleviate the hallmarks of RC muscle degeneration to address current limitations in postsurgical recovery.
Collapse
Affiliation(s)
- Chengcheng Fu
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Department of Orthopedic Surgery, Columbia University, New York City, New York, USA
| | - Leesa M Galatz
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Woojin M Han
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| |
Collapse
|
224
|
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
|
225
|
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
|
226
|
Schmidt M, Weidemann A, Poser C, Bigot A, von Maltzahn J. Stimulation of Non-canonical NF-κB Through Lymphotoxin-β-Receptor Impairs Myogenic Differentiation and Regeneration of Skeletal Muscle. Front Cell Dev Biol 2021; 9:721543. [PMID: 34676210 PMCID: PMC8523804 DOI: 10.3389/fcell.2021.721543] [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: 06/07/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Myogenic differentiation, muscle stem cell functionality, and regeneration of skeletal muscle are cellular processes under tight control of various signaling pathways. Here, we investigated the role of non-canonical NF-κB signaling in myogenic differentiation, muscle stem cell functionality, and regeneration of skeletal muscle. We stimulated non-canonical NF-κB signaling with an agonistically acting antibody of the lymphotoxin beta receptor (LTβR). Interestingly, we found that stimulation of non-canonical NF-κB signaling through the LTβR agonist impairs myogenic differentiation, muscle stem cell function, and regeneration of skeletal muscle. Furthermore, we show that stimulation of non-canonical NF-κB signaling by the LTβR agonist coincides with activation of canonical NF-κB signaling. We suggest a direct crosstalk between canonical and non-canonical NF-κB signaling during myogenic differentiation which is required for proper myogenic differentiation and thereby regeneration of skeletal muscle.
Collapse
Affiliation(s)
- Manuel Schmidt
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Anja Weidemann
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Christine Poser
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Anne Bigot
- Center of Research in Myology-UMRS 974, Institute of Myology, INSERM, Sorbonne Université, Paris, France
| | | |
Collapse
|
227
|
The Role of Satellite Cells in Skeletal Muscle Regeneration-The Effect of Exercise and Age. BIOLOGY 2021; 10:biology10101056. [PMID: 34681155 PMCID: PMC8533525 DOI: 10.3390/biology10101056] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/03/2022]
Abstract
Simple Summary Studies describing the effects of various forms of exercise and age on muscle regeneration were reviewed. Satellite cells are a heterogeneous group of cells that includes stem cells and skeletal muscle progenitor cells. Each skeletal muscle fiber has its own pool of satellite cells that remain inactive until the muscle is damaged. Minor damage within the cell membrane of muscle fibers is patched by fusing intracellular vesicles with the damaged sarcolemma. More severe muscle damage initiates a multistep regeneration process in which satellite cells play an essential role. The condition that initiates the cascade of reactions is the formation of inflammation at the structural discontinuity site, resulting in satellite cell activation. The multitude of reactions and pathways occurring during this process means that many different substances are involved in it and control it. Not all of them are well-understood yet. In parallel, the body’s own population of satellite cells is being rebuilt so that more fibers can be regenerated in the future. Athletes and the elderly are primarily at risk for muscle damage, and pathologies in muscle fiber regeneration cause serious diseases. Abstract The population of satellite cells (mSCs) is highly diversified. The cells comprising it differ in their ability to regenerate their own population and differentiate, as well as in the properties they exhibit. The heterogeneity of this group of cells is evidenced by multiple differentiating markers that enable their recognition, classification, labeling, and characterization. One of the main tasks of satellite cells is skeletal muscle regeneration. Myofibers are often damaged during vigorous exercise in people who participate in sports activities. The number of satellite cells and the speed of the regeneration processes that depend on them affect the time structure of an athlete’s training. This process depends on inflammatory cells. The multitude of reactions and pathways that occur during the regeneration process results in the participation and control of many factors that are activated and secreted during muscle fiber damage and at different stages of its regeneration. However, not all of them are well understood yet. This paper presents the current state of knowledge on satellite cell-dependent skeletal muscle regeneration. Studies describing the effects of various forms of exercise and age on this process were reviewed.
Collapse
|
228
|
Wroblewski OM, Nguyen MH, Cederna P, Larkin LM. Impact of Cell-Seeding Density and Cell Confluence on Human Tissue Engineered Skeletal Muscle. Tissue Eng Part A 2021; 28:420-432. [PMID: 34652973 PMCID: PMC9131361 DOI: 10.1089/ten.tea.2021.0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tissue engineering methodologies have the potential to treat volumetric muscle loss (VML) via the growth of exogenous skeletal muscle grafts from small autogenous muscle biopsies. A significant obstacle preventing the widespread use of engineered skeletal muscle grafts in a clinical setting is the high number of skeletal muscle stem cells, known as satellite cells, required for fabrication of human-sized skeletal muscle tissue. Additionally, there is a lack of work adapting engineered constructs created for animal models into skeletal muscle engineered from a primary human skeletal muscle cell source. For this study, we used scaffold-free tissue-engineered skeletal muscle units (SMUs) to determine the impact of cell seeding density on the ability to fabricate functional human engineered skeletal muscle. Following established protocols, human skeletal muscle isolates were cultured into SMUs at five different cell-seeding densities: 1,000 cells/cm2, 2,500 cells/cm2, 5,000 cells/cm2, 10,000 cells/cm2, and 25,000 cells/cm2. Following previous human SMU work, SMUs prepared at a cell seeding density of 10,000 cells/cm2 served as controls. Additionally, the impact of cell monolayer confluency on the outcome of human cell-sourced SMU fabrication was investigated at both the 1,000 cells/cm2 and 10,000 cells/cm2 seeding densities. Light microscopy was used to examine myotube formation and hypertrophy in cell monolayers. After the formation of three-dimensional constructs, SMUs underwent maximum tetanic isometric force production measurements and immunohistochemical staining to examine SMU contractile function and muscle-like structure, respectively. Results indicate that the 25,000 cells/cm2 cell-seeding density was detrimental to the contractile function of human cell-sourced SMUs, which had significantly lower maximum tetanic forces compared to SMUs seeded at lower densities. Compared to control, low cell-seeding densities (1,000 cells/cm2 - 5,000 cells/cm2) have no detrimental impact on SMU skeletal muscle growth, maturation, or contractility. Cell cultures seeded at 1,000 cells/cm2 and allowed to proliferate to 90-100% confluency prior to treatment in muscle differentiation media (MDM) resulted in SMUs with greater contractile forces and total muscle-structure compared to cell cultures switched to MDM when underconfluent or overconfluent. In conclusion, initial cell-seeding density for SMU fabrication can be decreased to as low as 1,000 cells/cm2 without negatively impacting SMU muscle-like structure and function.
Collapse
Affiliation(s)
- Olga Maria Wroblewski
- University of Michigan Department of Biomedical Engineering, 505527, Ann Arbor, Michigan, United States;
| | - Matthew Hung Nguyen
- University of Michigan Department of Molecular and Integrative Physiology, 200746, Ann Arbor, Michigan, United States;
| | - Paul Cederna
- University of Michigan Medical School, 12266, Surgery, Ann Arbor, Michigan, United States;
| | - Lisa Marie Larkin
- University of Michigan Department of Molecular and Integrative Physiology, 200746, Ann Arbor, Michigan, United States;
| |
Collapse
|
229
|
Gao Z, Lu A, Daquinag AC, Yu Y, Huard M, Tseng C, Gao X, Huard J, Kolonin MG. Partial Ablation of Non-Myogenic Progenitor Cells as a Therapeutic Approach to Duchenne Muscular Dystrophy. Biomolecules 2021; 11:biom11101519. [PMID: 34680151 PMCID: PMC8534118 DOI: 10.3390/biom11101519] [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: 07/02/2021] [Revised: 09/27/2021] [Accepted: 10/12/2021] [Indexed: 01/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by the loss of dystrophin, remains incurable. Reduction in muscle regeneration with DMD is associated with the accumulation of fibroadipogenic progenitors (FAPs) differentiating into myofibroblasts and leading to a buildup of the collagenous tissue aggravating DMD pathogenesis. Mesenchymal stromal cells (MSCs) expressing platelet-derived growth factor receptors (PDGFRs) are activated in muscle during DMD progression and give rise to FAPs promoting DMD progression. Here, we hypothesized that muscle dysfunction in DMD could be delayed via genetic or pharmacologic depletion of MSC-derived FAPs. In this paper, we test this hypothesis in dystrophin-deficient mdx mice. To reduce fibro/adipose infiltration and potentiate muscle progenitor cells (MPCs), we used a model for inducible genetic ablation of proliferating MSCs via a suicide transgene, viral thymidine kinase (TK), expressed under the Pdgfrb promoter. We also tested if MSCs from fat tissue, the adipose stromal cells (ASCs), contribute to FAPs and could be targeted in DMD. Pharmacological ablation was performed with a hunter-killer peptide D-CAN targeting ASCs. MSC depletion with these approaches resulted in increased endurance, measured based on treadmill running, as well as grip strength, without significantly affecting fibrosis. Although more research is needed, our results suggest that depletion of pathogenic MSCs mitigates muscle damage and delays the loss of muscle function in mouse models of DMD.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Cell Proliferation/genetics
- Disease Models, Animal
- Dystrophin/genetics
- Humans
- Mesenchymal Stem Cells/metabolism
- Mice
- Mice, Inbred mdx
- Muscle, Skeletal/growth & development
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/therapy
- Myofibroblasts/cytology
- Myofibroblasts/metabolism
- Promoter Regions, Genetic/genetics
- Receptors, Platelet-Derived Growth Factor/genetics
- Stem Cells/cytology
- Stem Cells/metabolism
Collapse
Affiliation(s)
- Zhanguo Gao
- Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA; (Z.G.); (A.C.D.); (Y.Y.)
| | - Aiping Lu
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA; (A.L.); (M.H.); (X.G.)
| | - Alexes C. Daquinag
- Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA; (Z.G.); (A.C.D.); (Y.Y.)
| | - Yongmei Yu
- Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA; (Z.G.); (A.C.D.); (Y.Y.)
| | - Matthieu Huard
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA; (A.L.); (M.H.); (X.G.)
| | - Chieh Tseng
- M.D. Anderson Cancer Center, The University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Xueqin Gao
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA; (A.L.); (M.H.); (X.G.)
| | - Johnny Huard
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA; (A.L.); (M.H.); (X.G.)
- Correspondence: (J.H.); (M.G.K.); Tel.: +970-479-1595 (J.H.); +713-500-3146 (M.G.K.)
| | - Mikhail G. Kolonin
- Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA; (Z.G.); (A.C.D.); (Y.Y.)
- Correspondence: (J.H.); (M.G.K.); Tel.: +970-479-1595 (J.H.); +713-500-3146 (M.G.K.)
| |
Collapse
|
230
|
Soliman H, Theret M, Scott W, Hill L, Underhill TM, Hinz B, Rossi FMV. Multipotent stromal cells: One name, multiple identities. Cell Stem Cell 2021; 28:1690-1707. [PMID: 34624231 DOI: 10.1016/j.stem.2021.09.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multipotent stromal cells (MSCs) are vital for development, maintenance, function, and regeneration of most tissues. They can differentiate along multiple connective lineages, but unlike most other stem/progenitor cells, they carry out various other functions while maintaining their developmental potential. MSCs function as damage sensors, respond to injury by fostering regeneration through secretion of trophic factors as well as extracellular matrix (ECM) molecules, and contribute to fibrotic reparative processes when regeneration fails. Tissue-specific MSC identity, fate(s), and function(s) are being resolved through fate mapping coupled with single cell "omics," providing unparalleled insights into the secret lives of tissue-resident MSCs.
Collapse
Affiliation(s)
- Hesham Soliman
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Aspect Biosystems, Vancouver, BC V6P 6P2, Canada
| | - Marine Theret
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Wilder Scott
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Lesley Hill
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Tully Michael Underhill
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
231
|
Zhang C, Jia S, Huang J, Peng H, Zhang J, Liu L, Zhang W, Xin H, Wang X. A carbonized wormwood modified photothermal microneedle patch for the repair of damaged skeletal muscles. J Mater Chem B 2021; 9:8014-8020. [PMID: 34477628 DOI: 10.1039/d1tb00610j] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, we aimed to achieve an efficient repair of damaged skeletal muscles using polyvinyl alcohol (PVA) soluble microneedle patches (MNP) loaded with carbonized wormwood and prostaglandin E2 (inflammatory factors). The introduction of carbonized wormwood imparted the MNP with near-infrared light heating characteristics that improved the efficiency of prostaglandin E2 delivery while also promoting circulation in the damaged muscle area. Our experimental results showed that, compared with the classical moxibustion treatment, the system could more quickly restore muscle strength and the cross-sectional area of muscle bundle fibers in a mouse model of muscular injury. In addition, it could also successfully induce the proliferation and differentiation of muscle stem cells to effectively repair injured muscle tissues. Above all, this light-controlled photothermal MN (microneedle) drug-delivery system avoided the common problems of traditional moxibustion such as large levels of smoke, slow efficacy and risk of scalding. Collectively, we put forward a safe, accurate and efficient approach for skeletal muscle damage treatment using carbonized wormwood.
Collapse
Affiliation(s)
- Chuxi Zhang
- The First Clinical Medical College, Nanchang University, Nanchang, Jiangxi 330088, China
| | - Shuang Jia
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, China.
| | - Jinlong Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, China.
| | - Haichuan Peng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, China.
| | - Jiao Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, China.
| | - Lubing Liu
- The First Clinical Medical College, Nanchang University, Nanchang, Jiangxi 330088, China
| | - Wei Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, China.
| | - Hongbo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, China.
| | - Xiaolei Wang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330088, China. .,College of Chemistry, Nanchang University, Nanchang, Jiangxi 330088, China
| |
Collapse
|
232
|
Perreault LR, Le TT, Oudin MJ, Black LD. RNA sequencing indicates age-dependent shifts in the cardiac fibroblast transcriptome between fetal, neonatal, and adult developmental ages. Physiol Genomics 2021; 53:414-429. [PMID: 34281425 PMCID: PMC8560366 DOI: 10.1152/physiolgenomics.00074.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/16/2021] [Indexed: 11/22/2022] Open
Abstract
Cardiac fibroblasts are responsible for extracellular matrix turnover and repair in the cardiac environment and serve to help facilitate immune responses. However, it is well established that they have a significant phenotypic heterogeneity with respect to location, physiological conditions, and developmental age. The goal of this study was to provide an in-depth transcriptomic profile of cardiac fibroblasts derived from rat hearts at fetal, neonatal, and adult developmental ages to ascertain variations in gene expression that may drive functional differences in these cells at these specific stages of development. We performed RNA sequencing (RNA-seq) of cardiac fibroblasts isolated from fetal, neonatal, and adult rats and compared with the rat genome. Principal component analysis of RNA-seq data suggested that data variance was predominantly due to developmental age. Differential expression and gene set enrichment analysis against Gene Ontology and Kyoto Encyclopedia of Genes and Genomes datasets indicated an array of differences across developmental ages, including significant decreases in cardiac development and cardiac function-associated genes with age and a significant increase in immune- and inflammatory-associated functions, particularly immune cell signaling and cytokine and chemokine production, with respect to increasing developmental age. These results reinforce established evidence of diverse phenotypic heterogeneity of fibroblasts with respect to developmental age. Furthermore, based on our analysis of gene expression, age-specific alterations in cardiac fibroblasts may play a crucial role in observed differences in cardiac inflammation and immune response observed across developmental ages.
Collapse
Affiliation(s)
- Luke R Perreault
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Thanh T Le
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Madeleine J Oudin
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Cellular, Molecular, and Developmental Biology Program, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Lauren D Black
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
- Cellular, Molecular, and Developmental Biology Program, Tufts Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
233
|
Miao W, Ma Z, Tang Z, Yu L, Liu S, Huang T, Wang P, Wu T, Song Z, Zhang H, Li Y, Zhou L. Integrative ATAC-seq and RNA-seq Analysis of the Longissimus Muscle of Luchuan and Duroc Pigs. Front Nutr 2021; 8:742672. [PMID: 34660666 PMCID: PMC8511529 DOI: 10.3389/fnut.2021.742672] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
Luchuan pig is a typical obese pig breed in China, and the diameter and area of its longissimus dorsi muscle fibers are significantly smaller than those of Duroc (lean) pig. Skeletal muscle fiber characteristics are related to meat quality of livestock. There is a significant correlation between the quality of different breeds of pork and the characteristics of muscle fiber, which is an important factor affecting the quality of pork. The diameter and area of muscle fibers are related to muscle growth and development. Therefore, we used the assay for transposase-accessible chromatin using sequencing (ATAC-seq) and RNA sequencing (RNA-seq) analysis to investigate the potential mechanism underlying the difference in skeletal muscle growth and development between the two types of pigs. First, transposase-accessible chromatin was analyzed to map the landscape of open chromatin regions and transcription factor binding sites. We identified several transcription factors that potentially affected muscle growth and development, including TFAP4, MAX, NHLH1, FRX5, and TGIF1. We also found that transcription factors with basic helix-loop-helix structures had a preference for binding to genes involved in muscle development. Then, by integrating ATAC-seq and RNA-seq, we found that the Wnt signaling pathway, the mTOR signaling pathway, and other classical pathways regulate skeletal muscle development. In addition, some pathways that might regulate skeletal muscle growth, such as parathyroid hormone synthesis, secretion, and action, synthesis and degradation of ketone bodies, and the thyroid hormone signaling pathway, which were significantly enriched. After further study, we identified a number of candidate genes (ASNS, CARNS1, G0S2, PPP1R14C, and SH3BP5) that might be associated with muscle development. We also found that the differential regulation of chromatin openness at the level of some genes was contrary to the differential regulation at the level of transcription, suggesting that transcription factors and transcriptional repressors may be involved in the regulation of gene expression. Our study provided an in-depth understanding of the mechanism behind the differences in muscle fibers from two species of pig and provided an important foundation for further research on improving the quality of pork.
Collapse
Affiliation(s)
- Weiwei Miao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Zeqiang Ma
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Zhanyang Tang
- Tilapia Seed Farm, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Lin Yu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Siqi Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Tengda Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Peng Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Tian Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Ziyi Song
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Haojie Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yixing Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Lei Zhou
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
234
|
Saito Y, Chikenji TS. Diverse Roles of Cellular Senescence in Skeletal Muscle Inflammation, Regeneration, and Therapeutics. Front Pharmacol 2021; 12:739510. [PMID: 34552495 PMCID: PMC8450382 DOI: 10.3389/fphar.2021.739510] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/23/2021] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle undergoes vigorous tissue remodeling after injury. However, aging, chronic inflammatory diseases, sarcopenia, and neuromuscular disorders cause muscle loss and degeneration, resulting in muscular dysfunction. Cellular senescence, a state of irreversible cell cycle arrest, acts during normal embryonic development and remodeling after tissue damage; when these processes are complete, the senescent cells are eliminated. However, the accumulation of senescent cells is a hallmark of aging tissues or pathological contexts and may lead to progressive tissue degeneration. The mechanisms responsible for the effects of senescent cells have not been fully elucidated. Here, we review current knowledge about the beneficial and detrimental effects of senescent cells in tissue repair, regeneration, aging, and age-related disease, especially in skeletal muscle. We also discuss how senescence of muscle stem cells and muscle-resident fibro-adipogenic progenitors affects muscle pathologies or regeneration, and consider the possibility that immunosenescence leads to muscle pathogenesis. Finally, we explore senotherapy, the therapeutic targeting of senescence to treat age-related disease, from the standpoint of improving muscle regeneration.
Collapse
Affiliation(s)
- Yuki Saito
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takako S Chikenji
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Health Sciences, School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
235
|
Sun KX, Jiang XY, Li X, Su YJ, Wang JL, Zhang L, Yang YM, Zhu XJ. Deletion of phosphatidylserine flippase β-subunit Tmem30a in satellite cells leads to delayed skeletal muscle regeneration. Zool Res 2021; 42:650-659. [PMID: 34472226 PMCID: PMC8455468 DOI: 10.24272/j.issn.2095-8137.2021.195] [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] [Indexed: 11/07/2022] Open
Abstract
Phosphatidylserine (PS) is distributed asymmetrically in the plasma membrane of eukaryotic cells. Phosphatidylserine flippase (P4-ATPase) transports PS from the outer leaflet of the lipid bilayer to the inner leaflet of the membrane to maintain PS asymmetry. The β subunit TMEM30A is indispensable for transport and proper function of P4-ATPase. Previous studies have shown that the ATP11A and TMEM30A complex is the molecular switch for myotube formation. However, the role of Tmem30a in skeletal muscle regeneration remains elusive. In the current study, Tmem30a was highly expressed in the tibialis anterior (TA) muscles of dystrophin-null (mdx) mice and BaCl2-induced muscle injury model mice. We generated a satellite cell (SC)-specific Tmem30a conditional knockout (cKO) mouse model to investigate the role of Tmem30a in skeletal muscle regeneration. The regenerative ability of cKO mice was evaluated by analyzing the number and diameter of regenerated SCs after the TA muscles were injured by BaCl2-injection. Compared to the control mice, the cKO mice showed decreased Pax7+ and MYH3+ SCs, indicating diminished SC proliferation, and decreased expression of muscular regulatory factors (MYOD and MYOG), suggesting impaired myoblast proliferation in skeletal muscle regeneration. Taken together, these results demonstrate the essential role of Tmem30a in skeletal muscle regeneration.
Collapse
Affiliation(s)
- Kuan-Xiang Sun
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072 China
| | - Xiao-Yan Jiang
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xiao Li
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Yu-Jing Su
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Ju-Lin Wang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Lin Zhang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Ye-Ming Yang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Xian-Jun Zhu
- Health Management Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.,Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072 China.,Department of Ophthalmology, First People's Hospital of Shangqiu, Shangqiu, Henan 476000, China.,Key Laboratory of Tibetan Medicine Research, Chinese Academy of Sciences and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Xining, Qinghai 810008, China. E-mail:
| |
Collapse
|
236
|
Abstract
This protocol describes the biofabrication of 3D millimeter-scale human muscle units, embedding non-planar muscle fibers wrapped by fibroblasts-rich endomysium and intertwined with microvascular networks. Suspended muscle fibers are formed through the self-assembly of human myoblasts within cylindrical cavities generated in a sacrificial gelatin template cast in a 3D printed frame. Following myotube differentiation, muscle fibers are embedded in a 3D matrix containing endothelial cells and muscle-derived fibroblasts. The cellular complexity of the environment is instrumental to drive fibroblast migration towards muscle fibers and to induce the organ-specific differentiation of endothelial cells. This advanced 3D muscle model can be applied to analyze the biological mechanisms underlying specific muscle diseases which involve a complex remodeling of the muscle environment (e.g., muscular dystrophies and fibrosis) whereby the pathological interplay among different cell populations drives the onset and progression of the disease.
Collapse
|
237
|
Tonti OR, Larson H, Lipp SN, Luetkemeyer CM, Makam M, Vargas D, Wilcox SM, Calve S. Tissue-specific parameters for the design of ECM-mimetic biomaterials. Acta Biomater 2021; 132:83-102. [PMID: 33878474 PMCID: PMC8434955 DOI: 10.1016/j.actbio.2021.04.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/18/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) is a complex network of biomolecules that mechanically and biochemically directs cell behavior and is crucial for maintaining tissue function and health. The heterogeneous organization and composition of the ECM varies within and between tissue types, directing mechanics, aiding in cell-cell communication, and facilitating tissue assembly and reassembly during development, injury and disease. As technologies like 3D printing rapidly advance, researchers are better able to recapitulate in vivo tissue properties in vitro; however, tissue-specific variations in ECM composition and organization are not given enough consideration. This is in part due to a lack of information regarding how the ECM of many tissues varies in both homeostatic and diseased states. To address this gap, we describe the components and organization of the ECM, and provide examples for different tissues at various states of disease. While many aspects of ECM biology remain unknown, our goal is to highlight the complexity of various tissues and inspire engineers to incorporate unique components of the native ECM into in vitro platform design and fabrication. Ultimately, we anticipate that the use of biomaterials that incorporate key tissue-specific ECM will lead to in vitro models that better emulate human pathologies. STATEMENT OF SIGNIFICANCE: Biomaterial development primarily emphasizes the engineering of new materials and therapies at the expense of identifying key parameters of the tissue that is being emulated. This can be partially attributed to the difficulty in defining the 3D composition, organization, and mechanics of the ECM within different tissues and how these material properties vary as a function of homeostasis and disease. In this review, we highlight a range of tissues throughout the body and describe how ECM content, cell diversity, and mechanical properties change in diseased tissues and influence cellular behavior. Accurately mimicking the tissue of interest in vitro by using ECM specific to the appropriate state of homeostasis or pathology in vivo will yield results more translatable to humans.
Collapse
Affiliation(s)
- Olivia R Tonti
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Hannah Larson
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah N Lipp
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Callan M Luetkemeyer
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Megan Makam
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Diego Vargas
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sean M Wilcox
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States.
| |
Collapse
|
238
|
Ferrara PJ, Verkerke ARP, Maschek JA, Shahtout JL, Siripoksup P, Eshima H, Johnson JM, Petrocelli JJ, Mahmassani ZS, Green TD, McClung JM, Cox JE, Drummond MJ, Funai K. Low lysophosphatidylcholine induces skeletal muscle myopathy that is aggravated by high-fat diet feeding. FASEB J 2021; 35:e21867. [PMID: 34499764 DOI: 10.1096/fj.202101104r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/27/2021] [Accepted: 08/09/2021] [Indexed: 12/25/2022]
Abstract
Obesity alters skeletal muscle lipidome and promotes myopathy, but it is unknown whether aberrant muscle lipidome contributes to the reduction in skeletal muscle contractile force-generating capacity. Comprehensive lipidomic analyses of mouse skeletal muscle revealed a very strong positive correlation between the abundance of lysophosphatidylcholine (lyso-PC), a class of lipids that is known to be downregulated with obesity, with maximal tetanic force production. The level of lyso-PC is regulated primarily by lyso-PC acyltransferase 3 (LPCAT3), which acylates lyso-PC to form phosphatidylcholine. Tamoxifen-inducible skeletal muscle-specific overexpression of LPCAT3 (LPCAT3-MKI) was sufficient to reduce muscle lyso-PC content in both standard chow diet- and high-fat diet (HFD)-fed conditions. Strikingly, the assessment of skeletal muscle force-generating capacity ex vivo revealed that muscles from LPCAT3-MKI mice were weaker regardless of diet. Defects in force production were more apparent in HFD-fed condition, where tetanic force production was 40% lower in muscles from LPCAT3-MKI compared to that of control mice. These observations were partly explained by reductions in the cross-sectional area in type IIa and IIx fibers, and signs of muscle edema in the absence of fibrosis. Future studies will pursue the mechanism by which LPCAT3 may alter protein turnover to promote myopathy.
Collapse
Affiliation(s)
- Patrick J Ferrara
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Anthony R P Verkerke
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - J Alan Maschek
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Metabolomics, Mass Spectrometry, and Proteomics Core, University of Utah, Salt Lake City, Utah, USA
| | - Justin L Shahtout
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Piyarat Siripoksup
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Hiroaki Eshima
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of International Tourism, Nagasaki International University, Sasebo, Japan
| | - Jordan M Johnson
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - Jonathan J Petrocelli
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Ziad S Mahmassani
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Thomas D Green
- East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - Joseph M McClung
- East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA
| | - James E Cox
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Metabolomics, Mass Spectrometry, and Proteomics Core, University of Utah, Salt Lake City, Utah, USA.,Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| | - Micah J Drummond
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, Utah, USA.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,East Carolina Diabetes & Obesity Institute, East Carolina University, Greenville, North Carolina, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA.,Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
239
|
Uezumi A, Ikemoto-Uezumi M, Zhou H, Kurosawa T, Yoshimoto Y, Nakatani M, Hitachi K, Yamaguchi H, Wakatsuki S, Araki T, Morita M, Yamada H, Toyoda M, Kanazawa N, Nakazawa T, Hino J, Fukada SI, Tsuchida K. Mesenchymal Bmp3b expression maintains skeletal muscle integrity and decreases in age-related sarcopenia. J Clin Invest 2021; 131:139617. [PMID: 33170806 DOI: 10.1172/jci139617] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 10/29/2020] [Indexed: 12/19/2022] Open
Abstract
Age-related sarcopenia constitutes an important health problem associated with adverse outcomes. Sarcopenia is closely associated with fat infiltration in muscle, which is attributable to interstitial mesenchymal progenitors. Mesenchymal progenitors are nonmyogenic in nature but are required for homeostatic muscle maintenance. However, the underlying mechanism of mesenchymal progenitor-dependent muscle maintenance is not clear, nor is the precise role of mesenchymal progenitors in sarcopenia. Here, we show that mice genetically engineered to specifically deplete mesenchymal progenitors exhibited phenotypes markedly similar to sarcopenia, including muscle weakness, myofiber atrophy, alterations of fiber types, and denervation at neuromuscular junctions. Through searching for genes responsible for mesenchymal progenitor-dependent muscle maintenance, we found that Bmp3b is specifically expressed in mesenchymal progenitors, whereas its expression level is significantly decreased during aging or adipogenic differentiation. The functional importance of BMP3B in maintaining myofiber mass as well as muscle-nerve interaction was demonstrated using knockout mice and cultured cells treated with BMP3B. Furthermore, the administration of recombinant BMP3B in aged mice reversed their sarcopenic phenotypes. These results reveal previously unrecognized mechanisms by which the mesenchymal progenitors ensure muscle integrity and suggest that age-related changes in mesenchymal progenitors have a considerable impact on the development of sarcopenia.
Collapse
Affiliation(s)
- Akiyoshi Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Madoka Ikemoto-Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Heying Zhou
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Tamaki Kurosawa
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Yuki Yoshimoto
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Masashi Nakatani
- Faculty of Rehabilitation and Care, Seijoh University, Tokai, Japan
| | - Keisuke Hitachi
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Japan
| | - Hisateru Yamaguchi
- Department of Medical Technology, School of Nursing and Medical Care, Yokkaichi Nursing and Medical Care University, Yokkaichi, Japan
| | - Shuji Wakatsuki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toshiyuki Araki
- Department of Peripheral Nervous System Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Mitsuhiro Morita
- Department of Orthopaedic Surgery, Fujita Health University, Toyoake, Japan
| | - Harumoto Yamada
- Department of Orthopaedic Surgery, Fujita Health University, Toyoake, Japan
| | | | - Nobuo Kanazawa
- Department of Surgery, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology (TMGHIG), Tokyo, Japan
| | | | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kunihiro Tsuchida
- Division for Therapies against Intractable Diseases, Institute for Comprehensive Medical Science (ICMS), Fujita Health University, Toyoake, Japan
| |
Collapse
|
240
|
Murach KA, Fry CS, Dupont-Versteegden EE, McCarthy JJ, Peterson CA. Fusion and beyond: Satellite cell contributions to loading-induced skeletal muscle adaptation. FASEB J 2021; 35:e21893. [PMID: 34480776 PMCID: PMC9293230 DOI: 10.1096/fj.202101096r] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/14/2022]
Abstract
Satellite cells support adult skeletal muscle fiber adaptations to loading in numerous ways. The fusion of satellite cells, driven by cell-autonomous and/or extrinsic factors, contributes new myonuclei to muscle fibers, associates with load-induced hypertrophy, and may support focal membrane damage repair and long-term myonuclear transcriptional output. Recent studies have also revealed that satellite cells communicate within their niche to mediate muscle remodeling in response to resistance exercise, regulating the activity of numerous cell types through various mechanisms such as secretory signaling and cell-cell contact. Muscular adaptation to resistance and endurance activity can be initiated and sustained for a period of time in the absence of satellite cells, but satellite cell participation is ultimately required to achieve full adaptive potential, be it growth, function, or proprioceptive coordination. While significant progress has been made in understanding the roles of satellite cells in adult muscle over the last few decades, many conclusions have been extrapolated from regeneration studies. This review highlights our current understanding of satellite cell behavior and contributions to adaptation outside of regeneration in adult muscle, as well as the roles of satellite cells beyond fusion and myonuclear accretion, which are gaining broader recognition.
Collapse
Affiliation(s)
- Kevin A Murach
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Molecular Muscle Mass Regulation Laboratory, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, Arkansas, USA.,Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
| | - Christopher S Fry
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Athletic Training and Clinical Nutrition, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Esther E Dupont-Versteegden
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - John J McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Charlotte A Peterson
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky, USA.,Department of Physical Therapy, College of Health Sciences, University of Kentucky, Lexington, Kentucky, USA.,Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
241
|
Kanazawa Y, Nagano M, Koinuma S, Sujino M, Minami Y, Sugiyo S, Takeda I, Shigeyoshi Y. Basement membrane recovery process in rat soleus muscle after exercise-induced muscle injury. Connect Tissue Res 2021; 62:519-530. [PMID: 32619127 DOI: 10.1080/03008207.2020.1791839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: Collagen IV is a component of the basement membrane (BM) that provides mechanical support for muscle fibers. In addition, transcription factor 4 (TCF4) is highly expressed in muscle connective tissue fibroblasts and regulates muscle regeneration. However, the expression of collagen IV and TCF4 (+) cells in response to exercise-induced muscle injury is not well known. Here, we investigated the expression and localization of collagen IV and TCF4 (+) cells during the recovery process after muscle injury induced by different exercise loads.Materials and Methods: Muscle injury was observed in the soleus muscle of young Wistar rats after 12 or 18 sets-downhill running (DR) on a treadmill. After running, the rats were permitted to recover for a period of 0.5 days, 2 days, or 7 days.Results: Ectopic localization of collagen IV in injured muscle fibers was observed after DR, and the number increased at 0.5 days after 18 sets DR and at 2 days after 12 or 18 sets DR as compared to the number observed at baseline. BM disruption was observed after DR. TCF4 (+) cells appeared in the inside and around injured muscle fibers at 0.5 day of recovery. After 18 sets DR, TCF4 (+) cells were more abundant for a longer period than that observed after 12 sets DR.Conclusions: DR induces BM disruption accompanied by muscle fiber damage. It is possible that BM destruction may be accompanied by muscle damage and that TCF4 (+) cells contribute to muscle fiber and BM recovery.
Collapse
Affiliation(s)
- Yuji Kanazawa
- Department of Physical Therapy, Osaka University of Human Sciences, Shojyaku, Settsu, Japan.,Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine, Ohnohigashi, Osakasayama, Japan
| | - Mamoru Nagano
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine, Ohnohigashi, Osakasayama, Japan
| | - Satoshi Koinuma
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine, Ohnohigashi, Osakasayama, Japan
| | - Mitsugu Sujino
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine, Ohnohigashi, Osakasayama, Japan
| | - Yoichi Minami
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine, Ohnohigashi, Osakasayama, Japan
| | - Shinichi Sugiyo
- Department of Physical Therapy, Osaka University of Human Sciences, Shojyaku, Settsu, Japan
| | - Isao Takeda
- Department of Physical Therapy, Osaka University of Human Sciences, Shojyaku, Settsu, Japan
| | - Yasufumi Shigeyoshi
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine, Ohnohigashi, Osakasayama, Japan
| |
Collapse
|
242
|
Petrocelli JJ, Mahmassani ZS, Fix DK, Montgomery JA, Reidy PT, McKenzie AI, de Hart NM, Ferrara PJ, Kelley JJ, Eshima H, Funai K, Drummond MJ. Metformin and leucine increase satellite cells and collagen remodeling during disuse and recovery in aged muscle. FASEB J 2021; 35:e21862. [PMID: 34416035 DOI: 10.1096/fj.202100883r] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 11/11/2022]
Abstract
Loss of muscle mass and strength after disuse followed by impaired muscle recovery commonly occurs with aging. Metformin (MET) and leucine (LEU) individually have shown positive effects in skeletal muscle during atrophy conditions but have not been evaluated in combination nor tested as a remedy to enhance muscle recovery following disuse atrophy in aging. The purpose of this study was to determine if a dual treatment of metformin and leucine (MET + LEU) would prevent disuse-induced atrophy and/or promote muscle recovery in aged mice and if these muscle responses correspond to changes in satellite cells and collagen remodeling. Aged mice (22-24 months) underwent 14 days of hindlimb unloading (HU) followed by 7 or 14 days of reloading (7 or 14 days RL). MET, LEU, or MET + LEU was administered via drinking water and were compared to Vehicle (standard drinking water) and ambulatory baseline. We observed that during HU, MET + LEU resolved whole body grip strength and soleus muscle specific force decrements caused by HU. Gastrocnemius satellite cell abundance was increased with MET + LEU treatment but did not alter muscle size during disuse or recovery conditions. Moreover, MET + LEU treatment alleviated gastrocnemius collagen accumulation caused by HU and increased collagen turnover during 7 and 14 days RL driven by a decrease in collagen IV content. Transcriptional pathway analysis revealed that MET + LEU altered muscle hallmark pathways related to inflammation and myogenesis during HU. Together, the dual treatment of MET and LEU was able to increase muscle function, satellite cell content, and reduce collagen accumulation, thus improving muscle quality during disuse and recovery in aging.
Collapse
Affiliation(s)
- Jonathan J Petrocelli
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Ziad S Mahmassani
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Dennis K Fix
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | | | - Paul T Reidy
- Department of Kinesiology, Nutrition and Health, Miami University, Oxford, Ohio, USA
| | - Alec I McKenzie
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Naomi M de Hart
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
| | - Patrick J Ferrara
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Joshua J Kelley
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, USA
| | - Hiroaki Eshima
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
| | - Katsuhiko Funai
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, USA.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| | - Micah J Drummond
- Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, Utah, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
243
|
Translational control by DHX36 binding to 5'UTR G-quadruplex is essential for muscle stem-cell regenerative functions. Nat Commun 2021; 12:5043. [PMID: 34413292 PMCID: PMC8377060 DOI: 10.1038/s41467-021-25170-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 06/06/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle has a remarkable ability to regenerate owing to its resident stem cells (also called satellite cells, SCs). SCs are normally quiescent; when stimulated by damage, they activate and expand to form new fibers. The mechanisms underlying SC proliferative progression remain poorly understood. Here we show that DHX36, a helicase that unwinds RNA G-quadruplex (rG4) structures, is essential for muscle regeneration by regulating SC expansion. DHX36 (initially named RHAU) is barely expressed at quiescence but is highly induced during SC activation and proliferation. Inducible deletion of Dhx36 in adult SCs causes defective proliferation and muscle regeneration after damage. System-wide mapping in proliferating SCs reveals DHX36 binding predominantly to rG4 structures at various regions of mRNAs, while integrated polysome profiling shows that DHX36 promotes mRNA translation via 5′-untranslated region (UTR) rG4 binding. Furthermore, we demonstrate that DHX36 specifically regulates the translation of Gnai2 mRNA by unwinding its 5′ UTR rG4 structures and identify GNAI2 as a downstream effector of DHX36 for SC expansion. Altogether, our findings uncover DHX36 as an indispensable post-transcriptional regulator of SC function and muscle regeneration acting through binding and unwinding rG4 structures at 5′ UTR of target mRNAs. Skeletal muscle stem cells (or satellite cells, SCs) are normally quiescent but activate and expand in response to injury. Here the authors show that induction of DHX36 helicase during SC activation promotes mRNA translation by binding to 5′UTR mRNA G-quadruplexes (rG4) in targets including Gnai2 and unwinding them.
Collapse
|
244
|
P2Y2 promotes fibroblasts activation and skeletal muscle fibrosis through AKT, ERK, and PKC. BMC Musculoskelet Disord 2021; 22:680. [PMID: 34380439 PMCID: PMC8359595 DOI: 10.1186/s12891-021-04569-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Skeletal muscle atrophy and fibrosis are pathological conditions that contribute to morbidity in numerous conditions including aging, cachexia, and denervation. Muscle atrophy is characterized as reduction of muscle fiber size and loss of muscle mass while muscle fibrosis is due to fibroblasts activation and excessive production of extracellular matrix. Purinergic receptor P2Y2 has been implicated in fibrosis. This study aims to elucidate the roles of P2Y2 in sleketal muscle atrophy and fibrosis. METHODS Primary muscle fibroblasts were isolated from wild type and P2Y2 knockout (KO) mice and their proliferating and migrating abilities were assessed by CCK-8 and Transwell migration assays respectively. Fibroblasts were activated with TGF-β1 and assessed by western blot of myofibroblast markers including α-SMA, CTGF, and collagen I. Muscle atrophy and fibrosis were induced by transection of distal sciatic nerve and assessed using Masson staining. RESULTS P2Y2 KO fibroblasts proliferated and migrated significantly slower than WT fibroblasts with or without TGF-β1.The proliferation and ECM production were enhanced by P2Y2 agonist PSB-1114 and inhibited by antagonist AR-C118925. TGF-β1 induced fibrotic activation was abolished by P2Y2 ablation and inhibited by AKT, ERK, and PKC inhibitors. Ablation of P2Y2 reduced denervation induced muscle atrophy and fibrosis. CONCLUSIONS P2Y2 is a promoter of skeletal muscle atrophy and activation of fibroblasts after muscle injury, which signaling through AKT, ERK and PKC. P2Y2 could be a potential intervention target after muscle injury.
Collapse
|
245
|
Siddiqui SH, Park J, Kang D, Khan M, Shim K. Cortisol differentially affects the viability and myogenesis of mono- and co-cultured porcine gluteal muscles satellite cells and fibroblasts. Tissue Cell 2021; 73:101615. [PMID: 34419738 DOI: 10.1016/j.tice.2021.101615] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
Cortisol is a ubiquitously expressed stress hormone. In this study, we investigated the effects of exogenous cortisol on porcine gluteal muscles primary cultured satellite cells and fibroblasts. Satellite cells and fibroblasts were mono-or co-cultured, and cells in each type of culture were categorized into the control and cortisol-treated (treatment) groups. We selected 28 μmol mL-1 cortisol for treatment based on their efficacy. Cortisol treatment reduced viability of monocultured satellite cells and fibroblasts. In both monocultured and co-cultured cells, the nucleus in the treatment group was damaged than that control group. Moreover, the total cell cycle duration was shorter in the treatment group than the control group. PAX-7 expression was upregulated in the control group of co-cultured satellite cells and fibroblasts than those remaining groups. Moreover, MyoD expression was downregulated in the cortisol treated group of both mono-and co-cultured satellite cells compared with that in the control group. In co-cultured fibroblasts, MyoD and MyoG expression was upregulated than those remaining groups. The Cyto-C expression was upregulated in the treatment group compared to the control mono-and co-cultured both cells. These results suggest that the selected experimental dose of cortisol reduced cell viability and myogenesis-related gene expression in the monoculture compared to that in the co-culture of satellite cells and fibroblasts.
Collapse
Affiliation(s)
- Sharif Hasan Siddiqui
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Jinryong Park
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Darae Kang
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Mousumee Khan
- Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University, Jeonju, 54907, Republic of Korea
| | - Kwanseob Shim
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju, 54896, Republic of Korea; Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| |
Collapse
|
246
|
Xiong X, Gao H, Lin Y, Yechoor V, Ma K. Inhibition of Rev-erbα ameliorates muscular dystrophy. Exp Cell Res 2021; 406:112766. [PMID: 34364881 DOI: 10.1016/j.yexcr.2021.112766] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 01/25/2023]
Abstract
Duchene muscular dystrophy leads to progressive muscle structural and functional decline due to chronic degenerative-regenerative cycles. Enhancing the regenerative capacity of dystrophic muscle provides potential therapeutic options. We previously demonstrated that the circadian clock repressor Rev-erbα inhibited myogenesis and Rev-erbα ablation enhanced muscle regeneration. Here we show that Rev-erbα deficiency in the dystrophin-deficient mdx mice promotes regenerative myogenic response to ameliorate muscle damage. Loss of Rev-erbα in mdx mice improved dystrophic pathology and muscle wasting. Rev-erbα-deficient dystrophic muscle exhibit augmented myogenic response, enhanced neo-myofiber formation and attenuated inflammatory response. In mdx myoblasts devoid of Rev-erbα, myogenic differentiation was augmented together with up-regulation of Wnt signaling and proliferative pathways, suggesting that loss of Rev-erbα inhibition of these processes contributed to the improvement in regenerative myogenesis. Collectively, our findings revealed that the loss of Rev-erbα function protects dystrophic muscle from injury by promoting myogenic repair, and inhibition of its activity may have therapeutic utilities for muscular dystrophy.
Collapse
Affiliation(s)
- Xuekai Xiong
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Hongbo Gao
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Yayu Lin
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Vijay Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ke Ma
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
247
|
Lahmann I, Griger J, Chen JS, Zhang Y, Schuelke M, Birchmeier C. Met and Cxcr4 cooperate to protect skeletal muscle stem cells against inflammation-induced damage during regeneration. eLife 2021; 10:57356. [PMID: 34350830 PMCID: PMC8370772 DOI: 10.7554/elife.57356] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Acute skeletal muscle injury is followed by an inflammatory response, removal of damaged tissue, and the generation of new muscle fibers by resident muscle stem cells, a process well characterized in murine injury models. Inflammatory cells are needed to remove the debris at the site of injury and provide signals that are beneficial for repair. However, they also release chemokines, reactive oxygen species, as well as enzymes for clearance of damaged cells and fibers, which muscle stem cells have to withstand in order to regenerate the muscle. We show here that MET and CXCR4 cooperate to protect muscle stem cells against the adverse environment encountered during muscle repair. This powerful cyto-protective role was revealed by the genetic ablation of Met and Cxcr4 in muscle stem cells of mice, which resulted in severe apoptosis during early stages of regeneration. TNFα neutralizing antibodies rescued the apoptosis, indicating that TNFα provides crucial cell-death signals during muscle repair that are counteracted by MET and CXCR4. We conclude that muscle stem cells require MET and CXCR4 to protect them against the harsh inflammatory environment encountered in an acute muscle injury.
Collapse
Affiliation(s)
- Ines Lahmann
- Neurowissenschaftliches Forschungzentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| | - Joscha Griger
- Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| | - Jie-Shin Chen
- Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| | - Yao Zhang
- Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| | - Markus Schuelke
- Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carmen Birchmeier
- Neurowissenschaftliches Forschungzentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Developmental Biology/Signal Transduction Group, Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Society, Berlin, Germany
| |
Collapse
|
248
|
Song R, Zhao S, Xu Y, Hu J, Ke S, Li F, Tian G, Zheng X, Li J, Gu L, Xu Y. MRTF-A regulates myoblast commitment to differentiation by targeting PAX7 during muscle regeneration. J Cell Mol Med 2021; 25:8645-8661. [PMID: 34347392 PMCID: PMC8435411 DOI: 10.1111/jcmm.16820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Myocardin-related transcription factor-A/serum response factor (MRTF-A/SRF), a well-known transcriptional programme, has been proposed to play crucial roles in skeletal muscle development and function. However, whether MRTF-A participates in muscle regeneration and the molecular mechanisms are not completely understood. Here, we show that MRTF-A levels are highly correlated with myogenic genes using a RNA-seq assay, which reveal that MRTF-A knockdown in C2C12 cells significantly reduces PAX7 expression. Subsequent in vitro and in vivo data show that MRTF-A and PAX7 present identical expression patterns during myoblast differentiation and CTX-induced muscle injury and repair. Remarkably, MRTF-A overexpression promotes myoblast proliferation, while inhibiting cell differentiation and the expression of MyoD and MyoG. MRTF-A loss of function produces the opposite effect. Moreover, mice with lentivirus (MRTF-A) injection possesses more PAX7+ satellite cells, but less differentiating MyoD+ and MyoG+ cells, leading subsequently to diminished muscle regeneration. Our mechanistic results reveal that MRTF-A contributes to PAX7-mediated myoblast self-renewal, proliferation, and differentiation by binding to its distal CArG box. Overall, we propose that MRTF-A functions as a novel PAX7 regulator upon myoblast commitment to differentiation, which could provide pathways for dictating muscle stem cell fate and open new avenues to explore stem cell-based therapy for muscle degenerative diseases.
Collapse
Affiliation(s)
- Ruhui Song
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Shengnan Zhao
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Yue Xu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Jian Hu
- Animal Disease Control and Prevention Centre of Chongqing City, Chongqing, 401120, China
| | - Shuangao Ke
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Fan Li
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Gaohui Tian
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Xiao Zheng
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Jiajun Li
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Lixing Gu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Yao Xu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| |
Collapse
|
249
|
Gras S, Blasco A, Mòdol-Caballero G, Tarabal O, Casanovas A, Piedrafita L, Barranco A, Das T, Rueda R, Pereira SL, Navarro X, Esquerda JE, Calderó J. Beneficial effects of dietary supplementation with green tea catechins and cocoa flavanols on aging-related regressive changes in the mouse neuromuscular system. Aging (Albany NY) 2021; 13:18051-18093. [PMID: 34319911 PMCID: PMC8351677 DOI: 10.18632/aging.203336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022]
Abstract
Besides skeletal muscle wasting, sarcopenia entails morphological and molecular changes in distinct components of the neuromuscular system, including spinal cord motoneurons (MNs) and neuromuscular junctions (NMJs); moreover, noticeable microgliosis has also been observed around aged MNs. Here we examined the impact of two flavonoid-enriched diets containing either green tea extract (GTE) catechins or cocoa flavanols on age-associated regressive changes in the neuromuscular system of C57BL/6J mice. Compared to control mice, GTE- and cocoa-supplementation significantly improved the survival rate of mice, reduced the proportion of fibers with lipofuscin aggregates and central nuclei, and increased the density of satellite cells in skeletal muscles. Additionally, both supplements significantly augmented the number of innervated NMJs and their degree of maturity compared to controls. GTE, but not cocoa, prominently increased the density of VAChT and VGluT2 afferent synapses on MNs, which were lost in control aged spinal cords; conversely, cocoa, but not GTE, significantly augmented the proportion of VGluT1 afferent synapses on aged MNs. Moreover, GTE, but not cocoa, reduced aging-associated microgliosis and increased the proportion of neuroprotective microglial phenotypes. Our data indicate that certain plant flavonoids may be beneficial in the nutritional management of age-related deterioration of the neuromuscular system.
Collapse
Affiliation(s)
- Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Guillem Mòdol-Caballero
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Alejandro Barranco
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
| | - Tapas Das
- Abbott Nutrition, Research and Development, Columbus, OH 43215, USA
| | - Ricardo Rueda
- Abbott Nutrition, Research and Development, Granada, Spain
| | | | - Xavier Navarro
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Spain
| | - Josep E. Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| |
Collapse
|
250
|
Mini review: Biomaterials in repair and regeneration of nerve in a volumetric muscle loss. Neurosci Lett 2021; 762:136145. [PMID: 34332029 DOI: 10.1016/j.neulet.2021.136145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 06/28/2021] [Accepted: 07/26/2021] [Indexed: 01/23/2023]
Abstract
Volumetric muscle loss (VML) following a severe trauma or injury is beyond the intrinsic regenerative capacity of muscle tissues, and hence interventional therapy is required. Extensive muscle loss concomitant with damage to neuromuscular components overwhelms the muscles' remarkable regenerative capacity. The loss of nervous and vascular tissue leads to further damage and atrophy, so a combined treatment for neuromuscular junction (NMJ) along with the volumetric muscle regeneration is important. There have been immense advances in the field of tissue engineering for skeletal muscle tissue and peripheral nerve regeneration, but very few address the interdependence of the tissues and the need for combined therapies to repair and regenerate fully functional muscle tissue. This review addresses the problem and presents an overview of the biomaterials that have been studied for tissue engineering of neuromuscular tissues associated with skeletal muscles.
Collapse
|