1
|
Liu Y, Li S, Robertson R, Granet JA, Aubry I, Filippelli RL, Tremblay ML, Chang NC. PTPN1/2 inhibition promotes muscle stem cell differentiation in Duchenne muscular dystrophy. Life Sci Alliance 2025; 8:e202402831. [PMID: 39477543 PMCID: PMC11527974 DOI: 10.26508/lsa.202402831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal disease caused by mutations in the DMD gene that encodes dystrophin. Dystrophin deficiency also impacts muscle stem cells (MuSCs), resulting in impaired asymmetric stem cell division and myogenic commitment. Using MuSCs from DMD patients and the DMD mouse model mdx, we found that PTPN1 phosphatase expression is up-regulated and STAT3 phosphorylation is concomitantly down-regulated in DMD MuSCs. To restore STAT3-mediated myogenic signaling, we examined the effect of K884, a novel PTPN1/2 inhibitor, on DMD MuSCs. Treatment with K884 enhanced STAT3 phosphorylation and promoted myogenic differentiation of DMD patient-derived MuSCs. In MuSCs from mdx mice, K884 treatment increased the number of asymmetric cell divisions, correlating with enhanced myogenic differentiation. Interestingly, the pro-myogenic effect of K884 is specific to human and murine DMD MuSCs and is absent from control MuSCs. Moreover, PTPN1/2 loss-of-function experiments indicate that the pro-myogenic impact of K884 is mediated mainly through PTPN1. We propose that PTPN1/2 inhibition may serve as a therapeutic strategy to restore the myogenic function of MuSCs in DMD.
Collapse
MESH Headings
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Animals
- Cell Differentiation/drug effects
- Humans
- Mice
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics
- Mice, Inbred mdx
- STAT3 Transcription Factor/metabolism
- Stem Cells/metabolism
- Stem Cells/cytology
- Muscle Development/genetics
- Muscle Development/drug effects
- Disease Models, Animal
- Phosphorylation
- Signal Transduction/drug effects
- Muscle, Skeletal/metabolism
Collapse
Affiliation(s)
- Yiyang Liu
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Shulei Li
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Rebecca Robertson
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Jules A Granet
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Isabelle Aubry
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Romina L Filippelli
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Michel L Tremblay
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Natasha C Chang
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| |
Collapse
|
2
|
Catarinella G, Bracaglia A, Skafida E, Procopio P, Ruggieri V, Parisi C, De Bardi M, Borsellino G, Madaro L, Puri PL, Sacco A, Latella L. STAT3 inhibition recovers regeneration of aged muscles by restoring autophagy in muscle stem cells. Life Sci Alliance 2024; 7:e202302503. [PMID: 38843935 PMCID: PMC11157169 DOI: 10.26508/lsa.202302503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
Age-related reduction in muscle stem cell (MuSC) regenerative capacity is associated with cell-autonomous and non-cell-autonomous changes caused by alterations in systemic and skeletal muscle environments, ultimately leading to a decline in MuSC number and function. Previous studies demonstrated that STAT3 plays a key role in driving MuSC expansion and differentiation after injury-activated regeneration, by regulating autophagy in activated MuSCs. However, autophagy gradually declines in MuSCs during lifespan and contributes to the impairment of MuSC-mediated regeneration of aged muscles. Here, we show that STAT3 inhibition restores the autophagic process in aged MuSCs, thereby recovering MuSC ability to promote muscle regeneration in geriatric mice. We show that STAT3 inhibition could activate autophagy at the nuclear level, by promoting transcription of autophagy-related genes, and at the cytoplasmic level, by targeting STAT3/PKR phosphorylation of eIF2α. These results point to STAT3 inhibition as a potential intervention to reverse the age-related autophagic block that impairs MuSC ability to regenerate aged muscles. They also reveal that STAT3 regulates MuSC function by both transcription-dependent and transcription-independent regulation of autophagy.
Collapse
Affiliation(s)
| | - Andrea Bracaglia
- IRCCS Fondazione Santa Lucia, Rome, Italy
- PhD Program in Cellular and Molecular Biology, Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Emilia Skafida
- IRCCS Fondazione Santa Lucia, Rome, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Veronica Ruggieri
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Roma "La Sapienza", Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Cristina Parisi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Roma "La Sapienza", Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | | | | | - Luca Madaro
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Roma "La Sapienza", Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Rome, Italy
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Lucia Latella
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Institute of Translational Pharmacology, National Research Council of Italy, Rome, Italy
| |
Collapse
|
3
|
Dan-Jumbo SO, Riley SE, Cortes-Araya Y, Ho W, Lee S, Thrower T, Esteves CL, Donadeu FX. Derivation and long-term maintenance of porcine skeletal muscle progenitor cells. Sci Rep 2024; 14:9370. [PMID: 38653980 DOI: 10.1038/s41598-024-59767-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
Culture of muscle cells from livestock species has typically involved laborious enzyme-based approaches that yield heterogeneous populations with limited proliferative and myogenic differentiation capacity, thus limiting their use in physiologically-meaningful studies. This study reports the use of a simple explant culture technique to derive progenitor cell populations from porcine muscle that could be maintained and differentiated long-term in culture. Fragments of semitendinosus muscle from 4 to 8 week-old piglets (n = 4) were seeded on matrigel coated culture dishes to stimulate migration of muscle-derived progenitor cells (MDPCs). Cell outgrowths appeared within a few days and were serially passaged and characterised using RT-qPCR, immunostaining and flow cytometry. MDPCs had an initial mean doubling time of 1.4 days which increased to 2.5 days by passage 14. MDPC populations displayed steady levels of the lineage-specific markers, PAX7 and MYOD, up until at least passage 2 (positive immunostaining in about 40% cells for each gene), after which the expression of myogenic markers decreased gradually. Remarkably, MDPCs were able to readily generate myotubes in culture up until passage 8. Moreover, a decrease in myogenic capacity during serial passaging was concomitant with a gradual increase in the expression of the pre-adipocyte markers, CD105 and PDGFRA, and an increase in the ability of MDPCs to differentiate into adipocytes. In conclusion, explant culture provided a simple and efficient method to harvest enriched myogenic progenitors from pig skeletal muscle which could be maintained long-term and differentiated in vitro, thus providing a suitable system for studies on porcine muscle biology and applications in the expanding field of cultured meat.
Collapse
Affiliation(s)
- Susan O Dan-Jumbo
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Susanna E Riley
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Yennifer Cortes-Araya
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - William Ho
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Seungmee Lee
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Thomas Thrower
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Cristina L Esteves
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - F Xavier Donadeu
- Division of Translational Bioscience, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK.
| |
Collapse
|
4
|
Denes LT, Kelley CP, Wang ET. Multiplexed Immunofluorescence and Single-Molecule RNA Fluorescence In Situ Hybridization in Mouse Skeletal Myofibers. Methods Mol Biol 2024; 2784:163-176. [PMID: 38502485 DOI: 10.1007/978-1-0716-3766-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
RNA fluorescence in situ hybridization (FISH) is a powerful method to determine the abundance and localization of mRNA molecules in cells. While modern RNA FISH techniques allow quantification at single molecule resolution, most methods are optimized for mammalian cell culture and are not easily applied to in vivo tissue settings. Single-molecule RNA detection in skeletal muscle cells has been particularly challenging due to the thickness and high autofluorescence of adult muscle tissue and a lack of in vitro models for mature muscle cells (myofibers). Here, we present a method for isolation of adult myofibers from mouse skeletal muscle and detection of single mRNA molecules and proteins using multiplexed RNA FISH and immunofluorescence.
Collapse
Affiliation(s)
- Lance T Denes
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA.
| | - Chase P Kelley
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| | - Eric T Wang
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA
| |
Collapse
|
5
|
Zhang S, Yang F, Huang Y, He L, Li Y, Wan YCE, Ding Y, Chan KM, Xie T, Sun H, Wang H. ATF3 induction prevents precocious activation of skeletal muscle stem cell by regulating H2B expression. Nat Commun 2023; 14:4978. [PMID: 37591871 PMCID: PMC10435463 DOI: 10.1038/s41467-023-40465-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 07/27/2023] [Indexed: 08/19/2023] Open
Abstract
Skeletal muscle stem cells (also called satellite cells, SCs) are important for maintaining muscle tissue homeostasis and damage-induced regeneration. However, it remains poorly understood how SCs enter cell cycle to become activated upon injury. Here we report that AP-1 family member ATF3 (Activating Transcription Factor 3) prevents SC premature activation. Atf3 is rapidly and transiently induced in SCs upon activation. Short-term deletion of Atf3 in SCs accelerates acute injury-induced regeneration, however, its long-term deletion exhausts the SC pool and thus impairs muscle regeneration. The Atf3 loss also provokes SC activation during voluntary exercise and enhances the activation during endurance exercise. Mechanistically, ATF3 directly activates the transcription of Histone 2B genes, whose reduction accelerates nucleosome displacement and gene transcription required for SC activation. Finally, the ATF3-dependent H2B expression also prevents genome instability and replicative senescence in SCs. Therefore, this study has revealed a previously unknown mechanism for preserving the SC population by actively suppressing precocious activation, in which ATF3 is a key regulator.
Collapse
Affiliation(s)
- Suyang Zhang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, New Territories, Hong Kong SAR, China
| | - Feng Yang
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yile Huang
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liangqiang He
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, New Territories, Hong Kong SAR, China
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuying Li
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yi Ching Esther Wan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518172, China
| | - Yingzhe Ding
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518172, China
| | - Ting Xie
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Hao Sun
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Huating Wang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China.
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, New Territories, Hong Kong SAR, China.
| |
Collapse
|
6
|
Chen W, Perkins TJ, Rudnicki MA. Quantification of Muscle Satellite Stem Cell Divisions by High-Content Analysis. Methods Mol Biol 2023; 2587:537-553. [PMID: 36401049 DOI: 10.1007/978-1-0716-2772-3_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
High-content screening is commonly performed on 2D cultured cells, which is high throughput but has low biological relevance. In contrast, single myofiber culture assay preserves the satellite cell niche between the basal lamina and sarcolemma and consequently has high biological relevance but is low throughput. We describe here a high-content screening method that utilizes single myofiber culture that addresses the caveats of both techniques. Our method utilizes the transgenic reporter allele Myf5-Cre:R26R-eYFP to differentiate stem and committed cells within a dividing couplet that can be quantified by high-content throughput immunodetection and bioinformatic analysis.
Collapse
Affiliation(s)
- William Chen
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Theodore J Perkins
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
7
|
Montserrat-Vazquez S, Ali NJ, Matteini F, Lozano J, Zhaowei T, Mejia-Ramirez E, Marka G, Vollmer A, Soller K, Sacma M, Sakk V, Mularoni L, Mallm JP, Plass M, Zheng Y, Geiger H, Florian MC. Transplanting rejuvenated blood stem cells extends lifespan of aged immunocompromised mice. NPJ Regen Med 2022; 7:78. [PMID: 36581635 PMCID: PMC9800381 DOI: 10.1038/s41536-022-00275-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
One goal of regenerative medicine is to rejuvenate tissues and extend lifespan by restoring the function of endogenous aged stem cells. However, evidence that somatic stem cells can be targeted in vivo to extend lifespan is still lacking. Here, we demonstrate that after a short systemic treatment with a specific inhibitor of the small RhoGTPase Cdc42 (CASIN), transplanting aged hematopoietic stem cells (HSCs) from treated mice is sufficient to extend the healthspan and lifespan of aged immunocompromised mice without additional treatment. In detail, we show that systemic CASIN treatment improves strength and endurance of aged mice by increasing the myogenic regenerative potential of aged skeletal muscle stem cells. Further, we show that CASIN modifies niche localization and H4K16ac polarity of HSCs in vivo. Single-cell profiling reveals changes in HSC transcriptome, which underlie enhanced lymphoid and regenerative capacity in serial transplantation assays. Overall, we provide proof-of-concept evidence that a short systemic treatment to decrease Cdc42 activity improves the regenerative capacity of different endogenous aged stem cells in vivo, and that rejuvenated HSCs exert a broad systemic effect sufficient to extend murine health- and lifespan.
Collapse
Affiliation(s)
- Sara Montserrat-Vazquez
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | - Noelle J. Ali
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Francesca Matteini
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Lozano
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | - Tu Zhaowei
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Eva Mejia-Ramirez
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain ,grid.512890.7Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Gina Marka
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Angelika Vollmer
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Karin Soller
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Mehmet Sacma
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Vadim Sakk
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Loris Mularoni
- grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain
| | | | - Mireya Plass
- grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain ,grid.512890.7Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain ,grid.417656.7Gene Regulation of Cell Identity Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Yi Zheng
- grid.239573.90000 0000 9025 8099Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Hartmut Geiger
- grid.6582.90000 0004 1936 9748Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - M. Carolina Florian
- grid.417656.7Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain ,grid.417656.7Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Barcelona, Spain ,grid.512890.7Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
8
|
Ranu N, Laitila J, Dugdale HF, Mariano J, Kolb JS, Wallgren-Pettersson C, Witting N, Vissing J, Vilchez JJ, Fiorillo C, Zanoteli E, Auranen M, Jokela M, Tasca G, Claeys KG, Voermans NC, Palmio J, Huovinen S, Moggio M, Beck TN, Kontrogianni-Konstantopoulos A, Granzier H, Ochala J. NEB mutations disrupt the super-relaxed state of myosin and remodel the muscle metabolic proteome in nemaline myopathy. Acta Neuropathol Commun 2022; 10:185. [PMID: 36528760 PMCID: PMC9758823 DOI: 10.1186/s40478-022-01491-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Nemaline myopathy (NM) is one of the most common non-dystrophic genetic muscle disorders. NM is often associated with mutations in the NEB gene. Even though the exact NEB-NM pathophysiological mechanisms remain unclear, histological analyses of patients' muscle biopsies often reveal unexplained accumulation of glycogen and abnormally shaped mitochondria. Hence, the aim of the present study was to define the exact molecular and cellular cascade of events that would lead to potential changes in muscle energetics in NEB-NM. For that, we applied a wide range of biophysical and cell biology assays on skeletal muscle fibres from NM patients as well as untargeted proteomics analyses on isolated myofibres from a muscle-specific nebulin-deficient mouse model. Unexpectedly, we found that the myosin stabilizing conformational state, known as super-relaxed state, was significantly impaired, inducing an increase in the energy (ATP) consumption of resting muscle fibres from NEB-NM patients when compared with controls or with other forms of genetic/rare, acquired NM. This destabilization of the myosin super-relaxed state had dynamic consequences as we observed a remodeling of the metabolic proteome in muscle fibres from nebulin-deficient mice. Altogether, our findings explain some of the hitherto obscure hallmarks of NM, including the appearance of abnormal energy proteins and suggest potential beneficial effects of drugs targeting myosin activity/conformations for NEB-NM.
Collapse
Affiliation(s)
- Natasha Ranu
- grid.13097.3c0000 0001 2322 6764Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK
| | - Jenni Laitila
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark ,grid.7737.40000 0004 0410 2071The Folkhälsan Institute of Genetics and Department of Medical and Clinical Genetics, Medicum, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Hannah F. Dugdale
- grid.13097.3c0000 0001 2322 6764Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, UK ,grid.6571.50000 0004 1936 8542School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Jennifer Mariano
- grid.411024.20000 0001 2175 4264Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA
| | - Justin S. Kolb
- grid.134563.60000 0001 2168 186XDepartment of Cellular and Molecular Medicine, University of Arizona, Tucson, USA
| | - Carina Wallgren-Pettersson
- grid.7737.40000 0004 0410 2071The Folkhälsan Institute of Genetics and Department of Medical and Clinical Genetics, Medicum, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
| | - Nanna Witting
- grid.5254.60000 0001 0674 042XCopenhagen Neuromuscular Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - John Vissing
- grid.5254.60000 0001 0674 042XCopenhagen Neuromuscular Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Juan Jesus Vilchez
- grid.84393.350000 0001 0360 9602Neuromuscular and Ataxias Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) Spain, Valencia, Spain
| | - Chiara Fiorillo
- grid.5606.50000 0001 2151 3065Neuromuscular Disorders Unit, IRCCS Istituto Giannina Gaslini, DINOGMI, University of Genoa, Genoa, Italy
| | - Edmar Zanoteli
- grid.11899.380000 0004 1937 0722Department of Neurology, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Mari Auranen
- grid.7737.40000 0004 0410 2071Clinical Neurosciences, University of Helsinki and Helsinki University Hospital, NeurologyHelsinki, Finland
| | - Manu Jokela
- grid.1374.10000 0001 2097 1371Neurology, Clinical Medicine, University of Turku, Turku, Finland ,grid.410552.70000 0004 0628 215XNeurocenter, Turku University Hospital, Turku, Finland ,grid.502801.e0000 0001 2314 6254Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland
| | - Giorgio Tasca
- grid.414603.4Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy ,grid.1006.70000 0001 0462 7212John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Kristl G. Claeys
- grid.410569.f0000 0004 0626 3338Department of Neurology, University Hospitals Leuven, Leuven, Belgium ,grid.5596.f0000 0001 0668 7884Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Nicol C. Voermans
- grid.10417.330000 0004 0444 9382Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johanna Palmio
- grid.502801.e0000 0001 2314 6254Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland
| | - Sanna Huovinen
- grid.412330.70000 0004 0628 2985Department of Pathology, Fimlab Laboratories, Tampere University Hospital, Tampere, Finland
| | - Maurizio Moggio
- grid.414818.00000 0004 1757 8749Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Thomas Nyegaard Beck
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Henk Granzier
- grid.134563.60000 0001 2168 186XDepartment of Cellular and Molecular Medicine, University of Arizona, Tucson, USA
| | - Julien Ochala
- grid.5254.60000 0001 0674 042XDepartment of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Schüler SC, Liu Y, Dumontier S, Grandbois M, Le Moal E, Cornelison DDW, Bentzinger CF. Extracellular matrix: Brick and mortar in the skeletal muscle stem cell niche. Front Cell Dev Biol 2022; 10:1056523. [PMID: 36523505 PMCID: PMC9745096 DOI: 10.3389/fcell.2022.1056523] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Abstract
The extracellular matrix (ECM) is an interconnected macromolecular scaffold occupying the space between cells. Amongst other functions, the ECM provides structural support to tissues and serves as a microenvironmental niche that conveys regulatory signals to cells. Cell-matrix adhesions, which link the ECM to the cytoskeleton, are dynamic multi-protein complexes containing surface receptors and intracellular effectors that control various downstream pathways. In skeletal muscle, the most abundant tissue of the body, each individual muscle fiber and its associated muscle stem cells (MuSCs) are surrounded by a layer of ECM referred to as the basal lamina. The core scaffold of the basal lamina consists of self-assembling polymeric laminins and a network of collagens that tether proteoglycans, which provide lateral crosslinking, establish collateral associations with cell surface receptors, and serve as a sink and reservoir for growth factors. Skeletal muscle also contains the fibrillar collagenous interstitial ECM that plays an important role in determining tissue elasticity, connects the basal laminae to each other, and contains matrix secreting mesenchymal fibroblast-like cell types and blood vessels. During skeletal muscle regeneration fibroblast-like cell populations expand and contribute to the transitional fibronectin-rich regenerative matrix that instructs angiogenesis and MuSC function. Here, we provide a comprehensive overview of the role of the skeletal muscle ECM in health and disease and outline its role in orchestrating tissue regeneration and MuSC function.
Collapse
Affiliation(s)
- Svenja C. Schüler
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Yuguo Liu
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Simon Dumontier
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Michel Grandbois
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Emmeran Le Moal
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - DDW Cornelison
- Division of Biological Sciences Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - C. Florian Bentzinger
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
10
|
Yamakawa D, Tsuboi J, Kasahara K, Matsuda C, Nishimura Y, Kodama T, Katayama N, Watanabe M, Inagaki M. Cilia-Mediated Insulin/Akt and ST2/JNK Signaling Pathways Regulate the Recovery of Muscle Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2202632. [PMID: 36373718 PMCID: PMC9811445 DOI: 10.1002/advs.202202632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 10/20/2022] [Indexed: 06/04/2023]
Abstract
Following injury, skeletal muscle regenerates but fatty tissue accumulation is seen in aged muscle or muscular dystrophies. Fibro/adipogenic progenitors (FAPs) are key players in these events; however, the effect of primary cilia on FAPs remains unclear. Here, it is reported that genetic ablation of trichoplein (TCHP), a ciliary regulator, induces ciliary elongation on FAPs after injury, which promotes muscle regeneration while inhibiting adipogenesis. The defective adipogenic differentiation of FAPs is attributed to dysfunction of cilia-dependent lipid raft dynamics, which is critical for insulin/Akt signaling. It is also found that interleukin (IL) 13 is substantially produced by intramuscular FAPs, which are upregulated by ciliary elongation and contribute to regeneration. Mechanistically, upon injury, long cilia excessively activate the IL33/ST2/JNK axis to enhance IL13 production, facilitating myoblast proliferation and M2 macrophage polarization. The results indicate that FAPs organize the regenerative responses to skeletal muscle injury via cilia-mediated insulin/Akt and ST2/JNK signaling pathways.
Collapse
Affiliation(s)
- Daishi Yamakawa
- Department of PhysiologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Junya Tsuboi
- Department of Gastroenterology and HepatologyMie University Graduate School of MedicineTsuMie514‐8507Japan
- Department of Hematology and OncologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Kousuke Kasahara
- Department of PhysiologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Chise Matsuda
- Department of Oncogenic PathologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Yuhei Nishimura
- Department of Integrative PharmacologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Tatsuya Kodama
- Department of PhysiologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Naoyuki Katayama
- Department of Hematology and OncologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Masatoshi Watanabe
- Department of Oncogenic PathologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| | - Masaki Inagaki
- Department of PhysiologyMie University Graduate School of MedicineTsuMie514‐8507Japan
| |
Collapse
|
11
|
Buckley KH, Nestor-Kalinoski AL, Pizza FX. Intercellular Adhesion Molecule-1 Enhances Myonuclear Transcription during Injury-Induced Muscle Regeneration. Int J Mol Sci 2022; 23:7028. [PMID: 35806032 PMCID: PMC9267068 DOI: 10.3390/ijms23137028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/11/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
The local inflammatory environment of injured skeletal muscle contributes to the resolution of the injury by promoting the proliferation of muscle precursor cells during the initial stage of muscle regeneration. However, little is known about the extent to which the inflammatory response influences the later stages of regeneration when newly formed (regenerating myofibers) are accumulating myonuclei and undergoing hypertrophy. Our prior work indicated that the inflammatory molecule ICAM-1 facilitates regenerating myofiber hypertrophy through a process involving myonuclear positioning and/or transcription. The present study tested the hypothesis that ICAM-1 enhances global transcription within regenerating myofibers by augmenting the transcriptional activity of myonuclei positioned in linear arrays (nuclear chains). We found that transcription in regenerating myofibers was ~2-fold higher in wild type compared with ICAM-1-/- mice at 14 and 28 days post-injury. This occurred because the transcriptional activity of individual myonuclei in nuclei chains, nuclear clusters, and a peripheral location were ~2-fold higher in wild type compared with ICAM-1-/- mice during regeneration. ICAM-1's enhancement of transcription in nuclear chains appears to be an important driver of myofiber hypertrophy as it was statistically associated with an increase in myofiber size during regeneration. Taken together, our findings indicate that ICAM-1 facilitates myofiber hypertrophy after injury by enhancing myonuclear transcription.
Collapse
Affiliation(s)
- Kole H. Buckley
- School of Exercise and Rehabilitation Sciences, University of Toledo, 2801 W. Bancroft St., Toledo, OH 43606, USA;
| | | | - Francis X. Pizza
- School of Exercise and Rehabilitation Sciences, University of Toledo, 2801 W. Bancroft St., Toledo, OH 43606, USA;
| |
Collapse
|
12
|
Xia W, Qiu J, Peng Y, Snyder MM, Gu L, Huang K, Luo N, Yue F, Kuang S. Chchd10 is dispensable for myogenesis but critical for adipose browning. CELL REGENERATION (LONDON, ENGLAND) 2022; 11:14. [PMID: 35362877 PMCID: PMC8975916 DOI: 10.1186/s13619-022-00111-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/27/2022] [Indexed: 12/20/2022]
Abstract
The Chchd10 gene encodes a coiled-coil-helix-coiled-coil-helix-domain containing protein predicted to function in the mitochondrion and nucleus. Mutations of Chchd10 are associated with ALS, dementia and myopathy in humans and animal models, but how knockout of Chchd10 (Chchd10KO) affects various tissues especially skeletal muscle and adipose tissues remains unclear. Here we show that Chchd10 expression increases as myoblasts and preadipocytes differentiate. During myogenesis, CHCHD10 interacts with TAR DNA binding protein 43 (TDP-43) in regenerating myofibers in vivo and in newly differentiated myotubes ex vivo. Surprisingly, Chchd10KO mice had normal skeletal muscle development, growth and regeneration, with moderate defects in grip strength and motor performance. Chchd10KO similarly had no effects on development of brown and white adipose tissues (WAT). However, Chchd10KO mice had blunted response to acute cold and attenuated cold-induced browning of WAT, with markedly reduced UCP1 levels. Together, these results demonstrate that Chchd10 is dispensable for normal myogenesis and adipogenesis but is required for normal motility and cold-induced, mitochondrion-dependent browning of adipocytes. The data also suggest that human CHCHD10 mutations cause myopathy through a gain-of-function mechanism.
Collapse
Affiliation(s)
- Wei Xia
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, 071000, China. .,Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA. .,College of Animal and Veterinary Science, Southwest Minzu University, Chengdu, 610041, China.
| | - Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Ying Peng
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Madigan M Snyder
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Lijie Gu
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA.,College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Kuilong Huang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Nanjian Luo
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
13
|
Buckley KH, Nestor-Kalinoski AL, Pizza FX. Positional Context of Myonuclear Transcription During Injury-Induced Muscle Regeneration. Front Physiol 2022; 13:845504. [PMID: 35492593 PMCID: PMC9040890 DOI: 10.3389/fphys.2022.845504] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/02/2022] [Indexed: 01/31/2023] Open
Abstract
Fundamental aspects underlying downstream processes of skeletal muscle regeneration, such as myonuclear positioning and transcription are poorly understood. This investigation begins to address deficiencies in knowledge by examining the kinetics of myonuclear accretion, positioning, and global transcription during injury-induced muscle regeneration in mice. We demonstrate that myonuclear accretion plateaus within 7 days of an injury and that the majority (∼70%) of myonuclei are centrally aligned in linear arrays (nuclear chains) throughout the course of regeneration. Relatively few myonuclei were found in a peripheral position (∼20%) or clustered (∼10%) together during regeneration. Importantly, transcriptional activity of individual myonuclei in nuclear chains was high, and greater than that of peripheral or clustered myonuclei. Transcription occurring primarily in nuclear chains elevated the collective transcriptional activity of regenerating myofibers during the later stage of regeneration. Importantly, the number of myonuclei in chains and their transcriptional activity were statistically correlated with an increase in myofiber size during regeneration. Our findings demonstrate the positional context of transcription during regeneration and highlight the importance of centralized nuclear chains in facilitating hypertrophy of regenerating myofibers after injury.
Collapse
Affiliation(s)
- Kole H. Buckley
- School of Exercise and Rehabilitation Sciences, University of Toledo, Toledo, OH, United States
| | | | - Francis X. Pizza
- School of Exercise and Rehabilitation Sciences, University of Toledo, Toledo, OH, United States
| |
Collapse
|
14
|
Elashry MI, Kinde M, Klymiuk MC, Eldaey A, Wenisch S, Arnhold S. The effect of hypoxia on myogenic differentiation and multipotency of the skeletal muscle-derived stem cells in mice. Stem Cell Res Ther 2022; 13:56. [PMID: 35123554 PMCID: PMC8817503 DOI: 10.1186/s13287-022-02730-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/20/2022] [Indexed: 01/01/2023] Open
Abstract
Abstract
Background
Skeletal muscle-derived stem cells (SC) have become a promising approach for investigating myogenic differentiation and optimizing tissue regeneration. Muscle regeneration is performed by SC, a self-renewal cell population underlying the basal lamina of muscle fibers. Here, we examined the impact of hypoxia condition on the regenerative capacity of SC either in their native microenvironment or via isolation in a monolayer culture using ectopic differentiation inductions. Furthermore, the effect of low oxygen tension on myogenic differentiation protocols of the myoblasts cell line C2C12 was examined.
Methods
Hind limb muscles of wild type mice were processed for both SC/fiber isolation and myoblast extraction using magnetic beads. SC were induced for myogenic, adipogenic and osteogenic commitments under normoxic (21% O2) and hypoxic (3% O2) conditions. SC proliferation and differentiation were evaluated using histological staining, immunohistochemistry, morphometric analysis and RT-qPCR. The data were statistically analyzed using ANOVA.
Results
The data revealed enhanced SC proliferation and motility following differentiation induction after 48 h under hypoxia. Following myogenic induction, the number of undifferentiated cells positive for Pax7 were increased at 72 h under hypoxia. Hypoxia upregulated MyoD and downregulated Myogenin expression at day-7 post-myogenic induction. Hypoxia promoted both SC adipogenesis and osteogenesis under respective induction as shown by using Oil Red O and Alizarin Red S staining. The expression of adipogenic markers; peroxisome proliferator activated receptor gamma (PPARγ) and fatty acid-binding protein 4 (FABP4) were upregulated under hypoxia up to day 14 compared to normoxic condition. Enhanced osteogenic differentiation was detected under hypoxic condition via upregulation of osteocalcin and osteopontin expression up to day 14 as well as, increased calcium deposition at day 21. Hypoxia exposure increases the number of adipocytes and the size of fat vacuoles per adipocyte compared to normoxic culture. Combining the differentiation medium with dexamethasone under hypoxia improves the efficiency of the myogenic differentiation protocol of C2C12 by increasing the length of the myotubes.
Conclusions
Hypoxia exposure increases cell resources for clinical applications and promotes SC multipotency and thus beneficial for tissue regeneration.
Collapse
|
15
|
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: 11] [Impact Index Per Article: 3.7] [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
|
16
|
Zhuang J, Zhou T, Bai S, Zhao B, Wu X, Chen Y. Effects of Restricted Feeding on Growth Performance, Intestinal Immunity, and Skeletal Muscle Development in New Zealand Rabbits. Animals (Basel) 2022; 12:ani12020160. [PMID: 35049783 PMCID: PMC8772555 DOI: 10.3390/ani12020160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The high prevalence of gastrointestinal diseases in young rabbits is the major cause of impediment in the development of the rabbit industry. Presently, few companies have adopted methods of restricting feeding to improve the survival rate independent of the effect on their growth and development. To explore the effects of different feeding-restriction levels on the growth performance, intestinal immunity, and skeletal muscle development of meat rabbits, 198 New Zealand meat rabbits of 35 days old were selected and randomly divided into three groups: (1) a control group, (2) a 15% feeding restriction group, and (3) a 30% feeding restriction group, with 66 in each group with an equal number of males and females. The growth performance measurement and health-risk assessment indicators, measurement of digestive enzyme activity, immune and antioxidant indexes, and regulation mechanism were evaluated and explored. Finally, we found that a 30% feeding limit affected the growth and development of skeletal muscle in growing rabbits by regulating the PI3K/Akt signaling pathway. Abstract This study aimed to explore the effects of different feeding restriction levels on the growth performance, intestinal immunity, and skeletal muscle development of meat rabbits. Additionally, we studied whether complete compensatory growth could be obtained post 2 weeks of restricted feeding, in order to seek a scientific mode of feeding restriction. Each of three groups was exposed to 3 weeks of feeding restriction and 2 weeks of compensatory growth. The 15% feeding restriction showed a negligible effect on the final body-weight of the rabbits (p > 0.05), but significantly reduced the feed-to-weight ratio (p < 0.05); reduced diarrhea and mortality; and increased digestive enzyme activity and antioxidant capacity. However, a 30% feeding-restriction level substantially reduced the growth rate of the rabbits (p < 0.05), impaired skeletal muscle development, and showed no compensatory growth after 2 weeks of nutritional recovery. Additionally, immunoglobulin and antioxidant enzyme synthesis were impaired due to reduced nutritional levels, and levels of pro-inflammatory factors were increased during the compensation period. The IGF1 mRNA expression decreased significantly (p < 0.05), whereas MSTN and FOXO1 expression increased noticeably (p < 0.05). Moreover, protein levels of p-Akt and p-p70 decreased significantly in the 15% feeding restriction group. Overall, the 15% feeding limit unaffected the weight and skeletal muscle development of rabbits, whereas the 30% feeding limit affected the growth and development of skeletal muscle in growing rabbits. The PI3K/Akt signaling pathway is plausibly a mediator of this process.
Collapse
Affiliation(s)
| | | | | | | | | | - Yang Chen
- Correspondence: ; Tel.: +86-18762321870
| |
Collapse
|
17
|
Romagnoli C, Iantomasi T, Brandi ML. Available In Vitro Models for Human Satellite Cells from Skeletal Muscle. Int J Mol Sci 2021; 22:ijms222413221. [PMID: 34948017 PMCID: PMC8706222 DOI: 10.3390/ijms222413221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle accounts for almost 40% of the total adult human body mass. This tissue is essential for structural and mechanical functions such as posture, locomotion, and breathing, and it is endowed with an extraordinary ability to adapt to physiological changes associated with growth and physical exercise, as well as tissue damage. Moreover, skeletal muscle is the most age-sensitive tissue in mammals. Due to aging, but also to several diseases, muscle wasting occurs with a loss of muscle mass and functionality, resulting from disuse atrophy and defective muscle regeneration, associated with dysfunction of satellite cells, which are the cells responsible for maintaining and repairing adult muscle. The most established cell lines commonly used to study muscle homeostasis come from rodents, but there is a need to study skeletal muscle using human models, which, due to ethical implications, consist primarily of in vitro culture, which is the only alternative way to vertebrate model organisms. This review will survey in vitro 2D/3D models of human satellite cells to assess skeletal muscle biology for pre-clinical investigations and future directions.
Collapse
Affiliation(s)
- Cecilia Romagnoli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (C.R.); (T.I.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (C.R.); (T.I.)
| | - Maria Luisa Brandi
- F.I.R.M.O. Italian Foundation for the Research on Bone Diseases, Via Reginaldo Giuliani 195/A, 50141 Florence, Italy
- Correspondence:
| |
Collapse
|
18
|
Roux-Biejat P, Coazzoli M, Marrazzo P, Zecchini S, Di Renzo I, Prata C, Napoli A, Moscheni C, Giovarelli M, Barbalace MC, Catalani E, Bassi MT, De Palma C, Cervia D, Malaguti M, Hrelia S, Clementi E, Perrotta C. Acid Sphingomyelinase Controls Early Phases of Skeletal Muscle Regeneration by Shaping the Macrophage Phenotype. Cells 2021; 10:3028. [PMID: 34831250 PMCID: PMC8616363 DOI: 10.3390/cells10113028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle regeneration is a complex process involving crosstalk between immune cells and myogenic precursor cells, i.e., satellite cells. In this scenario, macrophage recruitment in damaged muscles is a mandatory step for tissue repair since pro-inflammatory M1 macrophages promote the activation of satellite cells, stimulating their proliferation and then, after switching into anti-inflammatory M2 macrophages, they prompt satellite cells' differentiation into myotubes and resolve inflammation. Here, we show that acid sphingomyelinase (ASMase), a key enzyme in sphingolipid metabolism, is activated after skeletal muscle injury induced in vivo by the injection of cardiotoxin. ASMase ablation shortens the early phases of skeletal muscle regeneration without affecting satellite cell behavior. Of interest, ASMase regulates the balance between M1 and M2 macrophages in the injured muscles so that the absence of the enzyme reduces inflammation. The analysis of macrophage populations indicates that these events depend on the altered polarization of M1 macrophages towards an M2 phenotype. Our results unravel a novel role of ASMase in regulating immune response during muscle regeneration/repair and suggest ASMase as a supplemental therapeutic target in conditions of redundant inflammation that impairs muscle recovery.
Collapse
Affiliation(s)
- Paulina Roux-Biejat
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Marco Coazzoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Pasquale Marrazzo
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Silvia Zecchini
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Ilaria Di Renzo
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, 40126 Bologna, Italy;
| | - Alessandra Napoli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Claudia Moscheni
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Matteo Giovarelli
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| | - Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Maria Teresa Bassi
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy;
| | - Clara De Palma
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, 20129 Milano, Italy;
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), Università degli Studi della Tuscia, 01100 Viterbo, Italy; (E.C.); (D.C.)
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, 47921 Rimini, Italy; (P.M.); (M.C.B.); (M.M.); (S.H.)
| | - Emilio Clementi
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
- Scientific Institute IRCCS “Eugenio Medea”, 23842 Bosisio Parini, Italy;
| | - Cristiana Perrotta
- Department of Biomedical and Clinical Sciences “Luigi Sacco” (DIBIC), Università degli Studi di Milano, 20157 Milano, Italy; (P.R.-B.); (M.C.); (S.Z.); (I.D.R.); (A.N.); (C.M.); (M.G.); (E.C.)
| |
Collapse
|
19
|
Macino M, Biferali B, Cipriano A, Ballarino M, Mozzetta C. Targeting the Expression of Long Noncoding RNAs in Murine Satellite Cells from Single Myofibers. Bio Protoc 2021; 11:e4209. [PMID: 34859124 DOI: 10.21769/bioprotoc.4209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/09/2021] [Accepted: 08/20/2021] [Indexed: 11/02/2022] Open
Abstract
LncRNAs have been recently implicated in the epigenetic control of muscle differentiation and their functional characterization has traditionally relied upon in vitro models of myogenic differentiation. However, the use of experimental paradigms to specifically target lncRNAs expression in muscle stem cells (MuSCs), also known as satellite cells, represents an important requisite to interrogate their function in more physiological contexts. Since isolation and culture of single myofibers preserves satellite cells within their physiological niche underneath the surrounding basal lamina, this procedure represents the optimal approach to follow satellite cell dynamics ex-vivo, such as activation from quiescence, expansion of committed progenitors, differentiation, and self-renewal. Here, we detail an optimized protocol to isolate viable single myofibers from the extensor digitorum longus (EDL) skeletal muscle of adult mice and to manipulate the expression of lncRNAs by antisense LNA GapmeRs-mediated knock-down (KD). Furthermore, we describe a method of EdU incorporation that, coupled to lncRNA KD and subsequent immunofluorescence analysis of proliferating, differentiating, and satellite cell-specific markers, permits the inference of lncRNAs function on muscle stem cells dynamics. Graphic abstract: Graphical representation of the single myofiber isolation method. Experimental workflow showing the main steps of the protocol procedure: EDL muscle harvesting from the mouse hindlimb; EDL digestion into single myofibers; transfection with antisense oligos and culture for 96h; immunofluorescence protocol and image outcome.
Collapse
Affiliation(s)
- Martina Macino
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) at Sapienza University of Rome, Rome, Italy.,Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Beatrice Biferali
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) at Sapienza University of Rome, Rome, Italy.,Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Andrea Cipriano
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Monica Ballarino
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Chiara Mozzetta
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) at Sapienza University of Rome, Rome, Italy
| |
Collapse
|
20
|
Sex differences in metabolic pathways are regulated by Pfkfb3 and Pdk4 expression in rodent muscle. Commun Biol 2021; 4:1264. [PMID: 34737380 PMCID: PMC8569015 DOI: 10.1038/s42003-021-02790-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 10/15/2021] [Indexed: 12/28/2022] Open
Abstract
Skeletal muscles display sexually dimorphic features. Biochemically, glycolysis and fatty acid β-oxidation occur preferentially in the muscles of males and females, respectively. However, the mechanisms of the selective utilization of these fuels remains elusive. Here, we obtain transcriptomes from quadriceps type IIB fibers of untreated, gonadectomized, and sex steroid-treated mice of both sexes. Analyses of the transcriptomes unveil two genes, Pfkfb3 (phosphofructokinase-2) and Pdk4 (pyruvate dehydrogenase kinase 4), that may function as switches between the two sexually dimorphic metabolic pathways. Interestingly, Pfkfb3 and Pdk4 show male-enriched and estradiol-enhanced expression, respectively. Moreover, the contribution of these genes to sexually dimorphic metabolism is demonstrated by knockdown studies with cultured type IIB muscle fibers. Considering that skeletal muscles as a whole are the largest energy-consuming organs, our results provide insights into energy metabolism in the two sexes, during the estrus cycle in women, and under pathological conditions involving skeletal muscles. Baba et al. analyzed the transcriptomes from quadriceps type IIB fibers of untreated, gonadectomized, and sex steroid-treated mice of both sexes and identified Pfkfb3 and Pdk4 as differentially regulated genes between males and diestrus females. The authors found that Pfkfb3 and Pdk4 may act as metabolic switches, showed male-enriched and estradiol-enhanced expression, respectively and contributed to sexually dimorphic metabolism.
Collapse
|
21
|
Microtubule-based transport is essential to distribute RNA and nascent protein in skeletal muscle. Nat Commun 2021; 12:6079. [PMID: 34707124 PMCID: PMC8551216 DOI: 10.1038/s41467-021-26383-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022] Open
Abstract
While the importance of RNA localization in highly differentiated cells is well appreciated, basic principles of RNA localization in skeletal muscle remain poorly characterized. Here, we develop a method to detect and quantify single molecule RNA localization patterns in skeletal myofibers, and uncover a critical role for directed transport of RNPs in muscle. We find that RNAs localize and are translated along sarcomere Z-disks, dispersing tens of microns from progenitor nuclei, regardless of encoded protein function. We find that directed transport along the lattice-like microtubule network of myofibers becomes essential to achieve this localization pattern as muscle development progresses; disruption of this network leads to extreme accumulation of RNPs and nascent protein around myonuclei. Our observations suggest that global active RNP transport may be required to distribute RNAs in highly differentiated cells and reveal fundamental mechanisms of gene regulation, with consequences for myopathies caused by perturbations to RNPs or microtubules.
Collapse
|
22
|
Schüler SC, Dumontier S, Rigaux J, Bentzinger CF. Visualization of the Skeletal Muscle Stem Cell Niche in Fiber Bundles. Curr Protoc 2021; 1:e263. [PMID: 34612611 PMCID: PMC9291523 DOI: 10.1002/cpz1.263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Skeletal muscle stem cells (MuSCs) reside in a complex niche composed of the muscle fiber plasma membrane and the laminin‐rich basal lamina surrounded by the microvasculature, as well as different supportive cell types such as fibro‐adipogenic progenitors residing in the interstitial extracellular matrix. Within the first few hours after tissue damage, MuSCs undergo cytoskeletal rearrangements and transcriptional changes that prime the cells for activation. Due to their time‐consuming nature, enzymatic methods for liberation of single muscle fibers with fully quiescent MuSCs are challenging. Moreover, during enzymatic digestion, important niche components including the microvasculature and the collagenous interstitial matrix are destroyed. Here, we provide a method for the visualization of MuSCs on muscle fibers in their intact niche. Our method relies on mechanical teasing of fiber bundles from fixed skeletal muscles. We demonstrate that teased muscle fiber bundles allow the investigator to capture a representative snapshot of the MuSC niche in skeletal muscle, and outline how stem cell morphology and different microenvironmental components can be visualized. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Isolation of fiber bundles Basic Protocol 2: Immunofluorescence staining of MuSCs on fiber bundles Support Protocol: Preparation of Sylgard dishes
Collapse
Affiliation(s)
- Svenja C Schüler
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Simon Dumontier
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jonathan Rigaux
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - C Florian Bentzinger
- Département de Pharmacologie-Physiologie, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
23
|
Luan P, D'Amico D, Andreux PA, Laurila PP, Wohlwend M, Li H, Imamura de Lima T, Place N, Rinsch C, Zanou N, Auwerx J. Urolithin A improves muscle function by inducing mitophagy in muscular dystrophy. Sci Transl Med 2021; 13:13/588/eabb0319. [PMID: 33827972 DOI: 10.1126/scitranslmed.abb0319] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 11/25/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy, and despite advances in genetic and pharmacological disease-modifying treatments, its management remains a major challenge. Mitochondrial dysfunction contributes to DMD, yet the mechanisms by which this occurs remain elusive. Our data in experimental models and patients with DMD show that reduced expression of genes involved in mitochondrial autophagy, or mitophagy, contributes to mitochondrial dysfunction. Mitophagy markers were reduced in skeletal muscle and in muscle stem cells (MuSCs) of a mouse model of DMD. Administration of the mitophagy activator urolithin A (UA) rescued mitophagy in DMD worms and mice and in primary myoblasts from patients with DMD, increased skeletal muscle respiratory capacity, and improved MuSCs' regenerative ability, resulting in the recovery of muscle function and increased survival in DMD mouse models. These data indicate that restoration of mitophagy alleviates symptoms of DMD and suggest that UA may have potential therapeutic applications for muscular dystrophies.
Collapse
Affiliation(s)
- Peiling Luan
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Davide D'Amico
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.,Amazentis SA, Ecole Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, 1015 Lausanne, Switzerland
| | - Pénélope A Andreux
- Amazentis SA, Ecole Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, 1015 Lausanne, Switzerland
| | - Pirkka-Pekka Laurila
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Martin Wohlwend
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Hao Li
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, Quartier UNIL-Centre, Faculty of Biology-Medicine, University of Lausanne, Bâtiment Synathlon, 1015 Lausanne, Switzerland
| | - Chris Rinsch
- Amazentis SA, Ecole Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, 1015 Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, Quartier UNIL-Centre, Faculty of Biology-Medicine, University of Lausanne, Bâtiment Synathlon, 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
24
|
Rice JC, Weekley BH, Kanholm T, Chen Z, Lee S, Fernandez DJ, Abrahamson R, Castaldi A, Borok Z, Dynlacht BD, An W. MMP-2 is a novel histone H3 N-terminal protease necessary for myogenic gene activation. Epigenetics Chromatin 2021; 14:23. [PMID: 34001241 PMCID: PMC8130154 DOI: 10.1186/s13072-021-00398-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/06/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Selective proteolysis of the histone H3 N-terminal tail (H3NT) is frequently observed during eukaryotic development, generating a cleaved histone H3 (H3cl) product within a small, but significant, portion of the genome. Although increasing evidence supports a regulatory role for H3NT proteolysis in gene activation, the nuclear H3NT proteases and the biological significance of H3NT proteolysis remain largely unknown. RESULTS In this study, established cell models of skeletal myogenesis were leveraged to investigate H3NT proteolysis. These cells displayed a rapid and progressive accumulation of a single H3cl product within chromatin during myoblast differentiation. Using conventional approaches, we discovered that the canonical extracellular matrix (ECM) protease, matrix metalloproteinase 2 (MMP-2), is the principal H3NT protease of myoblast differentiation that cleaves H3 between K18-Q19. Gelatin zymography demonstrated progressive increases in nuclear MMP-2 activity, concomitant with H3cl accumulation, during myoblast differentiation. RNAi-mediated depletion of MMP-2 impaired H3NT proteolysis and resulted in defective myogenic gene activation and myoblast differentiation. Supplementation of MMP-2 ECM activity in MMP-2-depleted cells was insufficient to rescue defective H3NT proteolysis and myogenic gene activation. CONCLUSIONS This study revealed that MMP-2 is a novel H3NT protease and the principal H3NT protease of myoblast differentiation. The results indicate that myogenic signaling induces MMP-2-dependent H3NT proteolysis at early stages of myoblast differentiation. Importantly, the results support the necessity of nuclear MMP-2 H3NT protease activity, independent of MMP-2 activity in the ECM, for myogenic gene activation and proficient myoblast differentiation.
Collapse
Affiliation(s)
- Judd C Rice
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, 1450 Biggy Street, HNRT 6506, Los Angeles, CA, 90033, USA.
| | - Benjamin H Weekley
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, 1450 Biggy Street, HNRT 6506, Los Angeles, CA, 90033, USA
| | - Tomas Kanholm
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, 1450 Biggy Street, HNRT 6506, Los Angeles, CA, 90033, USA
| | - Zhihui Chen
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, 1450 Biggy Street, HNRT 6506, Los Angeles, CA, 90033, USA
| | - Sunyoung Lee
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, 1450 Biggy Street, HNRT 6506, Los Angeles, CA, 90033, USA
| | - Daniel J Fernandez
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, Los Angeles, CA, 90033, USA
| | - Rachel Abrahamson
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, 1450 Biggy Street, HNRT 6506, Los Angeles, CA, 90033, USA
| | - Alessandra Castaldi
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Zea Borok
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, 1450 Biggy Street, HNRT 6506, Los Angeles, CA, 90033, USA
- Hastings Center for Pulmonary Research and Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Brian D Dynlacht
- Department of Pathology and Cancer Institute, NYU School of Medicine,, New York, NY, 10016, USA
| | - Woojin An
- Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, University of Southern California Keck School of Medicine, 1450 Biggy Street, HNRT 6506, Los Angeles, CA, 90033, USA
| |
Collapse
|
25
|
Proietti D, Giordani L, De Bardi M, D’Ercole C, Lozanoska-Ochser B, Amadio S, Volonté C, Marinelli S, Muchir A, Bouché M, Borsellino G, Sacco A, Puri PL, Madaro L. Activation of skeletal muscle-resident glial cells upon nerve injury. JCI Insight 2021; 6:143469. [PMID: 33661767 PMCID: PMC8119188 DOI: 10.1172/jci.insight.143469] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/25/2021] [Indexed: 01/06/2023] Open
Abstract
Here, we report on the identification of Itga7-expressing muscle-resident glial cells activated by loss of neuromuscular junction (NMJ) integrity. Gene expression analysis at the bulk and single-cell level revealed that these cells are distinct from Itga7-expressing muscle satellite cells. We show that a selective activation and expansion of Itga7+ glial cells occur in response to muscle nerve lesion. Upon activation, muscle glial-derived progenies expressed neurotrophic genes, including nerve growth factor receptor, which enables their isolation by FACS. We show that activated muscle glial cells also expressed genes potentially implicated in extracellular matrix remodeling at NMJs. We found that tenascin C, which was highly expressed by muscle glial cells, activated upon nerve injury and preferentially localized to NMJ. Interestingly, we observed that the activation of muscle glial cells by acute nerve injury was reversible upon NMJ repair. By contrast, in a mouse model of ALS, in which NMJ degeneration is progressive, muscle glial cells steadily increased over the course of the disease. However, they exhibited an impaired neurotrophic activity, suggesting that pathogenic activation of glial cells may be implicated in ALS progression.
Collapse
Affiliation(s)
- Daisy Proietti
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | - Lorenzo Giordani
- Sorbonne Université, INSERM UMRS 974, Association Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | | | - Chiara D’Ercole
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | - Biliana Lozanoska-Ochser
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | | | - Cinzia Volonté
- IRCCS Fondazione Santa Lucia, Rome, Italy
- CNR, National Research Council, Institute for Systems Analysis and Computer Science, Rome, Italy
| | - Sara Marinelli
- CNR, National Research Council, Institute of Biochemistry and Cell Biology, Monterotondo Scalo, Rome, Italy
| | - Antoine Muchir
- Sorbonne Université, INSERM UMRS 974, Association Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Marina Bouché
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| | | | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Luca Madaro
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, University of Rome “la Sapienza”, Rome, Italy
| |
Collapse
|
26
|
Furuichi Y, Kawabata Y, Aoki M, Mita Y, Fujii NL, Manabe Y. Excess Glucose Impedes the Proliferation of Skeletal Muscle Satellite Cells Under Adherent Culture Conditions. Front Cell Dev Biol 2021; 9:640399. [PMID: 33732705 PMCID: PMC7957019 DOI: 10.3389/fcell.2021.640399] [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: 12/11/2020] [Accepted: 02/04/2021] [Indexed: 01/08/2023] Open
Abstract
Glucose is a major energy source consumed by proliferating mammalian cells. Therefore, in general, proliferating cells have the preference of high glucose contents in extracellular environment. Here, we showed that high glucose concentrations impede the proliferation of satellite cells, which are muscle-specific stem cells, under adherent culture conditions. We found that the proliferation activity of satellite cells was higher in glucose-free DMEM growth medium (low-glucose medium with a glucose concentration of 2 mM) than in standard glucose DMEM (high-glucose medium with a glucose concentration of 19 mM). Satellite cells cultured in the high-glucose medium showed a decreased population of reserve cells, identified by staining for Pax7 expression, suggesting that glucose concentration affects cell fate determination. In conclusion, glucose is a factor that decides the cell fate of skeletal muscle-specific stem cells. Due to this unique feature of satellite cells, hyperglycemia may negatively affect the regenerative capability of skeletal muscle myofibers and thus facilitate sarcopenia.
Collapse
Affiliation(s)
- Yasuro Furuichi
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Yuki Kawabata
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Miho Aoki
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Yoshitaka Mita
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Nobuharu L Fujii
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| | - Yasuko Manabe
- Department of Health Promotion Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
27
|
Khodabukus A. Tissue-Engineered Skeletal Muscle Models to Study Muscle Function, Plasticity, and Disease. Front Physiol 2021; 12:619710. [PMID: 33716768 PMCID: PMC7952620 DOI: 10.3389/fphys.2021.619710] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle possesses remarkable plasticity that permits functional adaptations to a wide range of signals such as motor input, exercise, and disease. Small animal models have been pivotal in elucidating the molecular mechanisms regulating skeletal muscle adaptation and plasticity. However, these small animal models fail to accurately model human muscle disease resulting in poor clinical success of therapies. Here, we review the potential of in vitro three-dimensional tissue-engineered skeletal muscle models to study muscle function, plasticity, and disease. First, we discuss the generation and function of in vitro skeletal muscle models. We then discuss the genetic, neural, and hormonal factors regulating skeletal muscle fiber-type in vivo and the ability of current in vitro models to study muscle fiber-type regulation. We also evaluate the potential of these systems to be utilized in a patient-specific manner to accurately model and gain novel insights into diseases such as Duchenne muscular dystrophy (DMD) and volumetric muscle loss. We conclude with a discussion on future developments required for tissue-engineered skeletal muscle models to become more mature, biomimetic, and widely utilized for studying muscle physiology, disease, and clinical use.
Collapse
Affiliation(s)
- Alastair Khodabukus
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
28
|
Su R, Wang B, Zhang M, Luo Y, Wang D, Zhao L, Su L, Duan Y, Faucitano L, Jin Y. Effects of energy supplements on the differentiation of skeletal muscle satellite cells. Food Sci Nutr 2021; 9:357-366. [PMID: 33473298 PMCID: PMC7802567 DOI: 10.1002/fsn3.2001] [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: 07/16/2020] [Revised: 10/19/2020] [Accepted: 10/25/2020] [Indexed: 01/14/2023] Open
Abstract
To investigate the effects of the activator of AMPK and high glucose on the differentiation of mouse SMSCs, primary SMSCs were isolated from mouse extensor digitorum longus muscle and grown to near confluence (80%). Postconfluent cells were cultured in a growth medium with different inductors: AICAR, glucose, and AICAR mixed with glucose. The specific protein expressions of SMSCs, myoblasts, adipocytes, and brown adipocytes were analyzed on days 0, 3, 5, 7, and 10. The results showed treatment with AICAR in SMSCs markedly activated AMPK phosphorylation (p < .05) and increased protein expression of Pax7 and MyoD (p < .05), high concentrations of intracellular glucose upregulated UCP-1 protein expression and enhanced lipid accumulation (p < .05), the cowork of AICAR and glucose affected a decrease on MyoD, PPARg, and UCP-1 expression (p < .05) and an increase on Pax7. The present study indicated that the certain energy supplements influence the direction of SMSC differentiation which may contribution on the structure of muscle and meat quality, sequentially.
Collapse
Affiliation(s)
- Rina Su
- College of Food Science and EngineeringInner Mongolia Agriculture UniversityHohhotChina
- Inner Mongolia Vocational college of Chemical EngineeringHohhotChina
| | - Bohui Wang
- College of Food Science and EngineeringInner Mongolia Agriculture UniversityHohhotChina
| | - Min Zhang
- College of Food Science and EngineeringInner Mongolia Agriculture UniversityHohhotChina
| | - Yulong Luo
- College of Food Science and EngineeringInner Mongolia Agriculture UniversityHohhotChina
| | - Debao Wang
- College of Food Science and EngineeringInner Mongolia Agriculture UniversityHohhotChina
| | - Lihua Zhao
- College of Food Science and EngineeringInner Mongolia Agriculture UniversityHohhotChina
| | - Lin Su
- College of Food Science and EngineeringInner Mongolia Agriculture UniversityHohhotChina
| | - Yan Duan
- College of Food Science and EngineeringInner Mongolia Agriculture UniversityHohhotChina
| | - Luigi Faucitano
- Agriculture and Agri‐Food CanadaSherbrooke Research and Development CentreSherbrookeQCCanada
| | - Ye Jin
- College of Food Science and EngineeringInner Mongolia Agriculture UniversityHohhotChina
| |
Collapse
|
29
|
Lv W, Jin J, Xu Z, Luo H, Guo Y, Wang X, Wang S, Zhang J, Zuo H, Bai W, Peng Y, Tang J, Zhao S, Zuo B. lncMGPF is a novel positive regulator of muscle growth and regeneration. J Cachexia Sarcopenia Muscle 2020; 11:1723-1746. [PMID: 32954689 PMCID: PMC7749533 DOI: 10.1002/jcsm.12623] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 07/24/2020] [Accepted: 08/23/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) play critical regulatory roles in diverse biological processes and diseases. While a large number of lncRNAs have been identified in skeletal muscles until now, their function and underlying mechanisms in skeletal myogenesis remain largely unclear. METHODS We characterized a novel functional lncRNA designated lncMGPF (lncRNA muscle growth promoting factor) using RACE, Northern blot, fluorescence in situ hybridization and quantitative real-time PCR. Its function was determined by gene overexpression, interference, and knockout experiments in C2C12 myoblasts, myogenic progenitor cells, and an animal model. The molecular mechanism by which lncMGPF regulates muscle differentiation was mainly examined by cotransfection experiments, luciferase reporter assay, RNA immunoprecipitation, RNA pull-down, and RNA stability analyses. RESULTS We report that lncMGPF, which is highly expressed in muscles and positively regulated by myoblast determination factor (MyoD), promotes myogenic differentiation of muscle cells in vivo and in vitro. lncMGPF knockout in mice substantially decreases growth rate, reduces muscle mass, and impairs muscle regeneration. Overexpression of lncMGPF in muscles can rescue the muscle phenotype of knockout mice and promote muscle growth of wild-type mice. Mechanistically, lncMGPF promotes muscle differentiation by acting as a molecular sponge of miR-135a-5p and thus increasing the expression of myocyte enhancer factor 2C (MEF2C), as well as by enhancing human antigen R-mediated messenger RNA stabilization of myogenic regulatory genes such as MyoD and myogenin (MyoG). We confirm that pig lncRNA AK394747 and human lncRNA MT510647 are homologous to mouse lncMGPF, with conserved function and mechanism during myogenesis. CONCLUSIONS Our data reveal that lncMGPF is a novel positive regulator of myogenic differentiation, muscle growth and regeneration in mice, pigs, and humans.
Collapse
Affiliation(s)
- Wei Lv
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jianjun Jin
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zaiyan Xu
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Hongmei Luo
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yubo Guo
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaojing Wang
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Shanshan Wang
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jiali Zhang
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hao Zuo
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wei Bai
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yaxing Peng
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Junming Tang
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medicine Science, Hubei University of Medicine, Shiyan, China
| | - Shuhong Zhao
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| |
Collapse
|
30
|
Hall A, Fontelonga T, Wright A, Bugda Gwilt K, Widrick J, Pasut A, Villa F, Miranti CK, Gibbs D, Jiang E, Meng H, Lawlor MW, Gussoni E. Tetraspanin CD82 is necessary for muscle stem cell activation and supports dystrophic muscle function. Skelet Muscle 2020; 10:34. [PMID: 33243288 PMCID: PMC7693590 DOI: 10.1186/s13395-020-00252-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Background Tetraspanins are a family of proteins known to assemble protein complexes at the cell membrane. They are thought to play diverse cellular functions in tissues by modifying protein-binding partners, thus bringing complexity and diversity in their regulatory networks. Previously, we identified the tetraspanin KAI/CD82 as a prospective marker for human muscle stem cells. CD82 expression appeared decreased in human Duchenne muscular dystrophy (DMD) muscle, suggesting a functional link to muscular dystrophy, yet whether this decrease is a consequence of dystrophic pathology or a compensatory mechanism in an attempt to rescue muscle from degeneration is currently unknown. Methods We studied the consequences of loss of CD82 expression in normal and dystrophic skeletal muscle and examined the dysregulation of downstream functions in mice aged up to 1 year. Results Expression of CD82 is important to sustain satellite cell activation, as in its absence there is decreased cell proliferation and less efficient repair of injured muscle. Loss of CD82 in dystrophic muscle leads to a worsened phenotype compared to control dystrophic mice, with decreased pulmonary function, myofiber size, and muscle strength. Mechanistically, decreased myofiber size in CD82−/− dystrophic mice is not due to altered PTEN/AKT signaling, although increased phosphorylation of mTOR at Ser2448 was observed. Conclusion Basal CD82 expression is important to dystrophic muscle, as its loss leads to significantly weakened myofibers and impaired muscle function, accompanied by decreased satellite cell activity that is unable to protect and repair myofiber damage. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-020-00252-3.
Collapse
Affiliation(s)
- Arielle Hall
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Tatiana Fontelonga
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Alec Wright
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Katlynn Bugda Gwilt
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Jeffrey Widrick
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Alessandra Pasut
- Laboratory of Angiogenesis and Vascular metabolism, Center for Cancer Biology, VIB and KU Leuven, 3000, Leuven, Belgium
| | - Francesco Villa
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cynthia K Miranti
- Department of Cellular and Molecular Medicine, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Devin Gibbs
- Molecular Biology Institute, UCLA, Los Angeles, CA, 90095, USA
| | - Evan Jiang
- The University of Pennsylvania, College of Arts and Sciences, Philadelphia, PA, 19104, USA
| | - Hui Meng
- Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Michael W Lawlor
- Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Emanuela Gussoni
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, 02115, USA. .,The Stem Cell Program at Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
31
|
Buchanan SM, Price FD, Castiglioni A, Gee AW, Schneider J, Matyas MN, Hayhurst M, Tabebordbar M, Wagers AJ, Rubin LL. Pro-myogenic small molecules revealed by a chemical screen on primary muscle stem cells. Skelet Muscle 2020; 10:28. [PMID: 33036659 PMCID: PMC7547525 DOI: 10.1186/s13395-020-00248-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/14/2020] [Indexed: 11/10/2022] Open
Abstract
Satellite cells are the canonical muscle stem cells that regenerate damaged skeletal muscle. Loss of function of these cells has been linked to reduced muscle repair capacity and compromised muscle health in acute muscle injury and congenital neuromuscular diseases. To identify new pathways that can prevent loss of skeletal muscle function or enhance regenerative potential, we established an imaging-based screen capable of identifying small molecules that promote the expansion of freshly isolated satellite cells. We found several classes of receptor tyrosine kinase (RTK) inhibitors that increased freshly isolated satellite cell numbers in vitro. Further exploration of one of these compounds, the RTK inhibitor CEP-701 (also known as lestaurtinib), revealed potent activity on mouse satellite cells both in vitro and in vivo. This expansion potential was not seen upon exposure of proliferating committed myoblasts or non-myogenic fibroblasts to CEP-701. When delivered subcutaneously to acutely injured animals, CEP-701 increased both the total number of satellite cells and the rate of muscle repair, as revealed by an increased cross-sectional area of regenerating fibers. Moreover, freshly isolated satellite cells expanded ex vivo in the presence of CEP-701 displayed enhanced muscle engraftment potential upon in vivo transplantation. We provide compelling evidence that certain RTKs, and in particular RET, regulate satellite cell expansion during muscle regeneration. This study demonstrates the power of small molecule screens of even rare adult stem cell populations for identifying stem cell-targeting compounds with therapeutic potential.
Collapse
Affiliation(s)
- Sean M Buchanan
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Feodor D Price
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Alessandra Castiglioni
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA.,Cancer Immunology Department, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Amanda Wagner Gee
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Joel Schneider
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Mark N Matyas
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Monica Hayhurst
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Mohammadsharif Tabebordbar
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Amy J Wagers
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Lee L Rubin
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA.
| |
Collapse
|
32
|
Xu D, Zhao L, Jiang J, Li S, Sun Z, Huang X, Li C, Wang T, Sun L, Li X, Jiang Z, Zhang L. A potential therapeutic effect of catalpol in Duchenne muscular dystrophy revealed by binding with TAK1. J Cachexia Sarcopenia Muscle 2020; 11:1306-1320. [PMID: 32869445 PMCID: PMC7567147 DOI: 10.1002/jcsm.12581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/29/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a progressive muscle disease caused by the loss of dystrophin, which results in inflammation, fibrosis, and the inhibition of myoblast differentiation in skeletal muscle. Catalpol, an iridoid glycoside, improves skeletal muscle function by enhancing myogenesis; it has potential to treat DMD. We demonstrate the positive effects of catalpol in dystrophic skeletal muscle. METHODS mdx (loss of dystrophin) mice (n = 18 per group) were treated with catalpol (200 mg/kg) for six consecutive weeks. Serum analysis, skeletal muscle performance and histology, muscle contractile function, and gene and protein expression were performed. Molecular docking and ligand-target interactions, RNA interference, immunofluorescence, and plasmids transfection were utilized to explore the protective mechanism in DMD by which catalpol binding with transforming growth factor-β-activated kinase 1 (TAK1) in skeletal muscle. RESULTS Six weeks of catalpol treatment improved whole-body muscle health in mdx mice, which was characterized by reduced plasma creatine kinase (n = 18, -35.1%, P < 0.05) and lactic dehydrogenase (n = 18, -10.3%, P < 0.05) activity. These effects were accompanied by enhanced grip strength (n = 18, +25.4%, P < 0.05) and reduced fibrosis (n = 18, -29.0% for hydroxyproline content, P < 0.05). Moreover, catalpol treatment protected against muscle fatigue and promoted muscle recovery in the tibialis anterior (TA) and diaphragm (DIA) muscles (n = 6, +69.8%, P < 0.05 and + 74.8%, P < 0.001, respectively), which was accompanied by enhanced differentiation in primary myoblasts from DMD patients (n = 6, male, mean age: 4.7 ± 1.9 years) and mdx mice. In addition, catalpol eliminated p-TAK1 overexpression in mdx mice (n = 12, -21.3%, P < 0.05) and primary myoblasts. The catalpol-induced reduction in fibrosis and increased myoblast differentiation resulted from the inhibition of TAK1 phosphorylation, leading to reduced myoblast trans-differentiation into myofibroblasts. Catalpol inhibited the phosphorylation of TAK1 by binding to TAK1, possibly at Asp-206, Thr-208, Asn-211, Glu-297, Lys-294, and Tyr-293. CONCLUSIONS Our findings show that catalpol and TAK1 inhibitors substantially improve whole-body muscle health and the function of dystrophic skeletal muscles and may provide a novel therapy for DMD.
Collapse
Affiliation(s)
- Dengqiu Xu
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Lei Zhao
- Department of NeurologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Jingwei Jiang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Sijia Li
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Zeren Sun
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Xiaofei Huang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Chunjie Li
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Tao Wang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Jiangsu Center for Pharmacodynamics Research and EvaluationChina Pharmaceutical UniversityNanjingChina
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Xihua Li
- Department of NeurologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Key Laboratory of Drug Quality Control and PharmacovigilanceChina Pharmaceutical UniversityNanjingChina
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Center for Drug Research and DevelopmentGuangdong Pharmaceutical UniversityGuangzhouChina
- Key Laboratory of Drug Quality Control and PharmacovigilanceChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
33
|
Stange K, Ahrens HE, von Maltzahn J, Röntgen M. Isolation and ex vivo cultivation of single myofibers from porcine muscle. In Vitro Cell Dev Biol Anim 2020; 56:585-592. [PMID: 32964376 PMCID: PMC7532130 DOI: 10.1007/s11626-020-00492-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/06/2020] [Indexed: 11/27/2022]
Abstract
The isolation and cultivation of intact, single myofibers presents a superior approach for studying myogenic cells in their native position. The cells’ characteristics remain more similar to muscle tissue than in cell culture. Nevertheless, no routinely used method in higher vertebrates exists. Therefore, we aimed at establishing the isolation and cultivation of single myofibers from porcine muscle. For the first time, we implemented the isolation of intact myofibers from porcine fibularis tertius muscle by enzymatic digestion and their subsequent cultivation under floating conditions. Confocal microscopy showed intact myofibrill structures in isolated myofibers. Myogenic cells were able to proliferate at their parent myofiber as shown by the increase of myonuclear number during culture. Additionally, the described method can be used to investigate myogenic cells migrated from isolated myofibers. These cells expressed myogenic markers and were able to differentiate. In the future, our method can be used for genetic manipulation of cells at myofibers, investigation of growth factors or pharmacological substances, and determination of interactions between myofibers and associated cells. Working with isolated myofibers has the potential to bridge conventional cell culture and animal experiments. Adapting the method to porcine muscle allows for application possibilities in veterinary medicine as well as in biomedical research, which cannot be addressed in rodent model systems.
Collapse
Affiliation(s)
- Katja Stange
- Institute of Muscle Biology and Growth, Growth and Development Unit, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Hellen Elisa Ahrens
- Research Group Stem Cells in Regeneration of Skeletal Muscle, Leibniz Institute on Aging, 07745, Jena, Germany
| | - Julia von Maltzahn
- Research Group Stem Cells in Regeneration of Skeletal Muscle, Leibniz Institute on Aging, 07745, Jena, Germany
| | - Monika Röntgen
- Institute of Muscle Biology and Growth, Growth and Development Unit, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
34
|
Splicing Factor SRSF1 Is Essential for Satellite Cell Proliferation and Postnatal Maturation of Neuromuscular Junctions in Mice. Stem Cell Reports 2020; 15:941-954. [PMID: 32888503 PMCID: PMC7561493 DOI: 10.1016/j.stemcr.2020.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 02/07/2023] Open
Abstract
Satellite cells are main muscle stem cells that could provide myonuclei for myofiber growth and synaptic-specific gene expression during the early postnatal development. Here, we observed that splicing factor SRSF1 is highly expressed in myoblasts and its expression is closely related with satellite cell activation and proliferation. By genetic deletion of SRSF1 in myogenic progenitors, we found that SRSF1 is critical for satellite cell proliferation in vitro and in vivo. Most notably we also observed that SRSF1 is required for the functional neuromuscular junction (NMJ) formation, as SRSF1-deficient mice fail to form mature pretzel-like NMJs, which leads to muscle weakness and premature death in mice. Finally, we demonstrated that SRSF1 contributes to muscle innervation and muscle development likely by regulating a restricted set of tissue-specific alternative splicing events. Thus, our data define a unique role for SRSF1 in postnatal skeletal muscle growth and function in mice. SRSF1 is highly expressed in activated satellite cells Loss of SRSF1 dramatically impairs satellite cell proliferation in vitro and in vivo SRSF1 is also required for the functional neuromuscular junction formation in mice SRSF1-deficient mice display muscle weakness and die prematurely
Collapse
|
35
|
Liu J, Huang ZP, Nie M, Wang G, Silva WJ, Yang Q, Freire PP, Hu X, Chen H, Deng Z, Pu WT, Wang DZ. Regulation of myonuclear positioning and muscle function by the skeletal muscle-specific CIP protein. Proc Natl Acad Sci U S A 2020; 117:19254-19265. [PMID: 32719146 PMCID: PMC7430979 DOI: 10.1073/pnas.1922911117] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The appropriate arrangement of myonuclei within skeletal muscle myofibers is of critical importance for normal muscle function, and improper myonuclear localization has been linked to a variety of skeletal muscle diseases, such as centronuclear myopathy and muscular dystrophies. However, the molecules that govern myonuclear positioning remain elusive. Here, we report that skeletal muscle-specific CIP (sk-CIP) is a regulator of nuclear positioning. Genetic deletion of sk-CIP in mice results in misalignment of myonuclei along the myofibers and at specialized structures such as neuromuscular junctions (NMJs) and myotendinous junctions (MTJs) in vivo, impairing myonuclear positioning after muscle regeneration, leading to severe muscle dystrophy in mdx mice, a mouse model of Duchenne muscular dystrophy. sk-CIP is localized to the centrosome in myoblasts and relocates to the outer nuclear envelope in myotubes upon differentiation. Mechanistically, we found that sk-CIP interacts with the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex and the centriole Microtubule Organizing Center (MTOC) proteins to coordinately modulate myonuclear positioning and alignment. These findings indicate that sk-CIP may function as a muscle-specific anchoring protein to regulate nuclear position in multinucleated muscle cells.
Collapse
MESH Headings
- Animals
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Co-Repressor Proteins
- Humans
- Mice
- Mice, Inbred mdx
- Mice, Knockout
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/physiopathology
- Myoblasts/metabolism
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/metabolism
- Myopathies, Structural, Congenital/physiopathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Organ Specificity
Collapse
Affiliation(s)
- Jianming Liu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Zhan-Peng Huang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Center for Translational Medicine, National Health Commission (NHC) Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510275, China
| | - Mao Nie
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Gang Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - William J Silva
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Laboratório de Biologia Celular e Molecular do Músculo Estriado, University of São Paulo, CEP 05508-000 Cidade Universitária, São Paulo, Brazil
| | - Qiumei Yang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Animal Sciences, Sichuan Agriculture University, Chengdu, 611130, China
| | - Paula P Freire
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Morphology, Institute of Biosciences, São Paulo State University, CEP 18618-000, Botucatu, São Paulo, Brazil
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Huaqun Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Department of Biology, Nanjing Normal University, Nanjing, 225300, China
| | - Zhongliang Deng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115;
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| |
Collapse
|
36
|
Wu J, Weisshaar N, Hotz-Wagenblatt A, Madi A, Ma S, Mieg A, Hering M, Mohr K, Schlimbach T, Borgers H, Cui G. Skeletal muscle antagonizes antiviral CD8 + T cell exhaustion. SCIENCE ADVANCES 2020; 6:eaba3458. [PMID: 32582853 PMCID: PMC7292629 DOI: 10.1126/sciadv.aba3458] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/28/2020] [Indexed: 05/05/2023]
Abstract
CD8+ T cells become functionally impaired or "exhausted" in chronic infections, accompanied by unwanted body weight reduction and muscle mass loss. Whether muscle regulates T cell exhaustion remains incompletely understood. We report that mouse skeletal muscle increased interleukin (IL)-15 production during LCMV clone 13 chronic infection. Muscle-specific ablation of Il15 enhanced the CD8+ T cell exhaustion phenotype. Muscle-derived IL-15 was required to maintain a population of CD8+CD103+ muscle-infiltrating lymphocytes (MILs). MILs resided in a less inflamed microenvironment, expressed more T cell factor 1 (Tcf1), and had higher proliferative potential than splenic T cells. MILs differentiated into functional effector T cells after reentering lymphoid tissues. Increasing muscle mass via muscle-specific inhibition of TGFβ signaling enhanced IL-15 production and antiviral CD8+ T cell responses. We conclude that skeletal muscle antagonizes T cell exhaustion by protecting T cell proliferative potential from inflammation and replenishing the effector T cell progeny pool in lymphoid organs.
Collapse
Affiliation(s)
- Jingxia Wu
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Nina Weisshaar
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Agnes Hotz-Wagenblatt
- Core Facility Omics IT and Data Management, DKFZ, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Alaa Madi
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Sicong Ma
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Alessa Mieg
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Marvin Hering
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Kerstin Mohr
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Tilo Schlimbach
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Helena Borgers
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Guoliang Cui
- T Cell Metabolism Group (D140), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Corresponding author.
| |
Collapse
|
37
|
Lee CY, Petkova M, Morales-Gonzalez S, Gimber N, Schmoranzer J, Meisel A, Böhmerle W, Stenzel W, Schuelke M, Schwarz JM. A spontaneous missense mutation in the chromodomain helicase DNA-binding protein 8 (CHD8) gene: a novel association with congenital myasthenic syndrome. Neuropathol Appl Neurobiol 2020; 46:588-601. [PMID: 32267004 DOI: 10.1111/nan.12617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 03/23/2020] [Indexed: 12/13/2022]
Abstract
AIMS Congenital myasthenic syndromes (CMS) are characterized by muscle weakness, ptosis and episodic apnoea. Mutations affect integral protein components of the neuromuscular junction (NMJ). Here we searched for the genetic basis of CMS in female monozygotic twins. METHODS We employed whole-exome sequencing for mutation detection and Sanger sequencing for segregation analysis. Immunohistology was done with antibodies against CHD8, rapsyn, β-catenin (βCAT) and golgin on fi-bro-blasts, human and mouse muscle. We recorded superresolution images of the NMJ using 3D-structured illumination microscopy. RESULTS We discovered a spontaneous missense mutation in CHD8 [chr14:g.21,884,051G>A, GRCh37.p11 | c.1732C>T, NM_00117062 | p.(R578C)], the gene encoding chromodomain helicase DNA-binding protein 8. This is the first missense mutation affecting Duplin, the short 110 kDa isoform of CHD8. It is known that CHD8/Duplin negatively regulates βCAT signalling in the WNT pathway and plays a role in chromatin remodelling. Inactivating CHD8 mutations are associated with autism spectrum disorder and intellectual disability in combination with facial dysmorphism, overgrowth and macrocephalus. No muscle-specific phenotype has been reported to date. Co-immunostaining with rapsyn on human and mouse muscle revealed a strong presence of CHD8 at the NMJ being located towards the sarcoplasmic side of the rapsyn cluster, where it co-localizes with βCAT. CONCLUSION We hypothesize CHD8 to have a role in the maintenance of the structural integrity and function of the NMJ. Both patients benefited from treatment with 3,4-diaminopyridine, a reversible blocker of voltage-gated potassium channels at the nerve terminal that prolongs the action potential and increases acetylcholine release.
Collapse
Affiliation(s)
- C Y Lee
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - M Petkova
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - S Morales-Gonzalez
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - N Gimber
- Advanced Medical Bioimaging Core Facility (AMBIO), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - J Schmoranzer
- Advanced Medical Bioimaging Core Facility (AMBIO), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - A Meisel
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - W Böhmerle
- Department of Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - W Stenzel
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - M Schuelke
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - J M Schwarz
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Neuropediatrics, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
38
|
Benedetti A, Fiore PF, Madaro L, Lozanoska-Ochser B, Bouché M. Targeting PKCθ Promotes Satellite Cell Self-Renewal. Int J Mol Sci 2020; 21:ijms21072419. [PMID: 32244482 PMCID: PMC7177808 DOI: 10.3390/ijms21072419] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 11/16/2022] Open
Abstract
Skeletal muscle regeneration following injury depends on the ability of satellite cells (SCs) to proliferate, self-renew, and eventually differentiate. The factors that regulate the process of self-renewal are poorly understood. In this study we examined the role of PKCθ in SC self-renewal and differentiation. We show that PKCθ is expressed in SCs, and its active form is localized to the chromosomes, centrosomes, and midbody during mitosis. Lack of PKCθ promotes SC symmetric self-renewal division by regulating Pard3 polarity protein localization, without affecting the overall proliferation rate. Genetic ablation of PKCθ or its pharmacological inhibition in vivo did not affect SC number in healthy muscle. By contrast, after induction of muscle injury, lack or inhibition of PKCθ resulted in a significant expansion of the quiescent SC pool. Finally, we show that lack of PKCθ does not alter the inflammatory milieu after acute injury in muscle, suggesting that the enhanced self-renewal ability of SCs in PKCθ-/- mice is not due to an alteration in the inflammatory milieu. Together, these results suggest that PKCθ plays an important role in SC self-renewal by stimulating their expansion through symmetric division, and it may represent a promising target to manipulate satellite cell self-renewal in pathological conditions.
Collapse
|
39
|
Blackburn DM, Lazure F, Soleimani VD. RNA Sequencing of Single Myofibers from Mus musculus. Bio Protoc 2020; 10:e3525. [PMID: 33654749 DOI: 10.21769/bioprotoc.3525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/26/2020] [Accepted: 01/26/2020] [Indexed: 11/02/2022] Open
Abstract
Whole transcriptome analysis is a key method in biology that allows researchers to determine the effect a condition has on gene regulation. One difficulty in RNA sequencing of muscle is that traditional methods are performed on the whole muscle, but this captures non-myogenic cells that are part of the muscle. In order to analyze only the transcriptome of myofibers we combine single myofiber isolation with SMART-Seq to provide high resolution genome wide expression of a single myofiber.
Collapse
Affiliation(s)
- Darren M Blackburn
- Department of Human Genetics, McGill University, 3640 rue University, Montreal, QC, H3A 0C7 Canada.,Molecular and Regenerative Medicine Axis, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, 3640 rue University, Montreal, QC, H3A 0C7 Canada.,Molecular and Regenerative Medicine Axis, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, 3640 rue University, Montreal, QC, H3A 0C7 Canada.,Molecular and Regenerative Medicine Axis, Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Cote-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
| |
Collapse
|
40
|
Serum-free media for the growth of primary bovine myoblasts. Cytotechnology 2019; 72:111-120. [PMID: 31884572 PMCID: PMC7002633 DOI: 10.1007/s10616-019-00361-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/30/2019] [Indexed: 12/11/2022] Open
Abstract
The demand for meat is expected to exceed production capacity by livestock in the coming decennia. Therefore, cultured beef might be a viable alternative to traditional livestock-derived beef. One of the problems however is the sustainability of cultured beef through the use of fetal bovine serum. We aimed to identify a serum-free medium or a serum-replacement that is as effective as the current method used for culturing bovine myoblasts. Cells were harvested from a female Blanc Bleu Belge cow and myoblasts were subsequently isolated. Cells were cultured in either Advanced DMEM containing 20% FBS and 10% HS or one of the chemically-defined, serum-free media for 6 days. MTS was used as a measure of cell proliferation at day 1, 4 or 6 and microscopic pictures were taken to assess cell morphology. FBM™, TesR™ and Essential 8™ are commercially available xeno-free media developed for human PSCs and fibroblasts, with the highest potential to sustain bovine myoblast proliferation. Of the supplements tested, XenoFree™ and a custom-prepared growth factor mix failed to stimulate cell proliferation. LipoGro™ stimulated cell proliferation in some cases but also changed the phenotype of myoblasts to an adipocyte-like phenotype. We conclude that serum-free media stimulate exponential cell expansion, albeit not to the extent of the current growth medium containing up to 30% serum. Further research is needed to investigate whether prolonged cell culture or an adaptation period could further increase cell proliferation.
Collapse
|
41
|
Jia Z, Nie Y, Yue F, Kong Y, Gu L, Gavin TP, Liu X, Kuang S. A requirement of Polo-like kinase 1 in murine embryonic myogenesis and adult muscle regeneration. eLife 2019; 8:e47097. [PMID: 31393265 PMCID: PMC6687435 DOI: 10.7554/elife.47097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 07/17/2019] [Indexed: 01/22/2023] Open
Abstract
Muscle development and regeneration require delicate cell cycle regulation of embryonic myoblasts and adult muscle satellite cells (MuSCs). Through analysis of the Polo-like kinase (Plk) family cell-cycle regulators in mice, we show that Plk1's expression closely mirrors myoblast dynamics during embryonic and postnatal myogenesis. Cell-specific deletion of Plk1 in embryonic myoblasts leads to depletion of myoblasts, developmental failure and prenatal lethality. Postnatal deletion of Plk1 in MuSCs does not perturb their quiescence but depletes activated MuSCs as they enter the cell cycle, leading to regenerative failure. The Plk1-null MuSCs are arrested at the M-phase, accumulate DNA damage, and apoptose. Mechanistically, Plk1 deletion upregulates p53, and inhibition of p53 promotes survival of the Plk1-null myoblasts. Pharmacological inhibition of Plk1 similarly inhibits proliferation but promotes differentiation of myoblasts in vitro, and blocks muscle regeneration in vivo. These results reveal for the first time an indispensable role of Plk1 in developmental and regenerative myogenesis.
Collapse
Affiliation(s)
- Zhihao Jia
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Yaohui Nie
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
- Department of Health and KinesiologyPurdue UniversityWest LafayetteUnited States
| | - Feng Yue
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Yifan Kong
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Lijie Gu
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Timothy P Gavin
- Department of Health and KinesiologyPurdue UniversityWest LafayetteUnited States
| | - Xiaoqi Liu
- Department of BiochemistryPurdue UniversityWest LafayetteUnited States
- Center for Cancer ResearchPurdue UniversityWest LafayetteUnited States
| | - Shihuan Kuang
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
- Center for Cancer ResearchPurdue UniversityWest LafayetteUnited States
| |
Collapse
|
42
|
Chen S, Ding H, Yao X, Xie L. Isolation and Culture of Single Myofiber and Immunostaining of Satellite Cells from Adult C57BL/6J Mice. Bio Protoc 2019; 9:e3313. [PMID: 33654822 DOI: 10.21769/bioprotoc.3313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/28/2019] [Accepted: 06/30/2019] [Indexed: 11/02/2022] Open
Abstract
Myofiber isolation followed with ex vivo culture could recapitulate and visualize satellite cells (SCs) activation, proliferation, and differentiation. This approach could be taken to understand the physiology of satellite cells and the molecular mechanism of regulatory factors, in terms of the involvement of intrinsic factors over SCs quiescence, activation, proliferation and differentiation. Single myofiber culture has several advantages that the traditional approach such as FASC and cryosection could not compete with. For example, myofiber isolation and culture could be used to observe SCs activation, proliferation and differentiation at a continuous manner within their physiological "niche" environment while FACS or cryosection could only capture single time-point upon external stimulation to activate satellite cells by BaCl2, Cardiotoxin or ischemia. Furthermore, in vitro transfection with siRNA or overexpression vector could be performed under ex vivo culture to understand the detailed molecular function of a specific gene on SCs physiology. With these advantages, the physiological state of SCs could be analyzed at multiple designated time-points by immunofluorescence staining. In this protocol, we provide an efficient and practical protocol to isolate single myofiber from EDL muscle, followed with ex vivo culture and immunostaining.
Collapse
Affiliation(s)
- Shujie Chen
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangdong 510070, China
| | - Hongrong Ding
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangdong 510070, China
| | - Xiangping Yao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangdong 510070, China.,Zhujiang Hospital, Southern Medical University, Guangdong 510282, China
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Guangdong Institute of Microbiology, Guangdong Academy of Sciences, Guangdong 510070, China.,Zhujiang Hospital, Southern Medical University, Guangdong 510282, China
| |
Collapse
|
43
|
Pita-Thomas W, Mahar M, Joshi A, Gan D, Cavalli V. HDAC5 promotes optic nerve regeneration by activating the mTOR pathway. Exp Neurol 2019; 317:271-283. [PMID: 30910408 DOI: 10.1016/j.expneurol.2019.03.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/14/2019] [Accepted: 03/21/2019] [Indexed: 11/30/2022]
Abstract
Neurons in the central nervous system (CNS) regenerate poorly compared to their counterparts in the peripheral nervous system. We previously showed that, in peripheral sensory neurons, nuclear HDAC5 inhibits the expression of regenerative associated genes. After nerve injury, HDAC5 is exported to the cytoplasm to promote axon regeneration. Here we investigated the role of HDAC5 in retinal ganglion cells (RGCs), a CNS neuron which fails to survive and regenerate axons after injury. In contrast to PNS neurons, we found that HDAC5 is mostly cytoplasmic in naïve RGCs and its localization is not affected by optic nerve injury, suggesting that HDAC5 does not directly suppress regenerative associated genes in these cells. Manipulation of the PKCμ pathway, the canonical pathway that regulates HDAC5 localization in PNS neurons by phosphorylating serine 259 and 498, and other pathways that regulate nuclear/cytoplasmic transport, did not affect HDAC5 cytoplasmic localization in RGC. Also, an HDAC5 mutant whose serine 259 and 488 were replaced by alanine (HDAC5AA) to prevent phosphorylation and nuclear export showed a predominantly cytoplasmic localization, suggesting that HDAC5 resides mostly in the cytoplasm in RGCs. Interestingly, expression of HDAC5AA, but not HDAC5 wild type, in RGCs in vivo promoted optic nerve regeneration and RGC survival. Mechanistically, we found that HDAC5AA stimulated the survival and regeneration of RGCs by activating the mTOR pathway. Consistently, the combination of HDAC5AA expression and the stimulation of the immune system by zymosan injection had an additive effect in promoting robust axon regeneration. These results reveal the potential of manipulating HDAC5 phosphorylation state to activate the mTOR pathway, offering a new therapeutic target to design drugs that promote axon regeneration in the optic nerve.
Collapse
Affiliation(s)
- Wolfgang Pita-Thomas
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, United States of America
| | - Marcus Mahar
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, United States of America
| | - Avni Joshi
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, United States of America
| | - Di Gan
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, United States of America; Department of Neuroscience, Brandeis University, Waltham, MA 02453, United States of America
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, United States of America; Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, United States of America; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, United States of America.
| |
Collapse
|
44
|
Thoudam T, Ha CM, Leem J, Chanda D, Park JS, Kim HJ, Jeon JH, Choi YK, Liangpunsakul S, Huh YH, Kwon TH, Park KG, Harris RA, Park KS, Rhee HW, Lee IK. PDK4 Augments ER-Mitochondria Contact to Dampen Skeletal Muscle Insulin Signaling During Obesity. Diabetes 2019; 68:571-586. [PMID: 30523025 PMCID: PMC6385748 DOI: 10.2337/db18-0363] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 11/20/2018] [Indexed: 12/17/2022]
Abstract
Mitochondria-associated endoplasmic reticulum membrane (MAM) is a structural link between mitochondria and endoplasmic reticulum (ER). MAM regulates Ca2+ transport from the ER to mitochondria via an IP3R1-GRP75-VDAC1 complex-dependent mechanism. Excessive MAM formation may cause mitochondrial Ca2+ overload and mitochondrial dysfunction. However, the exact implication of MAM formation in metabolic syndromes remains debatable. Here, we demonstrate that PDK4 interacts with and stabilizes the IP3R1-GRP75-VDAC1 complex at the MAM interface. Obesity-induced increase in PDK4 activity augments MAM formation and suppresses insulin signaling. Conversely, PDK4 inhibition dampens MAM formation and improves insulin signaling by preventing MAM-induced mitochondrial Ca2+ accumulation, mitochondrial dysfunction, and ER stress. Furthermore, Pdk4-/- mice exhibit reduced MAM formation and are protected against diet-induced skeletal muscle insulin resistance. Finally, forced formation and stabilization of MAMs with synthetic ER-mitochondria linker prevented the beneficial effects of PDK4 deficiency on insulin signaling. Overall, our findings demonstrate a critical mediatory role of PDK4 in the development of skeletal muscle insulin resistance via enhancement of MAM formation.
Collapse
Affiliation(s)
- Themis Thoudam
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Chae-Myeong Ha
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Dipanjan Chanda
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jong-Seok Park
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hyo-Jeong Kim
- Electron Microscopy Research Center, Korea Basic Science Institute, Ochang, Chungbuk, Republic of Korea
| | - Jae-Han Jeon
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Yeon-Kyung Choi
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Richard L. Roudebush VA Medical Center, Indianapolis, IN
| | - Yang Hoon Huh
- Electron Microscopy Research Center, Korea Basic Science Institute, Ochang, Chungbuk, Republic of Korea
| | - Tae-Hwan Kwon
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Keun-Gyu Park
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Kyu-Sang Park
- Department of Physiology, Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Gangwon-Do, Republic of Korea
| | - Hyun-Woo Rhee
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - In-Kyu Lee
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
45
|
Recessive mutations in muscle-specific isoforms of FXR1 cause congenital multi-minicore myopathy. Nat Commun 2019; 10:797. [PMID: 30770808 PMCID: PMC6377633 DOI: 10.1038/s41467-019-08548-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 01/18/2019] [Indexed: 02/06/2023] Open
Abstract
FXR1 is an alternatively spliced gene that encodes RNA binding proteins (FXR1P) involved in muscle development. In contrast to other tissues, cardiac and skeletal muscle express two FXR1P isoforms that incorporate an additional exon-15. We report that recessive mutations in this particular exon of FXR1 cause congenital multi-minicore myopathy in humans and mice. Additionally, we show that while Myf5-dependent depletion of all FXR1P isoforms is neonatal lethal, mice carrying mutations in exon-15 display non-lethal myopathies which vary in severity depending on the specific effect of each mutation on the protein.
Collapse
|
46
|
Loss of microRNA-23-27-24 clusters in skeletal muscle is not influential in skeletal muscle development and exercise-induced muscle adaptation. Sci Rep 2019; 9:1092. [PMID: 30705375 PMCID: PMC6355808 DOI: 10.1038/s41598-018-37765-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/06/2018] [Indexed: 01/21/2023] Open
Abstract
MicroRNAs are small regulatory noncoding RNAs that repress gene expression at the posttranscriptional level. Previous studies have reported that the expression of miR-23, miR-27, and miR-24, driven from two miR-23–27–24 clusters, is altered by various pathophysiological conditions. However, their functions in skeletal muscle have not been clarified. To define the roles of the miR-23–27–24 clusters in skeletal muscle, we generated double-knockout (dKO) mice muscle-specifically lacking the miR-23–27–24 clusters. The dKO mice were viable and showed normal growth. The contractile and metabolic features of the muscles, represented by the expression of the myosin heavy chain and the oxidative markers PGC1-α and COX IV, were not altered in the dKO mice compared with wild-type mice. The dKO mice showed increased cross-sectional areas of the oxidative fibers. However, this dKO did not induce functional changes in the muscles. The dKO mice also showed normal adaptation to voluntary wheel running for 4 weeks, including the glycolytic-to-oxidative fiber type switch, and increases in mitochondrial markers, succinate dehydrogenase activity, and angiogenesis. In conclusion, our data demonstrate that the miR-23–27–24 clusters have subtle effects on skeletal muscle development and endurance-exercise-induced muscle adaptation.
Collapse
|
47
|
Hüttner SS, Ahrens HE, Schmidt M, Henze H, Jung MJ, Schüler SC, von Maltzahn J. Isolation and Culture of Individual Myofibers and Their Adjacent Muscle Stem Cells from Aged and Adult Skeletal Muscle. Methods Mol Biol 2019; 2045:25-36. [PMID: 30838602 DOI: 10.1007/7651_2019_209] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The isolation and culture of single floating myofibers with their adjacent muscle stem cells allow the analysis and comparison of muscle stem cells from aged and young mice. This method has the advantage that muscle stem cells are cultured on the myofiber, thereby culturing them in conditions as close to their endogenous niche as possible. Here we describe the isolation, culture, transfection with siRNA, and subsequent immunostaining for muscle stem cells on their adjacent myofibers from aged and young mice.
Collapse
Affiliation(s)
- Sören S Hüttner
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Jena, Germany
| | - Hellen E Ahrens
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Jena, Germany
| | - Manuel Schmidt
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Jena, Germany
| | - Henriette Henze
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Jena, Germany
| | | | - Svenja C Schüler
- Leibniz-Institute on Aging - Fritz-Lipmann-Institute, Jena, Germany
| | | |
Collapse
|
48
|
The RNA-binding proteins Zfp36l1 and Zfp36l2 act redundantly in myogenesis. Skelet Muscle 2018; 8:37. [PMID: 30526691 PMCID: PMC6286576 DOI: 10.1186/s13395-018-0183-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/22/2018] [Indexed: 01/07/2023] Open
Abstract
Background Members of the ZFP36 family of RNA-binding proteins regulate gene expression post-transcriptionally by binding to AU-rich elements in the 3’UTR of mRNA and stimulating mRNA degradation. The proteins within this family target different transcripts in different tissues. In particular, ZFP36 targets myogenic transcripts and may have a role in adult muscle stem cell quiescence. Our study examined the requirement of ZFP36L1 and ZFP36L2 in adult muscle cell fate regulation. Methods We generated single and double conditional knockout mice in which Zfp36l1 and/or Zfp36l2 were deleted in Pax7-expressing cells. Immunostained muscle sections were used to analyse resting skeletal muscle, and a cardiotoxin-induced injury model was used to determine the regenerative capacity of muscle. Results We show that ZFP36L1 and ZFP36L2 proteins are expressed in satellite cells. Mice lacking the two proteins in Pax7-expressing cells have reduced body weight and have reduced skeletal muscle mass. Furthermore, the number of satellite cells is reduced in adult skeletal muscle and the capacity of this muscle to regenerate following muscle injury is diminished. Conclusion ZFP36L1 and ZFP36L2 act redundantly in myogenesis. These findings add further intricacy to the regulation of the cell fate of Pax7-expressing cells in skeletal muscle by RNA-binding proteins. Electronic supplementary material The online version of this article (10.1186/s13395-018-0183-9) contains supplementary material, which is available to authorized users.
Collapse
|
49
|
Kwong JQ, Huo J, Bround MJ, Boyer JG, Schwanekamp JA, Ghazal N, Maxwell JT, Jang YC, Khuchua Z, Shi K, Bers DM, Davis J, Molkentin JD. The mitochondrial calcium uniporter underlies metabolic fuel preference in skeletal muscle. JCI Insight 2018; 3:121689. [PMID: 30429366 DOI: 10.1172/jci.insight.121689] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 10/04/2018] [Indexed: 01/17/2023] Open
Abstract
The mitochondrial Ca2+ uniporter (MCU) complex mediates acute mitochondrial Ca2+ influx. In skeletal muscle, MCU links Ca2+ signaling to energy production by directly enhancing the activity of key metabolic enzymes in the mitochondria. Here, we examined the role of MCU in skeletal muscle development and metabolic function by generating mouse models for the targeted deletion of Mcu in embryonic, postnatal, and adult skeletal muscle. Loss of Mcu did not affect muscle growth and maturation or otherwise cause pathology. Skeletal muscle-specific deletion of Mcu in mice also did not affect myofiber intracellular Ca2+ handling, but it did inhibit acute mitochondrial Ca2+ influx and mitochondrial respiration stimulated by Ca2+, resulting in reduced acute exercise performance in mice. However, loss of Mcu also resulted in enhanced muscle performance under conditions of fatigue, with a preferential shift toward fatty acid metabolism, resulting in reduced body fat with aging. Together, these results demonstrate that MCU-mediated mitochondrial Ca2+ regulation underlies skeletal muscle fuel selection at baseline and under enhanced physiological demands, which affects total homeostatic metabolism.
Collapse
Affiliation(s)
- Jennifer Q Kwong
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Pediatrics, Division of Cardiovascular Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jiuzhou Huo
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael J Bround
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Justin G Boyer
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jennifer A Schwanekamp
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Nasab Ghazal
- Department of Pediatrics, Division of Cardiovascular Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Joshua T Maxwell
- Department of Pediatrics, Division of Cardiovascular Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Young C Jang
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Zaza Khuchua
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA.,Sechenov University, Moscow, Russia
| | - Kevin Shi
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, California, USA
| | - Jennifer Davis
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA.,Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
50
|
Aloysius A, DasGupta R, Dhawan J. The transcription factor Lef1 switches partners from β-catenin to Smad3 during muscle stem cell quiescence. Sci Signal 2018; 11:11/540/eaan3000. [PMID: 30042129 DOI: 10.1126/scisignal.aan3000] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skeletal muscle stem cells (MuSCs), also known as satellite cells, persist in adult mammals by entering a state of quiescence (G0) during the early postnatal period. Quiescence is reversed during damage-induced regeneration and re-established after regeneration. Entry of cultured myoblasts into G0 is associated with a specific, reversible induction of Wnt target genes, thus implicating members of the Tcf and Lef1 (Tcf/Lef) transcription factor family, which mediate transcriptional responses to Wnt signaling, in the initiation of quiescence. We found that the canonical Wnt effector β-catenin, which cooperates with Tcf/Lef, was dispensable for myoblasts to enter quiescence. Using pharmacological and genetic approaches in cultured C2C12 myoblasts and in MuSCs, we demonstrated that Tcf/Lef activity during quiescence depended not on β-catenin but on the transforming growth factor-β (TGF-β) effector and transcriptional coactivator Smad3, which colocalized with Lef1 at canonical Wnt-responsive elements and directly interacted with Lef1 specifically in G0 Depletion of Smad3, but not β-catenin, reduced Lef1 occupancy at target promoters, Tcf/Lef target gene expression, and self-renewal of myoblasts. In vivo, MuSCs underwent a switch from β-catenin-Lef1 to Smad3-Lef1 interactions during the postnatal switch from proliferation to quiescence, with β-catenin-Lef1 interactions recurring during damage-induced reactivation. Our findings suggest that the interplay of Wnt-Tcf/Lef and TGF-β-Smad3 signaling activates canonical Wnt target promoters in a manner that depends on β-catenin during myoblast proliferation but is independent of β-catenin during MuSC quiescence.
Collapse
Affiliation(s)
- Ajoy Aloysius
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India.,Centre for Cellular and Molecular Biology, Hyderabad 500007, India.,Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India
| | | | - Jyotsna Dhawan
- Centre for Cellular and Molecular Biology, Hyderabad 500007, India. .,Institute for Stem Cell Biology and Regenerative Medicine, Bangalore 560065, India
| |
Collapse
|