51
|
Santarelli P, Rosti V, Vivo M, Lanzuolo C. Chromatin organization of muscle stem cell. Curr Top Dev Biol 2024; 158:375-406. [PMID: 38670713 DOI: 10.1016/bs.ctdb.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
The proper functioning of skeletal muscles is essential throughout life. A crucial crosstalk between the environment and several cellular mechanisms allows striated muscles to perform successfully. Notably, the skeletal muscle tissue reacts to an injury producing a completely functioning tissue. The muscle's robust regenerative capacity relies on the fine coordination between muscle stem cells (MuSCs or "satellite cells") and their specific microenvironment that dictates stem cells' activation, differentiation, and self-renewal. Critical for the muscle stem cell pool is a fine regulation of chromatin organization and gene expression. Acquiring a lineage-specific 3D genome architecture constitutes a crucial modulator of muscle stem cell function during development, in the adult stage, in physiological and pathological conditions. The context-dependent relationship between genome structure, such as accessibility and chromatin compartmentalization, and their functional effects will be analysed considering the improved 3D epigenome knowledge, underlining the intimate liaison between environmental encounters and epigenetics.
Collapse
Affiliation(s)
- Philina Santarelli
- INGM Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy
| | - Valentina Rosti
- INGM Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy; CNR Institute of Biomedical Technologies, Milan, Italy
| | - Maria Vivo
- Università degli studi di Salerno, Fisciano, Italy.
| | - Chiara Lanzuolo
- INGM Istituto Nazionale Genetica Molecolare Romeo ed Enrica Invernizzi, Milan, Italy; CNR Institute of Biomedical Technologies, Milan, Italy.
| |
Collapse
|
52
|
Bachman JF, Chakkalakal JV. Satellite cells in the growth and maintenance of muscle. Curr Top Dev Biol 2024; 158:1-14. [PMID: 38670701 DOI: 10.1016/bs.ctdb.2024.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Embryonic skeletal muscle growth is contingent upon a population of somite derived satellite cells, however, the contribution of these cells to early postnatal skeletal muscle growth remains relatively high. As prepubertal postnatal development proceeds, the activity and contribution of satellite cells to skeletal muscle growth diminishes. Eventually, at around puberty, a population of satellite cells escapes terminal commitment, continues to express the paired box transcription factor Pax7, and reside in a quiescent state orbiting the myofiber periphery adjacent to the basal lamina. After adolescence, some satellite cell contributions to muscle maintenance and adaptation occur, however, their necessity is reduced relative to embryonic, early postnatal, and prepubertal growth.
Collapse
Affiliation(s)
| | - Joe V Chakkalakal
- Departments of Orthopedic Surgery and Cell Biology, Duke University School of Medicine, Durham NC, USA.
| |
Collapse
|
53
|
Kureel SK, Blair B, Sheetz MP. Recent Advancement in Elimination Strategies and Potential Rejuvenation Targets of Senescence. Adv Biol (Weinh) 2024; 8:e2300461. [PMID: 37857532 DOI: 10.1002/adbi.202300461] [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: 08/29/2023] [Indexed: 10/21/2023]
Abstract
Cellular senescence is a state of exiting the cell cycle, resisting apoptosis, and changing phenotype. Senescent cells (SCs) can be identified by large, distorted morphology and irreversible inability to replicate. In early development, senescence has beneficial roles like tissue patterning and wound healing, where SCs are cleared by the immune system. However, there is a steep rise in SC number as organisms age. The issue with SC accumulation stems from the loss of cellular function, alterations of the microenvironment, and secretions of pro-inflammatory molecules, consisting of cytokines, chemokines, matrix metalloproteinases (MMPs), interleukins, and extracellular matrix (ECM)-associated molecules. This secreted cocktail is referred to as the senescence-associated secretory phenotype (SASP), a hallmark of cellular senescence. The SASP promotes inflammation and displays a bystander effect where paracrine signaling turns proliferating cells into senescent states. To alleviate age-associated diseases, researchers have developed novel methods and techniques to selectively eliminate SCs in aged individuals. Although studies demonstrated that selectively killing SCs improves age-related disorders, there are drawbacks to SC removal. Considering favorable aspects of senescence in the body, this paper reviews recent advancements in elimination strategies and potential rejuvenation targets of senescence to bring researchers in the field up to date.
Collapse
Affiliation(s)
- Sanjay Kumar Kureel
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Brandon Blair
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Michael P Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
54
|
Whitlock JM. Muscle Progenitor Cell Fusion in the Maintenance of Skeletal Muscle. Results Probl Cell Differ 2024; 71:257-279. [PMID: 37996682 DOI: 10.1007/978-3-031-37936-9_13] [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: 11/25/2023]
Abstract
Skeletal muscle possesses a resident, multipotent stem cell population that is essential for its repair and maintenance throughout life. Here I highlight the role of this stem cell population in muscle repair and regeneration and review the genetic control of the process; the mechanistic steps of activation, migration, recognition, adhesion, and fusion of these cells; and discuss the novel recognition of the membrane signaling that coordinates myogenic cell-cell fusion, as well as the identification of a two-part fusogen system that facilitates it.
Collapse
Affiliation(s)
- Jarred M Whitlock
- Section on Membrane Biology, Eunice Kennedy Shrive National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
55
|
Lin KH, Hibbert JE, Lemens JL, Torbey MM, Steinert ND, Flejsierowicz PM, Melka KM, Lares M, Setaluri V, Hornberger TA. The role of satellite cell-derived TRIM28 in mechanical load- and injury-induced myogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572566. [PMID: 38187693 PMCID: PMC10769277 DOI: 10.1101/2023.12.20.572566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Satellite cells are skeletal muscle stem cells that contribute to postnatal muscle growth, and they endow skeletal muscle with the ability to regenerate after a severe injury. Here we discovered that this myogenic potential of satellite cells requires a protein called tripartite motif-containing 28 (TRIM28). Unexpectedly, multiple lines of both in vitro and in vivo evidence revealed that the myogenic function of TRIM28 is not dependent on changes in the phosphorylation of its serine 473 residue. Moreover, the functions of TRIM28 were not mediated through the regulation of satellite cell proliferation or differentiation. Instead, our findings indicate that TRIM28 regulates the ability of satellite cells to progress through the process of fusion. Specifically, we discovered that TRIM28 controls the expression of a fusogenic protein called myomixer and concomitant fusion pore formation. Collectively, the outcomes of this study expose the framework of a novel regulatory pathway that is essential for myogenesis.
Collapse
Affiliation(s)
- Kuan-Hung Lin
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jamie E. Hibbert
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Jake L. Lemens
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Melissa M. Torbey
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Nathaniel D. Steinert
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Philip M. Flejsierowicz
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Kiley M. Melka
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| | - Marcos Lares
- Department of Dermatology, University of Wisconsin - Madison, WI, USA
| | | | - Troy A. Hornberger
- Department of Comparative Biosciences, University of Wisconsin - Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin - Madison, WI, USA
| |
Collapse
|
56
|
Yaghi OK, Hanna BS, Langston PK, Michelson DA, Jayewickreme T, Marin-Rodero M, Benoist C, Mathis D. A discrete 'early-responder' stromal-cell subtype orchestrates immunocyte recruitment to injured tissue. Nat Immunol 2023; 24:2053-2067. [PMID: 37932455 PMCID: PMC10792729 DOI: 10.1038/s41590-023-01669-w] [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: 07/15/2022] [Accepted: 10/05/2023] [Indexed: 11/08/2023]
Abstract
Following acute injury, stromal cells promote tissue regeneration by a diversity of mechanisms. Time-resolved single-cell RNA sequencing of muscle mesenchymal stromal cells (MmSCs) responding to acute injury identified an 'early-responder' subtype that spiked on day 1 and expressed a notable array of transcripts encoding immunomodulators. IL-1β, TNF-α and oncostatin M each strongly and rapidly induced MmSCs transcribing this immunomodulatory program. Macrophages amplified the program but were not strictly required for its induction. Transfer of the inflammatory MmSC subtype, tagged with a unique surface marker, into healthy hindlimb muscle induced inflammation primarily driven by neutrophils and macrophages. Among the abundant inflammatory transcripts produced by this subtype, Cxcl5 was stroma-specific and highly upregulated with injury. Depletion of this chemokine early after injury revealed a substantial impact on recruitment of neutrophils, a prolongation of inflammation to later times and an effect on tissue regeneration. Mesenchymal stromal cell subtypes expressing a comparable inflammatory program were found in a mouse model of muscular dystrophy and in several other tissues and pathologies in both mice and humans. These 'early-responder' mesenchymal stromal cells, already in place, permit rapid and coordinated mobilization and amplification of critical cell collaborators in response to injury.
Collapse
Affiliation(s)
- Omar K Yaghi
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Bola S Hanna
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - P Kent Langston
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel A Michelson
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Teshika Jayewickreme
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Miguel Marin-Rodero
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
57
|
Hicks MR, Saleh KK, Clock B, Gibbs DE, Yang M, Younesi S, Gane L, Gutierrez-Garcia V, Xi H, Pyle AD. Regenerating human skeletal muscle forms an emerging niche in vivo to support PAX7 cells. Nat Cell Biol 2023; 25:1758-1773. [PMID: 37919520 PMCID: PMC10709143 DOI: 10.1038/s41556-023-01271-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 09/26/2023] [Indexed: 11/04/2023]
Abstract
Skeletal muscle stem and progenitor cells including those derived from human pluripotent stem cells (hPSCs) offer an avenue towards personalized therapies and readily fuse to form human-mouse myofibres in vivo. However, skeletal muscle progenitor cells (SMPCs) inefficiently colonize chimeric stem cell niches and instead associate with human myofibres resembling foetal niches. We hypothesized competition with mouse satellite cells (SCs) prevented SMPC engraftment into the SC niche and thus generated an SC ablation mouse compatible with human engraftment. Single-nucleus RNA sequencing of SC-ablated mice identified the absence of a transient myofibre subtype during regeneration expressing Actc1. Similarly, ACTC1+ human myofibres supporting PAX7+ SMPCs increased in SC-ablated mice, and after re-injury we found SMPCs could now repopulate into chimeric niches. To demonstrate ACTC1+ myofibres are essential to supporting PAX7 SMPCs, we generated caspase-inducible ACTC1 depletion human pluripotent stem cells, and upon SMPC engraftment we found a 90% reduction in ACTC1+ myofibres and a 100-fold decrease in PAX7 cell numbers compared with non-induced controls. We used spatial RNA sequencing to identify key factors driving emerging human niche formation between ACTC1+ myofibres and PAX7+ SMPCs in vivo. This revealed that transient regenerating human myofibres are essential for emerging niche formation in vivo to support PAX7 SMPCs.
Collapse
Affiliation(s)
- Michael R Hicks
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA.
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA.
- Physiology and Biophysics, University of California, Irvine, CA, USA.
| | - Kholoud K Saleh
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, USA
| | - Ben Clock
- Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Devin E Gibbs
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Mandee Yang
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Shahab Younesi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Lily Gane
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | | | - Haibin Xi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - April D Pyle
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA.
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
- Jonnson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
| |
Collapse
|
58
|
Koopmans PJ, Ismaeel A, Goljanek-Whysall K, Murach KA. The roles of miRNAs in adult skeletal muscle satellite cells. Free Radic Biol Med 2023; 209:228-238. [PMID: 37879420 PMCID: PMC10911817 DOI: 10.1016/j.freeradbiomed.2023.10.403] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023]
Abstract
Satellite cells are bona fide muscle stem cells that are indispensable for successful post-natal muscle growth and regeneration after severe injury. These cells also participate in adult muscle adaptation in several capacities. MicroRNAs (miRNAs) are post-transcriptional regulators of mRNA that are implicated in several aspects of stem cell function. There is evidence to suggest that miRNAs affect satellite cell behavior in vivo during development and myogenic progenitor behavior in vitro, but the role of miRNAs in adult skeletal muscle satellite cells is less studied. In this review, we provide evidence for how miRNAs control satellite cell function with emphasis on satellite cells of adult skeletal muscle in vivo. We first outline how miRNAs are indispensable for satellite cell viability and control the phases of myogenesis. Next, we discuss the interplay between miRNAs and myogenic cell redox status, senescence, and communication to other muscle-resident cells during muscle adaptation. Results from recent satellite cell miRNA profiling studies are also summarized. In vitro experiments in primary myogenic cells and cell lines have been invaluable for exploring the influence of miRNAs, but we identify a need for novel genetic tools to further interrogate how miRNAs control satellite cell behavior in adult skeletal muscle in vivo.
Collapse
Affiliation(s)
- Pieter Jan Koopmans
- Exercise Science Research Center, Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA; Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40506, USA
| | - Katarzyna Goljanek-Whysall
- School of Medicine, College of Medicine, Nursing, and Health Sciences, University of Galway, Galway, Ireland
| | - Kevin A Murach
- Exercise Science Research Center, Molecular Muscle Mass Regulation Laboratory, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA; Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, 72701, USA.
| |
Collapse
|
59
|
Shao A, Kissil JL, Fan CM. The L27 Domain of MPP7 enhances TAZ-YY1 Cooperation to Renew Muscle Stem Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.01.565166. [PMID: 37961392 PMCID: PMC10635061 DOI: 10.1101/2023.11.01.565166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Stem cells regenerate differentiated cells to maintain and repair tissues and organs. They also replenish themselves, i.e. self-renewal, for the regenerative process to last a lifetime. How stem cells renew is of critical biological and medical significance. Here we use the skeletal muscle stem cell (MuSC) to study this process. Using a combination of genetic, molecular, and biochemical approaches, we show that MPP7, AMOT, and TAZ/YAP form a complex that activates a common set of target genes. Among these targets, Carm1 can direct MuSC renewal. In the absence of MPP7, TAZ can support regenerative progenitors and activate Carm1 expression, but not to a level needed for self-renewal. Facilitated by the actin polymerization-responsive AMOT, TAZ recruits the L27 domain of MPP7 to up-regulate Carm1 to the level necessary to drive MuSC renewal. The promoter of Carm1, and those of other common downstream genes, also contain binding site(s) for YY1. We further demonstrate that the L27 domain of MPP7 enhances the interaction between TAZ and YY1 to activate Carm1. Our results define a renewal transcriptional program embedded within the progenitor program, by selectively up-regulating key gene(s) within the latter, through the combination of protein interactions and in a manner dependent on the promoter context.
Collapse
Affiliation(s)
- Anwen Shao
- Department of Embryology, Carnegie Institution for Science, 3520 San Martin Drive, Baltimore, MD 21218
| | - Joseph L. Kissil
- Department of Molecular Oncology, The H. Lee Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, FL 33612
| | - Chen-Ming Fan
- Department of Embryology, Carnegie Institution for Science, 3520 San Martin Drive, Baltimore, MD 21218
- Department of Biology, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218
| |
Collapse
|
60
|
Salga M, Samuel SG, Tseng HW, Gatin L, Girard D, Rival B, Barbier V, Bisht K, Shatunova S, Debaud C, Winkler IG, Paquereau J, Dinh A, Genêt G, Kerever S, Abback PS, Banzet S, Genêt F, Lévesque JP, Alexander KA. Bacterial Lipopolysaccharides Exacerbate Neurogenic Heterotopic Ossification Development. J Bone Miner Res 2023; 38:1700-1717. [PMID: 37602772 DOI: 10.1002/jbmr.4905] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/24/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023]
Abstract
Neurogenic heterotopic ossifications (NHO) are heterotopic bones that develop in periarticular muscles after severe central nervous system (CNS) injuries. Several retrospective studies have shown that NHO prevalence is higher in patients who suffer concomitant infections. However, it is unclear whether these infections directly contribute to NHO development or reflect the immunodepression observed in patients with CNS injury. Using our mouse model of NHO induced by spinal cord injury (SCI) between vertebrae T11 to T13 , we demonstrate that lipopolysaccharides (LPS) from gram-negative bacteria exacerbate NHO development in a toll-like receptor-4 (TLR4)-dependent manner, signaling through the TIR-domain-containing adapter-inducing interferon-β (TRIF/TICAM1) adaptor rather than the myeloid differentiation primary response-88 (MYD88) adaptor. We find that T11 to T13 SCI did not significantly alter intestinal integrity nor cause intestinal bacteria translocation or endotoxemia, suggesting that NHO development is not driven by endotoxins from the gut in this model of SCI-induced NHO. Relevant to the human pathology, LPS increased expression of osteoblast markers in cultures of human fibro-adipogenic progenitors isolated from muscles surrounding NHO biopsies. In a case-control retrospective study in patients with traumatic brain injuries, infections with gram-negative Pseudomonas species were significantly associated with NHO development. Together these data suggest a functional association between gram-negative bacterial infections and NHO development and highlights infection management as a key consideration to avoid NHO development in patients. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Marjorie Salga
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Australia
- University of Versailles Saint Quentin en Yvelines, END:ICAP U1179 INSERM, UFR Simone Veil-Santé, Montigny le Bretonneux, France
- UPOH (Unité Péri Opératoire du Handicap), Physical and Rehabilitation Medicine Department, Raymond-Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Garches, France
| | - Selwin G Samuel
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Australia
- Department of Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Chennai, India
| | - Hsu-Wen Tseng
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Laure Gatin
- University of Versailles Saint Quentin en Yvelines, END:ICAP U1179 INSERM, UFR Simone Veil-Santé, Montigny le Bretonneux, France
- UPOH (Unité Péri Opératoire du Handicap), Physical and Rehabilitation Medicine Department, Raymond-Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Garches, France
- Department of Orthopedic Surgery, Raymond Poincaré Hospital, AP-HP, Garches, France
| | - Dorothée Girard
- Institut de Recherche Biomédicale des Armées (IRBA), INSERM UMR-MD 1197, Clamart, France
| | - Bastien Rival
- Institut de Recherche Biomédicale des Armées (IRBA), INSERM UMR-MD 1197, Clamart, France
| | - Valérie Barbier
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Kavita Bisht
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Svetlana Shatunova
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Charlotte Debaud
- University of Versailles Saint Quentin en Yvelines, END:ICAP U1179 INSERM, UFR Simone Veil-Santé, Montigny le Bretonneux, France
| | - Ingrid G Winkler
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Julie Paquereau
- UPOH (Unité Péri Opératoire du Handicap), Physical and Rehabilitation Medicine Department, Raymond-Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Garches, France
| | - Aurélien Dinh
- Department of Infectious Diseases, Raymond Poincaré Hospital, AP-HP, Garches, France
| | - Guillaume Genêt
- University of Versailles Saint Quentin en Yvelines, END:ICAP U1179 INSERM, UFR Simone Veil-Santé, Montigny le Bretonneux, France
| | - Sébastien Kerever
- Department of Anesthesiology and Critical Care, Lariboisière University Hospital, AP-HP, Paris, France
| | - Paer-Sélim Abback
- Department of Anesthesiology and Critical Care, Beaujon Hospital, DMU Parabol, AP-HP, Clichy, France
| | - Sébastien Banzet
- Institut de Recherche Biomédicale des Armées (IRBA), INSERM UMR-MD 1197, Clamart, France
| | - François Genêt
- University of Versailles Saint Quentin en Yvelines, END:ICAP U1179 INSERM, UFR Simone Veil-Santé, Montigny le Bretonneux, France
- UPOH (Unité Péri Opératoire du Handicap), Physical and Rehabilitation Medicine Department, Raymond-Poincaré Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Garches, France
| | - Jean-Pierre Lévesque
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Kylie A Alexander
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| |
Collapse
|
61
|
Gartling G, Nakamura R, Bing R, Branski RC. A Novel Method for Thyroarytenoid Myofiber Culture. Laryngoscope 2023; 133:3109-3115. [PMID: 37227163 PMCID: PMC11881223 DOI: 10.1002/lary.30756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/20/2023] [Accepted: 04/29/2023] [Indexed: 05/26/2023]
Abstract
OBJECTIVES/HYPOTHESIS Myofiber culture has been employed to investigate muscle physiology in vitro and is well-established in the rodent hind limb. Thyroarytenoid (TA) myofiber culture has not been described, providing an opportunity to employ this method to investigate distinct TA myofiber functions. The purpose of this study was to assess the feasibility of a TA myofiber culture model. STUDY DESIGN In vitro. METHODS TA muscles from five Sprague Dawley rats were independently isolated and digested for 90 min. A smooth-tip, wide-bored pipette dissociated TA myofibers from cartilage, and the fibers were distributed on collagen-coated dishes and incubated at 37°C, 5% CO2 for 2 h. Myofiber specificity was determined via immunolabeling for desmin and myosin heavy chain (MHC). Myofibers viability was assessed over 7 days via esterase assay. Additional myofibers were immunolabeled for satellite cell marker Pax-7. Glucocorticoid (GC) receptor (GR) was immunolabeled following GC treatment. RESULTS The harvest technique yielded ~120 myofibers per larynx. By day 7, ~60% of the fibers remained attached and were calcein AM-positive/ethidium homodimer-negative, indicating viability. Myofibers were positive for desmin and MHC, indicating muscle specificity. Cells surrounding myofibers were positive for Pax-7, indicating the presence of myogenic satellite cells. Myofibers also responded to GC treatment as determined by GR nuclear translocation. CONCLUSION TA myofibers remained viable in culture for at least 7 days with a predictable response to exogenous stimuli. This technique provides novel investigative opportunities regarding TA structure and function. LEVEL OF EVIDENCE N/A Laryngoscope, 133:3109-3115, 2023.
Collapse
Affiliation(s)
- Gary Gartling
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York City, New York, USA
| | - Ryosuke Nakamura
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York City, New York, USA
| | - Renjie Bing
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York City, New York, USA
| | - Ryan C Branski
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York City, New York, USA
- Otolaryngology-Head and Neck Surgery, NYU Grossman School of Medicine, New York City, New York, USA
| |
Collapse
|
62
|
Peng J, Han L, Liu B, Song J, Wang Y, Wang K, Guo Q, Liu X, Li Y, Zhang J, Wu W, Li S, Fu X, Zhuang CL, Zhang W, Suo S, Hu P, Zhao Y. Gli1 marks a sentinel muscle stem cell population for muscle regeneration. Nat Commun 2023; 14:6993. [PMID: 37914731 PMCID: PMC10620419 DOI: 10.1038/s41467-023-42837-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023] Open
Abstract
Adult skeletal muscle regeneration is mainly driven by muscle stem cells (MuSCs), which are highly heterogeneous. Although recent studies have started to characterize the heterogeneity of MuSCs, whether a subset of cells with distinct exists within MuSCs remains unanswered. Here, we find that a population of MuSCs, marked by Gli1 expression, is required for muscle regeneration. The Gli1+ MuSC population displays advantages in proliferation and differentiation both in vitro and in vivo. Depletion of this population leads to delayed muscle regeneration, while transplanted Gli1+ MuSCs support muscle regeneration more effectively than Gli1- MuSCs. Further analysis reveals that even in the uninjured muscle, Gli1+ MuSCs have elevated mTOR signaling activity, increased cell size and mitochondrial numbers compared to Gli1- MuSCs, indicating Gli1+ MuSCs are displaying the features of primed MuSCs. Moreover, Gli1+ MuSCs greatly contribute to the formation of GAlert cells after muscle injury. Collectively, our findings demonstrate that Gli1+ MuSCs represents a distinct MuSC population which is more active in the homeostatic muscle and enters the cell cycle shortly after injury. This population functions as the tissue-resident sentinel that rapidly responds to injury and initiates muscle regeneration.
Collapse
Affiliation(s)
- Jiayin Peng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Lili Han
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Biao Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Jiawen Song
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Yuang Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Kunpeng Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai, 201210, PR China
| | - Qian Guo
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - XinYan Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Yu Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Jujin Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Wenqing Wu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Sheng Li
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200023, PR China
| | - Xin Fu
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200023, PR China
| | - Cheng-le Zhuang
- The 10th People's Hospital affiliated to Tongji University, Shanghai, 200072, PR China
| | - Weikang Zhang
- Guangzhou Laboratory-Guangzhou Medical University, Guangzhou, 510005, PR China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Shengbao Suo
- Guangzhou Laboratory-Guangzhou Medical University, Guangzhou, 510005, PR China
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, Guangdong, 510005, PR China
| | - Ping Hu
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200023, PR China.
- The 10th People's Hospital affiliated to Tongji University, Shanghai, 200072, PR China.
- Guangzhou Laboratory-Guangzhou Medical University, Guangzhou, 510005, PR China.
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510005, PR China.
| | - Yun Zhao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China.
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai, 201210, PR China.
| |
Collapse
|
63
|
Ninfali C, Siles L, Esteve-Codina A, Postigo A. The mesodermal and myogenic specification of hESCs depend on ZEB1 and are inhibited by ZEB2. Cell Rep 2023; 42:113222. [PMID: 37819755 DOI: 10.1016/j.celrep.2023.113222] [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: 03/12/2022] [Revised: 08/02/2023] [Accepted: 09/20/2023] [Indexed: 10/13/2023] Open
Abstract
Human embryonic stem cells (hESCs) can differentiate into any cell lineage. Here, we report that ZEB1 and ZEB2 promote and inhibit mesodermal-to-myogenic specification of hESCs, respectively. Knockdown and/or overexpression experiments of ZEB1, ZEB2, or PAX7 in hESCs indicate that ZEB1 is required for hESC Nodal/Activin-mediated mesodermal specification and PAX7+ human myogenic progenitor (hMuP) generation, while ZEB2 inhibits these processes. ZEB1 downregulation induces neural markers, while ZEB2 downregulation induces mesodermal/myogenic markers. Mechanistically, ZEB1 binds to and transcriptionally activates the PAX7 promoter, while ZEB2 binds to and activates the promoter of the neural OTX2 marker. Transplanting ZEB1 or ZEB2 knocked down hMuPs into the muscles of a muscular dystrophy mouse model, showing that hMuP engraftment and generation of dystrophin-positive myofibers depend on ZEB1 and are inhibited by ZEB2. The mouse model results suggest that ZEB1 expression and/or downregulating ZEB2 in hESCs may also enhance hESC regenerative capacity for human muscular dystrophy therapy.
Collapse
Affiliation(s)
- Chiara Ninfali
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036 Barcelona, Spain
| | - Laura Siles
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036 Barcelona, Spain
| | | | - Antonio Postigo
- Group of Gene Regulation in Stem Cells, Cell Plasticity, Differentiation, and Cancer, IDIBAPS, 08036 Barcelona, Spain; Molecular Targets Program, J.G. Brown Center, Louisville University Healthcare Campus, Louisville, KY 40202, USA; ICREA, 08010 Barcelona, Spain.
| |
Collapse
|
64
|
Mademtzoglou D, Geara P, Mourikis P, Relaix F. Pax7 haploinsufficiency impairs muscle stem cell function in Cre-recombinase mice and underscores the importance of appropriate controls. Stem Cell Res Ther 2023; 14:294. [PMID: 37833800 PMCID: PMC10576335 DOI: 10.1186/s13287-023-03506-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
Ever since its introduction as a genetic tool, the Cre-lox system has been widely used for molecular genetic studies in vivo in the context of health and disease, as it allows time- and cell-specific gene modifications. However, insertion of the Cre-recombinase cassette in the gene of interest can alter transcription, protein expression, or function, either directly, by modifying the landscape of the locus, or indirectly, due to the lack of genetic compensation or by indirect impairment of the non-targeted allele. This is sometimes the case when Cre-lox is used for muscle stem cell studies. Muscle stem cells are required for skeletal muscle growth, regeneration and to delay muscle disease progression, hence providing an attractive model for stem cell research. Since the transcription factor Pax7 is specifically expressed in all muscle stem cells, tamoxifen-inducible Cre cassettes (CreERT2) have been inserted into this locus by different groups to allow targeted gene recombination. Here we compare the two Pax7-CreERT2 mouse lines that are mainly used to evaluate muscle regeneration and development of pathological features upon deletion of specific factors or pathways. We applied diverse commonly used tamoxifen schemes of CreERT2 activation, and we analyzed muscle repair after cardiotoxin-induced injury. We show that consistently the Pax7-CreERT2 allele targeted into the Pax7 coding sequence (knock-in/knock-out allele) produces an inherent defect in regeneration, manifested as delayed post-injury repair and reduction in muscle stem cell numbers. In genetic ablation studies lacking proper controls, this inherent defect could be misinterpreted as being provoked by the deletion of the factor of interest. Instead, using an alternative Pax7-CreERT2 allele that maintains bi-allelic Pax7 expression or including appropriate controls can prevent misinterpretation of experimental data. The findings presented here can guide researchers establish appropriate experimental design for muscle stem cell genetic studies.
Collapse
Affiliation(s)
| | - Perla Geara
- Univ Paris Est Creteil, INSERM, IMRB, F-94010, Creteil, France
| | | | - Frederic Relaix
- Univ Paris Est Creteil, INSERM, IMRB, F-94010, Creteil, France.
- Ecole nationale vétérinaire d'Alfort, IMRB, F-94700, Maisons-Alfort, France.
- EFS, IMRB, F-94010, Creteil, France.
- AP-HP, Hopital Mondor, Service d'histologie, F-94010, Creteil, France.
| |
Collapse
|
65
|
Barsky ST, Monks DA. Androgen action on myogenesis throughout the lifespan; comparison with neurogenesis. Front Neuroendocrinol 2023; 71:101101. [PMID: 37669703 DOI: 10.1016/j.yfrne.2023.101101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/07/2023]
Abstract
Androgens' pleiotropic actions in promoting sex differences present not only a challenge to providing a comprehensive account of their function, but also an opportunity to gain insights by comparing androgenic actions across organ systems. Although often overlooked by neuroscientists, skeletal muscle is another androgen-responsive organ system which shares with the nervous system properties of electrochemical excitability, behavioral relevance, and remarkable capacity for adaptive plasticity. Here we review androgenic regulation of mitogenic plasticity in skeletal muscle with the goal of identifying areas of interest to those researching androgenic mechanisms mediating sexual differentiation of neurogenesis. We use an organizational-activational framework to relate broad areas of similarity and difference between androgen effects on mitogenesis in muscle and brain throughout the lifespan, from early organogenesis, through pubertal organization, adult activation, and aging. The focus of the review is androgenic regulation of muscle-specific stem cells (satellite cells), which share with neural stem cells essential functions in development, plasticity, and repair, albeit with distinct, muscle-specific features. Also considered are areas of paracrine and endocrine interaction between androgen action on muscle and nervous system, including mediation of neural plasticity of innervating and distal neural populations by muscle-produced trophic factors.
Collapse
Affiliation(s)
- Sabrina Tzivia Barsky
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario, Canada.
| | - Douglas Ashley Monks
- Department of Cell & Systems Biology, Faculty of Arts & Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychology, Faculty of Arts & Science, University of Toronto Mississauga, Mississauga, Ontario, Canada.
| |
Collapse
|
66
|
Jomard C, Gondin J. Influence of sexual dimorphism on satellite cell regulation and inflammatory response during skeletal muscle regeneration. Physiol Rep 2023; 11:e15798. [PMID: 37798097 PMCID: PMC10555529 DOI: 10.14814/phy2.15798] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 10/07/2023] Open
Abstract
After injury, skeletal muscle regenerates thanks to the key role of satellite cells (SC). The regeneration process is supported and coordinated by other cell types among which immune cells. Among the mechanisms involved in skeletal muscle regeneration, a sexual dimorphism, involving sex hormones and more particularly estrogens, has been suggested. However, the role of sexual dimorphism on skeletal muscle regeneration is not fully understood, likely to the use of various experimental settings in both animals and human. This review aims at addressing how sex and estrogens regulate both the SC and the inflammatory response during skeletal muscle regeneration by considering the different experimental designs used in both animal models (i.e., ovarian hormone deficiency, estrogen replacement or supplementation, treatments with estrogen receptors agonists/antagonists and models knockout for estrogen receptors) and human (hormone therapy replacement, pre vs. postmenopausal, menstrual cycle variation…).
Collapse
Affiliation(s)
- Charline Jomard
- Institut NeuroMyoGène (INMG), Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Université Claude Bernard LyonLyonFrance
| | - Julien Gondin
- Institut NeuroMyoGène (INMG), Physiopathologie et Génétique du Neurone et du Muscle (PGNM), Université Claude Bernard LyonLyonFrance
| |
Collapse
|
67
|
Bouche A, Borner B, Richard C, Grand Y, Hannouche D, Laumonier T. In vitro-generated human muscle reserve cells are heterogeneous for Pax7 with distinct molecular states and metabolic profiles. Stem Cell Res Ther 2023; 14:243. [PMID: 37679820 PMCID: PMC10486062 DOI: 10.1186/s13287-023-03483-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND The capacity of skeletal muscles to regenerate relies on Pax7+ muscle stem cells (MuSC). While in vitro-amplified MuSC are activated and lose part of their regenerative capacity, in vitro-generated human muscle reserve cells (MuRC) are very similar to quiescent MuSC with properties required for their use in cell-based therapies. METHODS In the present study, we investigated the heterogeneity of human MuRC and characterized their molecular signature and metabolic profile. RESULTS We observed that Notch signaling is active and essential for the generation of quiescent human Pax7+ MuRC in vitro. We also revealed, by immunofluorescence and flow cytometry, two distinct subpopulations of MuRC distinguished by their relative Pax7 expression. After 48 h in differentiation medium (DM), the Pax7High subpopulation represented 35% of the total MuRC pool and this percentage increased to 61% after 96 h in DM. Transcriptomic analysis revealed that Pax7High MuRC were less primed for myogenic differentiation as compared to Pax7Low MuRC and displayed a metabolic shift from glycolysis toward fatty acid oxidation. The bioenergetic profile of human MuRC displayed a 1.5-fold decrease in glycolysis, basal respiration and ATP-linked respiration as compared to myoblasts. We also observed that AMPKα1 expression was significantly upregulated in human MuRC that correlated with an increased phosphorylation of acetyl-CoA carboxylase (ACC). Finally, we showed that fatty acid uptake was increased in MuRC as compared to myoblasts, whereas no changes were observed for glucose uptake. CONCLUSIONS Overall, these data reveal that the quiescent MuRC pool is heterogeneous for Pax7 with a Pax7High subpopulation being in a deeper quiescent state, less committed to differentiation and displaying a reduced metabolic activity. Altogether, our data suggest that human Pax7High MuRC may constitute an appropriate stem cell source for potential therapeutic applications in skeletal muscle diseases.
Collapse
Affiliation(s)
- Axelle Bouche
- Cell Therapy and Musculoskeletal Disorders Laboratory, Department of Orthopedic Surgery, Geneva University Hospitals and Faculty of Medicine, University Medical Center, 1 rue Michel Servet, 1211, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland
| | - Benoit Borner
- Cell Therapy and Musculoskeletal Disorders Laboratory, Department of Orthopedic Surgery, Geneva University Hospitals and Faculty of Medicine, University Medical Center, 1 rue Michel Servet, 1211, Geneva, Switzerland
| | - Chloé Richard
- Cell Therapy and Musculoskeletal Disorders Laboratory, Department of Orthopedic Surgery, Geneva University Hospitals and Faculty of Medicine, University Medical Center, 1 rue Michel Servet, 1211, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland
| | - Ysaline Grand
- Cell Therapy and Musculoskeletal Disorders Laboratory, Department of Orthopedic Surgery, Geneva University Hospitals and Faculty of Medicine, University Medical Center, 1 rue Michel Servet, 1211, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland
| | - Didier Hannouche
- Cell Therapy and Musculoskeletal Disorders Laboratory, Department of Orthopedic Surgery, Geneva University Hospitals and Faculty of Medicine, University Medical Center, 1 rue Michel Servet, 1211, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland
| | - Thomas Laumonier
- Cell Therapy and Musculoskeletal Disorders Laboratory, Department of Orthopedic Surgery, Geneva University Hospitals and Faculty of Medicine, University Medical Center, 1 rue Michel Servet, 1211, Geneva, Switzerland.
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland.
| |
Collapse
|
68
|
Hüttner SS, Henze H, Elster D, Koch P, Anderer U, von Eyss B, von Maltzahn J. A dysfunctional miR-1-TRPS1-MYOG axis drives ERMS by suppressing terminal myogenic differentiation. Mol Ther 2023; 31:2612-2632. [PMID: 37452493 PMCID: PMC10492030 DOI: 10.1016/j.ymthe.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/12/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
Rhabdomyosarcoma is the most common pediatric soft tissue tumor, comprising two major subtypes: the PAX3/7-FOXO1 fusion-negative embryonal and the PAX3/7-FOXO1 fusion-positive alveolar subtype. Here, we demonstrate that the expression levels of the transcriptional repressor TRPS1 are specifically enhanced in the embryonal subtype, resulting in impaired terminal myogenic differentiation and tumor growth. During normal myogenesis, expression levels of TRPS1 have to decrease to allow myogenic progression, as demonstrated by overexpression of TRPS1 in myoblasts impairing myotube formation. Consequentially, myogenic differentiation in embryonal rhabdomyosarcoma in vitro as well as in vivo can be achieved by reducing TRPS1 levels. Furthermore, we show that TRPS1 levels in RD cells, the bona fide model cell line for embryonal rhabdomyosarcoma, are regulated by miR-1 and that TRPS1 and MYOD1 share common genomic binding sites. The myogenin (MYOG) promoter is one of the critical targets of TRPS1 and MYOD1; we demonstrate that TRPS1 restricts MYOG expression and thereby inhibits terminal myogenic differentiation. Therefore, reduction of TRPS1 levels in embryonal rhabdomyosarcoma might be a therapeutic approach to drive embryonal rhabdomyosarcoma cells into myogenic differentiation, thereby generating postmitotic myotubes.
Collapse
Affiliation(s)
- Sören S Hüttner
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Henriette Henze
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Dana Elster
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Philipp Koch
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Ursula Anderer
- Department of Cell Biology and Tissue Engineering, Brandenburg University of Technology Cottbus-Senftenberg, Universitätsplatz 1, 01968 Senftenberg, Germany
| | - Björn von Eyss
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Julia von Maltzahn
- Leibniz Institute on Aging - Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany; Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Universitätsplatz 1, 01968 Senftenberg, Germany.
| |
Collapse
|
69
|
Brondolin M, Herzog D, Sultan S, Warburton F, Vigilante A, Knight RD. Migration and differentiation of muscle stem cells are coupled by RhoA signalling during regeneration. Open Biol 2023; 13:230037. [PMID: 37726092 PMCID: PMC10508982 DOI: 10.1098/rsob.230037] [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: 02/09/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023] Open
Abstract
Skeletal muscle is highly regenerative and is mediated by a population of migratory adult muscle stem cells (muSCs). Effective muscle regeneration requires a spatio-temporally regulated response of the muSC population to generate sufficient muscle progenitor cells that then differentiate at the appropriate time. The relationship between muSC migration and cell fate is poorly understood and it is not clear how forces experienced by migrating cells affect cell behaviour. We have used zebrafish to understand the relationship between muSC cell adhesion, behaviour and fate in vivo. Imaging of pax7-expressing muSCs as they respond to focal injuries in trunk muscle reveals that they migrate by protrusive-based means. By carefully characterizing their behaviour in response to injury we find that they employ an adhesion-dependent mode of migration that is regulated by the RhoA kinase ROCK. Impaired ROCK activity results in reduced expression of cell cycle genes and increased differentiation in regenerating muscle. This correlates with changes to focal adhesion dynamics and migration, revealing that ROCK inhibition alters the interaction of muSCs to their local environment. We propose that muSC migration and differentiation are coupled processes that respond to changes in force from the environment mediated by RhoA signalling.
Collapse
Affiliation(s)
- Mirco Brondolin
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, London SE1 9RT, UK
| | - Dylan Herzog
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, London SE1 9RT, UK
| | - Sami Sultan
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, London SE1 9RT, UK
| | - Fiona Warburton
- Oral Clinical Research Unit, King's College London, London, London SE1 9RT, UK
| | | | - Robert D. Knight
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, London SE1 9RT, UK
| |
Collapse
|
70
|
Gallagher H, Hendrickse PW, Pereira MG, Bowen TS. Skeletal muscle atrophy, regeneration, and dysfunction in heart failure: Impact of exercise training. JOURNAL OF SPORT AND HEALTH SCIENCE 2023; 12:557-567. [PMID: 37040849 PMCID: PMC10466197 DOI: 10.1016/j.jshs.2023.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/30/2022] [Accepted: 02/20/2023] [Indexed: 05/31/2023]
Abstract
This review highlights some established and some more contemporary mechanisms responsible for heart failure (HF)-induced skeletal muscle wasting and weakness. We first describe the effects of HF on the relationship between protein synthesis and degradation rates, which determine muscle mass, the involvement of the satellite cells for continual muscle regeneration, and changes in myofiber calcium homeostasis linked to contractile dysfunction. We then highlight key mechanistic effects of both aerobic and resistance exercise training on skeletal muscle in HF and outline its application as a beneficial treatment. Overall, HF causes multiple impairments related to autophagy, anabolic-catabolic signaling, satellite cell proliferation, and calcium homeostasis, which together promote fiber atrophy, contractile dysfunction, and impaired regeneration. Although both wasting and weakness are partly rescued by aerobic and resistance exercise training in HF, the effects of satellite cell dynamics remain poorly explored.
Collapse
Affiliation(s)
- Harrison Gallagher
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Paul W Hendrickse
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Marcelo G Pereira
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - T Scott Bowen
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
71
|
Tatsukawa T, Kano K, Nakajima KI, Yazawa T, Eguchi R, Kabara M, Horiuchi K, Hayasaka T, Matsuo R, Hasebe N, Azuma N, Kawabe JI. NG2-positive pericytes regulate homeostatic maintenance of slow-type skeletal muscle with rapid myonuclear turnover. Stem Cell Res Ther 2023; 14:205. [PMID: 37592340 PMCID: PMC10433572 DOI: 10.1186/s13287-023-03433-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Skeletal muscle comprises almost 40% of the human body and is essential for movement, structural support and metabolic homeostasis. Size of multinuclear skeletal muscle is stably maintained under steady conditions with the sporadic fusion of newly produced myocytes to compensate for the muscular turnover caused by daily wear and tear. It is becoming clear that microvascular pericytes (PCs) exhibit myogenic activity. However, whether PCs act as myogenic stem cells for the homeostatic maintenance of skeletal muscles during adulthood remains uncertain. METHODS We utilized PC-fused myofibers using PC-specific lineage tracing mouse (NG2-CreERT/Rosa-tdTomato) to observe whether muscle resident PCs have myogenic potential during daily life. Genetic PC deletion mouse model (NG2-CreERT/DTA) was used to test whether PC differentiates to myofibers for maintenance of muscle structure and function under homeostatic condition. RESULTS Under steady breeding conditions, tdTomato-expressing PCs were infused into myofibers, and subsequently, PC-derived nuclei were incorporated into myofibers. Especially in type-I slow-type myofibers such as the soleus, tdTomato+ myofibers were already observed 3 days after PC labeling; their ratio reached a peak (approximately 80%) within 1 month and was maintained for more than 1 year. Consistently, the NG2+ PC-specific deletion induced muscular atrophy in a slow-type myofiber-specific manner under steady breeding conditions. The number of myonucleus per volume of each myofiber was constant during observation period. CONCLUSIONS These findings demonstrate that the turnover of myonuclei in slow-type myofibers is relatively fast, with PCs acting as myogenic stem cells-the suppliers of new myonuclei under steady conditions-and play a vital role in the homeostatic maintenance of slow-type muscles.
Collapse
Affiliation(s)
- Takamitsu Tatsukawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kohei Kano
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kei-Ichi Nakajima
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Ryoji Eguchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Maki Kabara
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kiwamu Horiuchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Taiki Hayasaka
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Risa Matsuo
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Dermatology, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Naoyuki Hasebe
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Nobuyoshi Azuma
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Jun-Ichi Kawabe
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
| |
Collapse
|
72
|
Pizza FX, Buckley KH. Regenerating Myofibers after an Acute Muscle Injury: What Do We Really Know about Them? Int J Mol Sci 2023; 24:12545. [PMID: 37628725 PMCID: PMC10454182 DOI: 10.3390/ijms241612545] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Injury to skeletal muscle through trauma, physical activity, or disease initiates a process called muscle regeneration. When injured myofibers undergo necrosis, muscle regeneration gives rise to myofibers that have myonuclei in a central position, which contrasts the normal, peripheral position of myonuclei. Myofibers with central myonuclei are called regenerating myofibers and are the hallmark feature of muscle regeneration. An important and underappreciated aspect of muscle regeneration is the maturation of regenerating myofibers into a normal sized myofiber with peripheral myonuclei. Strikingly, very little is known about processes that govern regenerating myofiber maturation after muscle injury. As knowledge of myofiber formation and maturation during embryonic, fetal, and postnatal development has served as a foundation for understanding muscle regeneration, this narrative review discusses similarities and differences in myofiber maturation during muscle development and regeneration. Specifically, we compare and contrast myonuclear positioning, myonuclear accretion, myofiber hypertrophy, and myofiber morphology during muscle development and regeneration. We also discuss regenerating myofibers in the context of different types of myofiber necrosis (complete and segmental) after muscle trauma and injurious contractions. The overall goal of the review is to provide a framework for identifying cellular and molecular processes of myofiber maturation that are unique to muscle regeneration.
Collapse
Affiliation(s)
- Francis X. Pizza
- Department of Exercise and Rehabilitation Sciences, University of Toledo, Toledo, OH 43606, USA
| | - Kole H. Buckley
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA;
| |
Collapse
|
73
|
Martins L, Amorim WW, Gregnani MF, de Carvalho Araújo R, Qadri F, Bader M, Pesquero JB. Kinin receptors regulate skeletal muscle regeneration: differential effects for B1 and B2 receptors. Inflamm Res 2023; 72:1583-1601. [PMID: 37464053 PMCID: PMC10499706 DOI: 10.1007/s00011-023-01766-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/20/2023] [Accepted: 07/02/2023] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVE AND DESIGN After traumatic skeletal muscle injury, muscle healing is often incomplete and produces extensive fibrosis. Bradykinin (BK) reduces fibrosis in renal and cardiac damage models through the B2 receptor. The B1 receptor expression is induced by damage, and blocking of the kallikrein-kinin system seems to affect the progression of muscular dystrophy. We hypothesized that both kinin B1 and B2 receptors could play a differential role after traumatic muscle injury, and the lack of the B1 receptor could produce more cellular and molecular substrates for myogenesis and fewer substrates for fibrosis, leading to better muscle healing. MATERIAL AND METHODS To test this hypothesis, tibialis anterior muscles of kinin receptor knockout animals were subjected to traumatic injury. Myogenesis, angiogenesis, fibrosis, and muscle functioning were evaluated. RESULTS Injured B1KO mice showed a faster healing progression of the injured area with a larger amount of central nucleated fiber post-injury when compared to control mice. In addition, they exhibited higher neovasculogenic capacity, maintaining optimal tissue perfusion for the post-injury phase; had higher amounts of myogenic markers with less inflammatory infiltrate and tissue destruction. This was followed by higher amounts of SMAD7 and lower amounts of p-SMAD2/3, which resulted in less fibrosis. In contrast, B2KO and B1B2KO mice showed more severe tissue destruction and excessive fibrosis. B1KO animals had better results in post-injury functional tests compared to control animals. CONCLUSIONS We demonstrate that injured skeletal muscle tissues have a better repair capacity with less fibrosis in the presence of B2 receptor and absence of B1 receptor, including better performances in functional tests.
Collapse
Affiliation(s)
- Leonardo Martins
- Division of Medical Sciences, Laboratory of Transcriptional Regulation, Institute of Medical Biology of Polish Academy of Sciences (IMB-PAN), 3a Tylna St., 90-364, Łódź, Poland.
- Center for Research and Molecular Diagnosis of Genetic Diseases, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil.
- Department of Biochemistry and Molecular Biology, Federal University of São Paulo, Rua Três de Maio 100, 4th Floor, São Paulo, 04044-020, Brazil.
| | - Weslley Wallace Amorim
- Center for Research and Molecular Diagnosis of Genetic Diseases, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil
| | - Marcos Fernandes Gregnani
- Laboratory of Exercise Genetics and Metabolism, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil
| | - Ronaldo de Carvalho Araújo
- Laboratory of Exercise Genetics and Metabolism, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil
| | - Fatimunnisa Qadri
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Robert-Rössle-Str. 10, 13125, Berlin, Germany
- Institute for Biology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
- Charité University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Potsdamer Str. 58, 10785, Berlin, Germany
| | - João Bosco Pesquero
- Center for Research and Molecular Diagnosis of Genetic Diseases, Federal University of São Paulo, Rua Pedro de Toledo 669, 9th Floor, São Paulo, 04039032, Brazil.
- Department of Biophysics, Federal University of São Paulo, Rua Botucatu 862, 6th Floor, São Paulo, 04023-062, Brazil.
| |
Collapse
|
74
|
Govindappa PK, Jagadeeshaprasad MG, Tortora P, Talukder MAH, Elfar JC. Effects of 4-Aminopyridine on Combined Nerve and Muscle Injury and Bone Loss. J Hand Surg Am 2023; 48:831.e1-831.e9. [PMID: 35418340 PMCID: PMC9548524 DOI: 10.1016/j.jhsa.2022.01.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 12/08/2021] [Accepted: 01/21/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE Musculoskeletal injuries are common, and peripheral nerve injury (PNI) causes significant muscle and bone loss within weeks. After PNI, 4-aminopyridine (4-AP) improves functional recovery and muscle atrophy. However, it is unknown whether 4-AP has any effect on isolated traumatic muscle injury and PNI-induced bone loss. METHODS A standardized crush injury was performed on the sciatic nerve and muscles in mice, and the mice were assigned to receive normal saline or 4-AP treatment daily for 21 days. The postinjury motor and sensory function recovery was assessed, injured muscles were processed for histomorphometry, and the tibial bone was scanned for bone density. RESULTS 4-Aminopyridine significantly accelerated the postinjury motor and sensory function recovery, improved muscle histomorphometry, increased muscle satellite cell numbers, and shifted muscle fiber types after combined nerve and muscle injury. Importantly, the 4-AP treatment significantly reduced PNI-induced bone loss. In contrast, in the case of isolated muscle injury, 4-AP had no effect on functional recovery and bone density, but it improved muscle-specific histomorphometry to a limited extent. CONCLUSIONS These findings demonstrate the potential beneficial effects of 4-AP on the recovery of muscle morphology and bone density after combined muscle and nerve injury. CLINICAL RELEVANCE Nerve injuries frequently involve muscle and result in rapid muscle and bone atrophy. In this scenario, 4-AP, in addition to accelerating nerve functional recovery, might work as an adjunctive agent to improve the recovery of injured muscle and attenuate PNI-induced bone loss.
Collapse
Affiliation(s)
- Prem Kumar Govindappa
- Department of Orthopaedics and Rehabilitation, Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Mashanipalya G Jagadeeshaprasad
- Department of Orthopaedics and Rehabilitation, Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Peter Tortora
- Department of Orthopaedics and Rehabilitation, Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - M A Hassan Talukder
- Department of Orthopaedics and Rehabilitation, Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - John C. Elfar
- Department of Orthopaedics and Rehabilitation, Center for Orthopaedic Research and Translational Science (CORTS), The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
75
|
Johnson AL, Kamal M, Parise G. The Role of Supporting Cell Populations in Satellite Cell Mediated Muscle Repair. Cells 2023; 12:1968. [PMID: 37566047 PMCID: PMC10417507 DOI: 10.3390/cells12151968] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
Skeletal muscle has a high capacity to repair and remodel in response to damage, largely through the action of resident muscle stem cells, termed satellite cells. Satellite cells are required for the proper repair of skeletal muscle through a process known as myogenesis. Recent investigations have observed relationships between satellite cells and other cell types and structures within the muscle microenvironment. These findings suggest that the crosstalk between inflammatory cells, fibrogenic cells, bone-marrow-derived cells, satellite cells, and the vasculature is essential for the restoration of muscle homeostasis. This review will discuss the influence of the cells and structures within the muscle microenvironment on satellite cell function and muscle repair.
Collapse
Affiliation(s)
| | | | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
76
|
Bathina S, Armamento-Villareal R. The complex pathophysiology of bone fragility in obesity and type 2 diabetes mellitus: therapeutic targets to promote osteogenesis. Front Endocrinol (Lausanne) 2023; 14:1168687. [PMID: 37576965 PMCID: PMC10422976 DOI: 10.3389/fendo.2023.1168687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/05/2023] [Indexed: 08/15/2023] Open
Abstract
Fractures associated with Type2 diabetes (T2DM) are major public health concerns in an increasingly obese and aging population. Patients with obesity or T2DM have normal or better than normal bone mineral density but at an increased risk for fractures. Hence it is crucial to understand the pathophysiology and mechanism of how T2DM and obesity result in altered bone physiology leading to increased fracture risk. Although enhanced osteoclast mediated bone resorption has been reported for these patients, the most notable observation among patients with T2DM is the reduction in bone formation from mostly dysfunction in osteoblast differentiation and survival. Studies have shown that obesity and T2DM are associated with increased adipogenesis which is most likely at the expense of reduced osteogenesis and myogenesis considering that adipocytes, osteoblasts, and myoblasts originate from the same progenitor cells. Furthermore, emerging data point to an inter-relationship between bone and metabolic homeostasis suggesting that these physiologic processes could be under the control of common regulatory pathways. Thus, this review aims to explore the complex mechanisms involved in lineage differentiation and their effect on bone pathophysiology in patients with obesity and T2DM along with an examination of potential novel pharmacological targets or a re-evaluation of existing drugs to improve bone homeostasis.
Collapse
Affiliation(s)
- Siresha Bathina
- Division of Endocrinology Diabetes and Metabolism, Baylor College of Medicine, Houston, TX, United States
- Center for Translational Research on Inflammatory Disease, Michael E. DeBakey Veterans Affairs (VA) Medical Center, Houston, TX, United States
| | - Reina Armamento-Villareal
- Division of Endocrinology Diabetes and Metabolism, Baylor College of Medicine, Houston, TX, United States
- Center for Translational Research on Inflammatory Disease, Michael E. DeBakey Veterans Affairs (VA) Medical Center, Houston, TX, United States
| |
Collapse
|
77
|
Song C, Hu Z, Xu D, Bian H, Lv J, Zhu X, Zhang Q, Su L, Yin H, Lu T, Li Y. STING signaling in inflammaging: a new target against musculoskeletal diseases. Front Immunol 2023; 14:1227364. [PMID: 37492580 PMCID: PMC10363987 DOI: 10.3389/fimmu.2023.1227364] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Stimulator of Interferon Gene (STING) is a critical signaling linker protein that plays a crucial role in the intrinsic immune response, particularly in the cytoplasmic DNA-mediated immune response in both pathogens and hosts. It is also involved in various signaling processes in vivo. The musculoskeletal system provides humans with morphology, support, stability, and movement. However, its aging can result in various diseases and negatively impact people's lives. While many studies have reported that cellular aging is a leading cause of musculoskeletal disorders, it also offers insight into potential treatments. Under pathological conditions, senescent osteoblasts, chondrocytes, myeloid cells, and muscle fibers exhibit persistent senescence-associated secretory phenotype (SASP), metabolic disturbances, and cell cycle arrest, which are closely linked to abnormal STING activation. The accumulation of cytoplasmic DNA due to chromatin escape from the nucleus following DNA damage or telomere shortening activates the cGAS-STING signaling pathway. Moreover, STING activation is also linked to mitochondrial dysfunction, epigenetic modifications, and impaired cytoplasmic DNA degradation. STING activation upregulates SASP and autophagy directly and indirectly promotes cell cycle arrest. Thus, STING may be involved in the onset and development of various age-related musculoskeletal disorders and represents a potential therapeutic target. In recent years, many STING modulators have been developed and used in the study of musculoskeletal disorders. Therefore, this paper summarizes the effects of STING signaling on the musculoskeletal system at the molecular level and current understanding of the mechanisms of endogenous active ligand production and accumulation. We also discuss the relationship between some age-related musculoskeletal disorders and STING, as well as the current status of STING modulator development.
Collapse
Affiliation(s)
- Chenyu Song
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Zhuoyi Hu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Dingjun Xu
- Department of Orthopaedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Zhejiang, China
| | - Huihui Bian
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Juan Lv
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xuanxuan Zhu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Qiang Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Li Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Heng Yin
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Tong Lu
- Department of Critical Care Medicine, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Yinghua Li
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
78
|
Picca A, Lozanoska-Ochser B, Calvani R, Coelho-Júnior HJ, Leewenburgh C, Marzetti E. Inflammatory, mitochondrial, and senescence-related markers: Underlying biological pathways of muscle aging and new therapeutic targets. Exp Gerontol 2023; 178:112204. [PMID: 37169101 DOI: 10.1016/j.exger.2023.112204] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/13/2023]
Abstract
The maintenance of functional health is pivotal for achieving independent life in older age. The aged muscle is characterized by ultrastructural changes, including loss of type I and type II myofibers and a greater proportion of cytochrome c oxidase deficient and succinate dehydrogenase positive fibers. Both intrinsic (e.g., altered proteostasis, DNA damage, and mitochondrial dysfunction) and extrinsic factors (e.g., denervation, altered metabolic regulation, declines in satellite cells, and inflammation) contribute to muscle aging. Being a hub for several cellular activities, mitochondria are key to myocyte viability and mitochondrial dysfunction has been implicated in age-associated physical decline. The maintenance of functional organelles via mitochondrial quality control (MQC) processes is, therefore, crucial to skeletal myofiber viability and organismal health. The autophagy-lysosome pathway has emerged as a critical step of MQC in muscle by disposing organelles and proteins via their tagging for autophagosome incorporation and delivery to the lysosome for clearance. This pathway was found to be altered in muscle of physically inactive older adults. A relationship between this pathway and muscle tissue composition of the lower extremities as well as physical performance was also identified. Therefore, integrating muscle structure and myocyte quality control measures in the evaluation of muscle health may be a promising strategy for devising interventions fostering muscle health.
Collapse
Affiliation(s)
- Anna Picca
- Department of Medicine and Surgery, LUM University, Casamassima, 70100 Bari, Italy; Fondazione Policlinico Universitario "Agostino Gemelli" IRCSS, 00168 Rome, Italy
| | - Biliana Lozanoska-Ochser
- Department of Medicine and Surgery, LUM University, Casamassima, 70100 Bari, Italy; DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCSS, 00168 Rome, Italy; Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Hélio José Coelho-Júnior
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCSS, 00168 Rome, Italy; Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
79
|
Norris AM, Appu AB, Johnson CD, Zhou LY, McKellar DW, Renault MA, Hammers D, Cosgrove BD, Kopinke D. Hedgehog signaling via its ligand DHH acts as cell fate determinant during skeletal muscle regeneration. Nat Commun 2023; 14:3766. [PMID: 37355632 PMCID: PMC10290686 DOI: 10.1038/s41467-023-39506-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
Successful muscle regeneration relies on the interplay of multiple cell populations. However, the signals required for this coordinated intercellular crosstalk remain largely unknown. Here, we describe how the Hedgehog (Hh) signaling pathway controls the fate of fibro/adipogenic progenitors (FAPs), the cellular origin of intramuscular fat (IMAT) and fibrotic scar tissue. Using conditional mutagenesis and pharmacological Hh modulators in vivo and in vitro, we identify DHH as the key ligand that acts as a potent adipogenic brake by preventing the adipogenic differentiation of FAPs. Hh signaling also impacts muscle regeneration, albeit indirectly through induction of myogenic factors in FAPs. Our results also indicate that ectopic and sustained Hh activation forces FAPs to adopt a fibrogenic fate resulting in widespread fibrosis. In this work, we reveal crucial post-developmental functions of Hh signaling in balancing tissue regeneration and fatty fibrosis. Moreover, they provide the exciting possibility that mis-regulation of the Hh pathway with age and disease could be a major driver of pathological IMAT formation.
Collapse
Affiliation(s)
- Alessandra M Norris
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Ambili Bai Appu
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Connor D Johnson
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Lylybell Y Zhou
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - David W McKellar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Marie-Ange Renault
- Biology of Cardiovascular Diseases, INSERM, University of Bordeaux, Pessac, France
| | - David Hammers
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Benjamin D Cosgrove
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
80
|
Pang KT, Loo LSW, Chia S, Ong FYT, Yu H, Walsh I. Insight into muscle stem cell regeneration and mechanobiology. Stem Cell Res Ther 2023; 14:129. [PMID: 37173707 PMCID: PMC10176686 DOI: 10.1186/s13287-023-03363-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Stem cells possess the unique ability to differentiate into specialized cell types. These specialized cell types can be used for regenerative medicine purposes such as cell therapy. Myosatellite cells, also known as skeletal muscle stem cells (MuSCs), play important roles in the growth, repair, and regeneration of skeletal muscle tissues. However, despite its therapeutic potential, the successful differentiation, proliferation, and expansion processes of MuSCs remain a significant challenge due to a variety of factors. For example, the growth and differentiation of MuSCs can be greatly influenced by actively replicating the MuSCs microenvironment (known as the niche) using mechanical forces. However, the molecular role of mechanobiology in MuSC growth, proliferation, and differentiation for regenerative medicine is still poorly understood. In this present review, we comprehensively summarize, compare, and critically analyze how different mechanical cues shape stem cell growth, proliferation, differentiation, and their potential role in disease development (Fig. 1). The insights developed from the mechanobiology of stem cells will also contribute to how these applications can be used for regenerative purposes using MuSCs.
Collapse
Affiliation(s)
- Kuin Tian Pang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.
- School of Chemistry, Chemical Engineering, and Biotechnology, Nanyang Technology University, 62 Nanyang Drive, N1.2-B3, Singapore, 637459, Singapore.
| | - Larry Sai Weng Loo
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sean Chia
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Francesca Yi Teng Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hanry Yu
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
- Interdisplinary Science and Engineering Program, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - Ian Walsh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
81
|
Luo L, Chua YJB, Liu T, Liang K, Chua MWJ, Ma W, Goh JW, Wang Y, Su J, Ho YS, Li CW, Liu KH, Teh BT, Yu K, Shyh-Chang N. Muscle Injuries Induce a Prostacyclin-PPARγ/PGC1a-FAO Spike That Boosts Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2301519. [PMID: 37140179 PMCID: PMC10375192 DOI: 10.1002/advs.202301519] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/14/2023] [Indexed: 05/05/2023]
Abstract
It is well-known that muscle regeneration declines with aging, and aged muscles undergo degenerative atrophy or sarcopenia. While exercise and acute injury are both known to induce muscle regeneration, the molecular signals that help trigger muscle regeneration have remained unclear. Here, mass spectrometry imaging (MSI) is used to show that injured muscles induce a specific subset of prostanoids during regeneration, including PGG1, PGD2, and the prostacyclin PGI2. The spike in prostacyclin promotes skeletal muscle regeneration via myoblasts, and declines with aging. Mechanistically, the prostacyclin spike promotes a spike in PPARγ/PGC1a signaling, which induces a spike in fatty acid oxidation (FAO) to control myogenesis. LC-MS/MS and MSI further confirm that an early FAO spike is associated with normal regeneration, but muscle FAO became dysregulated during aging. Functional experiments demonstrate that the prostacyclin-PPARγ/PGC1a-FAO spike is necessary and sufficient to promote both young and aged muscle regeneration, and that prostacyclin can synergize with PPARγ/PGC1a-FAO signaling to restore aged muscles' regeneration and physical function. Given that the post-injury prostacyclin-PPARγ-FAO spike can be modulated pharmacologically and via post-exercise nutrition, this work has implications for how prostacyclin-PPARγ-FAO might be fine-tuned to promote regeneration and treat muscle diseases of aging.
Collapse
Affiliation(s)
- Lanfang Luo
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yan-Jiang Benjamin Chua
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore City, 119077, Singapore
- Genome Institute of Singapore, Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore City, 138672, Singapore
| | - Taoyan Liu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Kun Liang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Min-Wen Jason Chua
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore City, 119077, Singapore
- Genome Institute of Singapore, Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore City, 138672, Singapore
| | - Wenwu Ma
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jun-Wei Goh
- Genome Institute of Singapore, Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Singapore City, 138672, Singapore
| | - Yuefan Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jiali Su
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ying Swan Ho
- Bioprocessing Technology Institute, Agency for Science Technology and Research, Singapore City, 138668, Singapore
| | - Chun-Wei Li
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P. R. China
| | - Ke Hui Liu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Bin Tean Teh
- Laboratory of Cancer Therapeutics, Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore City, 169857, Singapore
- Division of Medical Science, Laboratory of Cancer Epigenome, National Cancer Centre Singapore, Singapore City, 119074, Singapore
| | - Kang Yu
- Department of Clinical Nutrition, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, P. R. China
| | - Ng Shyh-Chang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
82
|
Lim S, Lee DE, Morena da Silva F, Koopmans PJ, Vechetti IJ, von Walden F, Greene NP, Murach KA. MicroRNA control of the myogenic cell transcriptome and proteome: the role of miR-16. Am J Physiol Cell Physiol 2023; 324:C1101-C1109. [PMID: 36971422 PMCID: PMC10191132 DOI: 10.1152/ajpcell.00071.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
MicroRNAs (miRs) control stem cell biology and fate. Ubiquitously expressed and conserved miR-16 was the first miR implicated in tumorigenesis. miR-16 is low in muscle during developmental hypertrophy and regeneration. It is enriched in proliferating myogenic progenitor cells but is repressed during differentiation. The induction of miR-16 blocks myoblast differentiation and myotube formation, whereas knockdown enhances these processes. Despite a central role for miR-16 in myogenic cell biology, how it mediates its potent effects is incompletely defined. In this investigation, global transcriptomic and proteomic analyses after miR-16 knockdown in proliferating C2C12 myoblasts revealed how miR-16 influences myogenic cell fate. Eighteen hours after miR-16 inhibition, ribosomal protein gene expression levels were higher relative to control myoblasts and p53 pathway-related gene abundance was lower. At the protein level at this same time point, miR-16 knockdown globally upregulated tricarboxylic acid (TCA) cycle proteins while downregulating RNA metabolism-related proteins. miR-16 inhibition induced specific proteins associated with myogenic differentiation such as ACTA2, EEF1A2, and OPA1. We extend prior work in hypertrophic muscle tissue and show that miR-16 is lower in mechanically overloaded muscle in vivo. Our data collectively point to how miR-16 is implicated in aspects of myogenic cell differentiation. A deeper understanding of the role of miR-16 in myogenic cells has consequences for muscle developmental growth, exercise-induced hypertrophy, and regenerative repair after injury, all of which involve myogenic progenitors.
Collapse
Affiliation(s)
- Seongkyun Lim
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - David E Lee
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Francielly Morena da Silva
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Pieter J Koopmans
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ivan J Vechetti
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States
| | - Ferdinand von Walden
- Neuropediatrics, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas P Greene
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, United States
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, United States
| |
Collapse
|
83
|
de Morree A, Rando TA. Regulation of adult stem cell quiescence and its functions in the maintenance of tissue integrity. Nat Rev Mol Cell Biol 2023; 24:334-354. [PMID: 36922629 PMCID: PMC10725182 DOI: 10.1038/s41580-022-00568-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 03/18/2023]
Abstract
Adult stem cells are important for mammalian tissues, where they act as a cell reserve that supports normal tissue turnover and can mount a regenerative response following acute injuries. Quiescent stem cells are well established in certain tissues, such as skeletal muscle, brain, and bone marrow. The quiescent state is actively controlled and is essential for long-term maintenance of stem cell pools. In this Review, we discuss the importance of maintaining a functional pool of quiescent adult stem cells, including haematopoietic stem cells, skeletal muscle stem cells, neural stem cells, hair follicle stem cells, and mesenchymal stem cells such as fibro-adipogenic progenitors, to ensure tissue maintenance and repair. We discuss the molecular mechanisms that regulate the entry into, maintenance of, and exit from the quiescent state in mice. Recent studies revealed that quiescent stem cells have a discordance between RNA and protein levels, indicating the importance of post-transcriptional mechanisms, such as alternative polyadenylation, alternative splicing, and translation repression, in the control of stem cell quiescence. Understanding how these mechanisms guide stem cell function during homeostasis and regeneration has important implications for regenerative medicine.
Collapse
Affiliation(s)
- Antoine de Morree
- Department of Neurology and Neurological Science, Stanford University School of Medicine, Stanford, CA, USA.
- Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | - Thomas A Rando
- Department of Neurology and Neurological Science, Stanford University School of Medicine, Stanford, CA, USA.
- Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.
- Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
- Broad Stem Cell Research Center, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
84
|
Hanna BS, Wang G, Galván-Peña S, Mann AO, Ramirez RN, Muñoz-Rojas AR, Smith K, Wan M, Benoist C, Mathis D. The gut microbiota promotes distal tissue regeneration via RORγ + regulatory T cell emissaries. Immunity 2023; 56:829-846.e8. [PMID: 36822206 PMCID: PMC10101925 DOI: 10.1016/j.immuni.2023.01.033] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 12/22/2022] [Accepted: 01/30/2023] [Indexed: 02/24/2023]
Abstract
Specific microbial signals induce the differentiation of a distinct pool of RORγ+ regulatory T (Treg) cells crucial for intestinal homeostasis. We discovered highly analogous populations of microbiota-dependent Treg cells that promoted tissue regeneration at extra-gut sites, notably acutely injured skeletal muscle and fatty liver. Inflammatory meditators elicited by tissue damage combined with MHC-class-II-dependent T cell activation to drive the accumulation of gut-derived RORγ+ Treg cells in injured muscle, wherein they regulated the dynamics and tenor of early inflammation and helped balance the proliferation vs. differentiation of local stem cells. Reining in IL-17A-producing T cells was a major mechanism underlying the rheostatic functions of RORγ+ Treg cells in compromised tissues. Our findings highlight the importance of gut-trained Treg cell emissaries in controlling the response to sterile injury of non-mucosal tissues.
Collapse
Affiliation(s)
- Bola S Hanna
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Gang Wang
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Silvia Galván-Peña
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alexander O Mann
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ricardo N Ramirez
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Andrés R Muñoz-Rojas
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kathleen Smith
- Internal Medicine Research Unit, Worldwide Research, Development & Medical, Pfizer Inc., Cambridge, MA, USA
| | - Min Wan
- Internal Medicine Research Unit, Worldwide Research, Development & Medical, Pfizer Inc., Cambridge, MA, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
85
|
Kotsaris G, Qazi TH, Bucher CH, Zahid H, Pöhle-Kronawitter S, Ugorets V, Jarassier W, Börno S, Timmermann B, Giesecke-Thiel C, Economides AN, Le Grand F, Vallecillo-García P, Knaus P, Geissler S, Stricker S. Odd skipped-related 1 controls the pro-regenerative response of fibro-adipogenic progenitors. NPJ Regen Med 2023; 8:19. [PMID: 37019910 PMCID: PMC10076435 DOI: 10.1038/s41536-023-00291-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 03/17/2023] [Indexed: 04/07/2023] Open
Abstract
Skeletal muscle regeneration requires the coordinated interplay of diverse tissue-resident- and infiltrating cells. Fibro-adipogenic progenitors (FAPs) are an interstitial cell population that provides a beneficial microenvironment for muscle stem cells (MuSCs) during muscle regeneration. Here we show that the transcription factor Osr1 is essential for FAPs to communicate with MuSCs and infiltrating macrophages, thus coordinating muscle regeneration. Conditional inactivation of Osr1 impaired muscle regeneration with reduced myofiber growth and formation of excessive fibrotic tissue with reduced stiffness. Osr1-deficient FAPs acquired a fibrogenic identity with altered matrix secretion and cytokine expression resulting in impaired MuSC viability, expansion and differentiation. Immune cell profiling suggested a novel role for Osr1-FAPs in macrophage polarization. In vitro analysis suggested that increased TGFβ signaling and altered matrix deposition by Osr1-deficient FAPs actively suppressed regenerative myogenesis. In conclusion, we show that Osr1 is central to FAP function orchestrating key regenerative events such as inflammation, matrix secretion and myogenesis.
Collapse
Affiliation(s)
- Georgios Kotsaris
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
| | - Taimoor H Qazi
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Bioengineering, University of Pennsylvania, 19104, Philadelphia, USA
- Weldon School of Biomedical Engineering, Purdue University, 47907, West Lafayette, IN, USA
| | - Christian H Bucher
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117, Berlin, Germany
| | - Hafsa Zahid
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
- International Max Planck Research School for Biology and Computing IMPRS-BAC, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Ihnestrasse 73, 14195, Berlin, Germany
| | - Sophie Pöhle-Kronawitter
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Vladimir Ugorets
- Institute of Chemistry and Biochemistry, Cell Signaling Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - William Jarassier
- Institut NeuroMyoGène, CNRS UMR 5261, Inserm U1315, Université Claude Bernard Lyon 1, 69008, Lyon, France
| | - Stefan Börno
- Max Planck Institute for Molecular Genetics, Ihnestrasse 73, 14195, Berlin, Germany
| | - Bernd Timmermann
- Max Planck Institute for Molecular Genetics, Ihnestrasse 73, 14195, Berlin, Germany
| | | | | | - Fabien Le Grand
- Institut NeuroMyoGène, CNRS UMR 5261, Inserm U1315, Université Claude Bernard Lyon 1, 69008, Lyon, France
| | - Pedro Vallecillo-García
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Petra Knaus
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- Institute of Chemistry and Biochemistry, Cell Signaling Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Sven Geissler
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Julius Wolff Institute, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117, Berlin, Germany
- Berlin Center for Advanced Therapies (BECAT), Charité Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin, Germany
| | - Sigmar Stricker
- Institute of Chemistry and Biochemistry, Musculoskeletal Development and Regeneration Group, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany.
- Berlin-Brandenburg School for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany.
| |
Collapse
|
86
|
Yeh CJ, Sattler KM, Lepper C. Molecular regulation of satellite cells via intercellular signaling. Gene 2023; 858:147172. [PMID: 36621659 PMCID: PMC9928918 DOI: 10.1016/j.gene.2023.147172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/21/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
Somatic stem cells are tissue-specific reserve cells tasked to sustain tissue homeostasis in adulthood and/or effect tissue regeneration after traumatic injury. The stem cells of skeletal muscle tissue are the satellite cells, which were originally described and named after their localization beneath the muscle fiber lamina and attached to the multi-nucleated muscle fibers. During adult homeostasis, satellite cells are maintained in quiescence, a state of reversible cell cycle arrest. Yet, upon injury, satellite cells are rapidly activated, becoming highly mitotically active to generate large numbers of myoblasts that differentiate and fuse to regenerate the injured muscle fibers. A subset self-renews to replenish the pool of muscle stem cells.Complex intrinsic gene regulatory networks maintain the quiescent state of satellite cells, or upon injury, direct their activation, proliferation, differentiation and self-renewal. Molecular cues from the satellite cells' environment provide the essential information as to when and where satellite cells are to stay quiescent or break quiescence and effect regenerative myogenesis. Predominantly, these cues are secreted, diffusible or membrane-bound ligands that bind to and activate their specific cognate receptors on the satellite cell to activate downstream signaling cascades and elicit context-specific cell behavior. This review aims to offer a concise overview of major intercellular signaling pathways regulating satellite cells during quiescence and in injury-induced skeletal muscle regeneration.
Collapse
Affiliation(s)
- Chung-Ju Yeh
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States
| | - Kristina M Sattler
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States
| | - Christoph Lepper
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
87
|
Hanna BS, Yaghi OK, Langston PK, Mathis D. The potential for Treg-enhancing therapies in tissue, in particular skeletal muscle, regeneration. Clin Exp Immunol 2023; 211:138-148. [PMID: 35972909 PMCID: PMC10019136 DOI: 10.1093/cei/uxac076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/29/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Foxp3+CD4+ regulatory T cells (Tregs) are famous for their role in maintaining immunological tolerance. With their distinct transcriptomes, growth-factor dependencies and T-cell receptor (TCR) repertoires, Tregs in nonlymphoid tissues, termed "tissue-Tregs," also perform a variety of functions to help assure tissue homeostasis. For example, they are important for tissue repair and regeneration after various types of injury, both acute and chronic. They exert this influence by controlling both the inflammatory tenor and the dynamics of the parenchymal progenitor-cell pool in injured tissues, thereby promoting efficient repair and limiting fibrosis. Thus, tissue-Tregs are seemingly attractive targets for immunotherapy in the context of tissue regeneration, offering several advantages over existing therapies. Using skeletal muscle as a model system, we discuss the existing literature on Tregs' role in tissue regeneration in acute and chronic injuries, and various approaches for their therapeutic modulation in such contexts, including exercise as a natural Treg modulator.
Collapse
Affiliation(s)
- Bola S Hanna
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| | - Omar K Yaghi
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| | - P Kent Langston
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital; Boston, USA
| |
Collapse
|
88
|
Effect of heme oxygenase-1 on the differentiation of human myoblasts and the regeneration of murine skeletal muscles after acute and chronic injury. Pharmacol Rep 2023; 75:397-410. [PMID: 36918494 DOI: 10.1007/s43440-023-00475-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/20/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Impaired muscle regeneration is a hallmark of Duchenne muscular dystrophy (DMD), a neuromuscular disorder caused by mutations in the DMD gene encoding dystrophin. The lack of heme oxygenase-1 (HO-1, Hmox1), a known anti-inflammatory and cytoprotective enzyme, was shown to aggravate DMD pathology. METHODS We evaluated the role of HO-1 overexpression in human induced pluripotent stem cell (hiPSC)-derived skeletal muscle cells (hiPSC-SkM) in vitro and in the regeneration process in vivo in wild-type mice. Furthermore, the effect of cobalt protoporphyrin IX (CoPP), a pharmacological inducer of HO-1 expression, on regeneration markers during myogenic hiPSC differentiation and progression of the dystrophic phenotype was analysed in the mdx mouse DMD model. RESULTS HO-1 has an impact on hiPSC-SkM generation by decreasing cell fusion capacity and the expression of myogenic regulatory factors and muscle-specific microRNAs (myomiRs). Also, strong induction of HO-1 by CoPP totally abolished hiPSC-SkM differentiation. Injection of HO-1-overexpressing hiPSC-SkM into the cardiotoxin (CTX)-injured muscle of immunodeficient wild-type mice was associated with decreased expression of miR-206 and Myh3 and lower number of regenerating fibers, suggesting some advanced regeneration. However, the very potent induction of HO-1 by CoPP did not exert any protective effect on necrosis, leukocyte infiltration, fibrosis, myofiber regeneration biomarkers, and exercise capacity of mdx mice. CONCLUSIONS In summary, HO-1 inhibits the expression of differentiation markers in human iPSC-derived myoblasts. Although moderate overexpression of HO-1 in the injected myoblast was associated with partially advanced muscle regeneration, the high systemic induction of HO-1 did not improve muscle regeneration. The appropriate threshold of HO-1 expression must be established for the therapeutic effect of HO-1 on muscle regeneration.
Collapse
|
89
|
Müthel S, Marg A, Ignak B, Kieshauer J, Escobar H, Stadelmann C, Spuler S. Cas9-induced single cut enables highly efficient and template-free repair of a muscular dystrophy causing founder mutation. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:494-511. [PMID: 36865086 PMCID: PMC9972404 DOI: 10.1016/j.omtn.2023.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/01/2023] [Indexed: 02/07/2023]
Abstract
With thousands of patients worldwide, CAPN3 c.550delA is the most frequent mutation causing severe, progressive, and untreatable limb girdle muscular dystrophy. We aimed to genetically correct this founder mutation in primary human muscle stem cells. We designed editing strategies providing CRISPR-Cas9 as plasmid and mRNA first in patient-derived induced pluripotent stem cells and applied this strategy then in primary human muscle stem cells from patients. Mutation-specific targeting yielded highly efficient and precise correction of CAPN3 c.550delA to wild type for both cell types. Most likely a single cut generated by SpCas9 resulted in a 5' staggered overhang of one base pair, which triggered an overhang-dependent base replication of an A:T at the mutation site. This recovered the open reading frame and the CAPN3 DNA sequence was repaired template-free to wild type, which led to CAPN3 mRNA and protein expression. Off-target analysis using amplicon sequencing of 43 in silico predicted sites demonstrates the safety of this approach. Our study extends previous usage of single cut DNA modification since our gene product has been repaired into the wild-type CAPN3 sequence with the perspective of a real cure.
Collapse
Affiliation(s)
- Stefanie Müthel
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Muscle Research Unit at the Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and the Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, 10117 Berlin, Germany
| | - Andreas Marg
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Muscle Research Unit at the Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and the Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, 10117 Berlin, Germany
| | - Busem Ignak
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Muscle Research Unit at the Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and the Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, 10117 Berlin, Germany
| | - Janine Kieshauer
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Muscle Research Unit at the Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and the Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, 10117 Berlin, Germany
| | - Helena Escobar
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Muscle Research Unit at the Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and the Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, 10117 Berlin, Germany
| | - Christian Stadelmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Muscle Research Unit at the Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and the Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, 10117 Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Simone Spuler
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
- Muscle Research Unit at the Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC) and the Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Experimental and Clinical Research Center, 10117 Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
90
|
Evano B, Sarde L, Tajbakhsh S. Temporal static and dynamic imaging of skeletal muscle in vivo. Exp Cell Res 2023; 424:113484. [PMID: 36693490 DOI: 10.1016/j.yexcr.2023.113484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
A major challenge in the study of living systems is understanding how tissues and organs are established, maintained during homeostasis, reconstituted following injury or deteriorated during disease. Most of the studies that interrogate in vivo cell biological properties of cell populations within tissues are obtained through static imaging approaches. However, in vertebrates, little is known about which, when, and how extracellular and intracellular signals are dynamically integrated to regulate cell behaviour and fates, due largely to technical challenges. Intravital imaging of cellular dynamics in mammalian models has exposed surprising properties that have been missed by conventional static imaging approaches. Here we highlight some selected examples of intravital imaging in mouse intestinal stem cells, hematopoietic stem cells, hair follicle stem cells, and neural stem cells in the brain, each of which have distinct features from an anatomical and niche-architecture perspective. Intravital imaging of mouse skeletal muscles is comparatively less advanced due to several technical constraints that will be discussed, yet this approach holds great promise as a complementary investigative method to validate findings obtained by static imaging, as well as a method for discovery.
Collapse
Affiliation(s)
- Brendan Evano
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, 75015, France; CNRS UMR 3738, Institut Pasteur, Paris, 75015, France
| | - Liza Sarde
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, 75015, France; CNRS UMR 3738, Institut Pasteur, Paris, 75015, France; Sorbonne Université, Complexité Du Vivant, F-75005, Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells and Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, 75015, France; CNRS UMR 3738, Institut Pasteur, Paris, 75015, France.
| |
Collapse
|
91
|
Wang Q, Zhao J, Chen H, Zhou J, Chen A, Zhang J, Wang Y, Mao Z, Wang J, Qiu X, Chen Y, Wang R, Zhang Y, Miao D, Jin J. Bmi-1 Overexpression Improves Sarcopenia Induced by 1,25(OH) 2 D 3 Deficiency and Downregulates GATA4-Dependent Rela Transcription. J Bone Miner Res 2023; 38:427-442. [PMID: 36625422 DOI: 10.1002/jbmr.4770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/26/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023]
Abstract
Sarcopenia increases with age, and an underlying mechanism needs to be determined to help with designing more effective treatments. This study aimed to determine whether 1,25(OH)2 D3 deficiency could cause cellular senescence and a senescence-associated secretory phenotype (SASP) in skeletal muscle cells to induce sarcopenia, whether GATA4 could be upregulated by 1,25(OH)2 D3 deficiency to promote SASP, and whether Bmi-1 reduces the expression of GATA4 and GATA4-dependent SASP induced by 1,25(OH)2 D3 deficiency in skeletal muscle cells. Bioinformatics analyses with RNA sequencing data in skeletal muscle from physiologically aged and young mice were conducted. Skeletal muscles from 2-month-old young and 2-year-old physiologically aged wild-type (WT) mice and 8-week-old WT, Bmi-1 mesenchymal transgene (Bmi-1Tg ), Cyp27b1 homozygous (Cyp27b1-/- ), and Bmi-1Tg Cyp27b1-/- mice were observed for grip strength, cell senescence, DNA damage, and NF-κB-mediated SASP signaling of skeletal muscle. We found that muscle-derived Bmi-1 and vitamin D receptor (VDR) decreased with physiological aging, and DNA damage and GATA4-dependent SASP activation led to sarcopenia. Furthermore, 1,25(OH)2 D3 deficiency promoted DNA damage-induced GATA4 accumulation in muscles. GATA4 upregulated Rela at the region from -1448 to -1412 bp at the transcriptional level to cause NF-κB-dependent SASP for aggravating cell senescence and muscular dysfunction and sarcopenia. Bmi-1 overexpression promoted the ubiquitination and degradation of GATA4 by binding RING1B, which prevented cell senescence, SASP, and dysfunctional muscle, and improved sarcopenia induced by 1,25(OH)2 D3 deficiency. Thus, Bmi-1 overexpression improves sarcopenia induced by 1,25(OH)2 D3 deficiency, downregulates GATA4-dependent Rela transcription, and sequentially inhibits GATA4-dependent SASP in muscle cells. Therefore, Bmi-1 overexpression could be used for translational gene therapy for the ubiquitination of GATA4 and prevention of sarcopenia. © 2023 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Qiuyi Wang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jingyu Zhao
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haiyun Chen
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Medical School of Nanjing University, Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Jiawen Zhou
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ao Chen
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jin'ge Zhang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue Wang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhiyuan Mao
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiachen Wang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuehan Qiu
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yutong Chen
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rong Wang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yongjie Zhang
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dengshun Miao
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,The Research Center for Aging, Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianliang Jin
- Department of Human Anatomy, Research Centre for Bone and Stem Cells; Key Laboratory for Aging & Disease; The State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
92
|
Zhao Y, Ding Y, He L, Zhou Q, Chen X, Li Y, Alfonsi MV, Wu Z, Sun H, Wang H. Multiscale 3D genome reorganization during skeletal muscle stem cell lineage progression and aging. SCIENCE ADVANCES 2023; 9:eabo1360. [PMID: 36800432 PMCID: PMC9937580 DOI: 10.1126/sciadv.abo1360] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/17/2023] [Indexed: 06/13/2023]
Abstract
Little is known about three-dimensional (3D) genome organization in skeletal muscle stem cells [also called satellite cells (SCs)]. Here, we comprehensively map the 3D genome topology reorganization during mouse SC lineage progression. Specifically, rewiring at the compartment level is most pronounced when SCs become activated. Marked loss in topologically associating domain (TAD) border insulation and chromatin looping also occurs during early activation process. Meanwhile, TADs can form TAD clusters and super-enhancer-containing TAD clusters orchestrate stage-specific gene expression. Furthermore, we uncover that transcription factor PAX7 is pivotal in enhancer-promoter (E-P) loop formation. We also identify cis-regulatory elements that are crucial for local chromatin organization at Pax7 locus and Pax7 expression. Lastly, we unveil that geriatric SC displays a prominent gain in long-range contacts and loss of TAD border insulation. Together, our results uncover that 3D chromatin extensively reorganizes at multiple architectural levels and underpins the transcriptome remodeling during SC lineage development and SC aging.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yingzhe Ding
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Liangqiang He
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Qin Zhou
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaona Chen
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuying Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Maria Vittoria Alfonsi
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhenguo Wu
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Hao Sun
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Huating Wang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
93
|
Flynn CGK, Ginkel PRV, Hubert KA, Guo Q, Hrycaj SM, McDermott AE, Madruga A, Miller AP, Wellik DM. Hox11-expressing interstitial cells contribute to adult skeletal muscle at homeostasis. Development 2023; 150:dev201026. [PMID: 36815629 PMCID: PMC10110422 DOI: 10.1242/dev.201026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 01/13/2023] [Indexed: 02/24/2023]
Abstract
Interstitial stromal cells play critical roles in muscle development, regeneration and repair and we have previously reported that Hoxa11 and Hoxd11 are expressed in the interstitial cells of muscles attached to the zeugopod, and are crucial for the proper embryonic patterning of these muscles. Hoxa11eGFP expression continues in a subset of muscle interstitial cells through adult stages. The induction of Hoxa11-CreERT2-mediated lineage reporting (Hoxa11iTom) at adult stages in mouse results in lineage induction only in the interstitial cells. However, Hoxa11iTom+ cells progressively contribute to muscle fibers at subsequent stages. The contribution to myofibers exceeds parallel Pax7-CreERT2-mediated lineage labeling. Nuclear-specific lineage labeling demonstrates that Hoxa11-expressing interstitial cells contribute nuclear contents to myofibers. Crucially, at no point after Hoxa11iTom induction are satellite cells lineage labeled. When examined in vitro, isolated Hoxa11iTom+ interstitial cells are not capable of forming myotubes, but Hoxa11iTom+ cells can contribute to differentiating myotubes, supporting Hox-expressing interstitial cells as a new population of muscle progenitors, but not stem cells. This work adds to a small but growing body of evidence that supports a satellite cell-independent source of muscle tissue in vivo.
Collapse
Affiliation(s)
- Corey G. K. Flynn
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Paul R. Van Ginkel
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Katharine A. Hubert
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Genetics Training Program, University of Wisconsin-Madison, Madison, WI 53703, USA
| | - Qingyuan Guo
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Cell and Molecular Biology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Steven M. Hrycaj
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Aubrey E. McDermott
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Angelo Madruga
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Anna P. Miller
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Deneen M. Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
94
|
Bagley JR, Denes LT, McCarthy JJ, Wang ET, Murach KA. The myonuclear domain in adult skeletal muscle fibres: past, present and future. J Physiol 2023; 601:723-741. [PMID: 36629254 PMCID: PMC9931674 DOI: 10.1113/jp283658] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
Most cells in the body are mononuclear whereas skeletal muscle fibres are uniquely multinuclear. The nuclei of muscle fibres (myonuclei) are usually situated peripherally which complicates the equitable distribution of gene products. Myonuclear abundance can also change under conditions such as hypertrophy and atrophy. Specialised zones in muscle fibres have different functions and thus distinct synthetic demands from myonuclei. The complex structure and regulatory requirements of multinuclear muscle cells understandably led to the hypothesis that myonuclei govern defined 'domains' to maintain homeostasis and facilitate adaptation. The purpose of this review is to provide historical context for the myonuclear domain and evaluate its veracity with respect to mRNA and protein distribution resulting from myonuclear transcription. We synthesise insights from past and current in vitro and in vivo genetically modified models for studying the myonuclear domain under dynamic conditions. We also cover the most contemporary knowledge on mRNA and protein transport in muscle cells. Insights from emerging technologies such as single myonuclear RNA-sequencing further inform our discussion of the myonuclear domain. We broadly conclude: (1) the myonuclear domain can be flexible during muscle fibre growth and atrophy, (2) the mechanisms and role of myonuclear loss and motility deserve further consideration, (3) mRNA in muscle is actively transported via microtubules and locally restricted, but proteins may travel far from a myonucleus of origin and (4) myonuclear transcriptional specialisation extends beyond the classic neuromuscular and myotendinous populations. A deeper understanding of the myonuclear domain in muscle may promote effective therapies for ageing and disease.
Collapse
Affiliation(s)
- James R. Bagley
- Muscle Physiology Laboratory, Department of Kinesiology, San Francisco State University, San Francisco, California
| | | | - John J. McCarthy
- The Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Physiology, College of Medicine, University of Kentucky
| | - Eric T. Wang
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, University of Florida, Gainesville, Florida
- Myology Institute, University of Florida
- Genetics Institute, University of Florida
| | - Kevin A. Murach
- Exercise Science Research Center, Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville, Arkansas
- Cell and Molecular Biology Graduate Program, University of Arkansas
| |
Collapse
|
95
|
Schneider J, Sundaravinayagam D, Blume A, Marg A, Grunwald S, Metzler E, Escobar H, Müthel S, Wang H, Wollersheim T, Weber-Carstens S, Akalin A, Di Virgilio M, Tursun B, Spuler S. Disintegration of the NuRD Complex in Primary Human Muscle Stem Cells in Critical Illness Myopathy. Int J Mol Sci 2023; 24:2772. [PMID: 36769095 PMCID: PMC9916927 DOI: 10.3390/ijms24032772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Critical illness myopathy (CIM) is an acquired, devastating, multifactorial muscle-wasting disease with incomplete recovery. The impact on hospital costs and permanent loss of quality of life is enormous. Incomplete recovery might imply that the function of muscle stem cells (MuSC) is impaired. We tested whether epigenetic alterations could be in part responsible. We characterized human muscle stem cells (MuSC) isolated from early CIM and analyzed epigenetic alterations (CIM n = 15, controls n = 21) by RNA-Seq, immunofluorescence, analysis of DNA repair, and ATAC-Seq. CIM-MuSC were transplanted into immunodeficient NOG mice to assess their regenerative potential. CIM-MuSC exhibited significant growth deficits, reduced ability to differentiate into myotubes, and impaired DNA repair. The chromatin structure was damaged, as characterized by alterations in mRNA of histone 1, depletion or dislocation of core proteins of nucleosome remodeling and deacetylase complex, and loosening of multiple nucleosome-spanning sites. Functionally, CIM-MuSC had a defect in building new muscle fibers. Further, MuSC obtained from the electrically stimulated muscle of CIM patients was very similar to control MuSC, indicating the impact of muscle contraction in the onset of CIM. CIM not only affects working skeletal muscle but has a lasting and severe epigenetic impact on MuSC.
Collapse
Affiliation(s)
- Joanna Schneider
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
- Charité Universitätsmedizin Berlin, Department of Pediatric Neurology, 13353 Berlin, Germany
- Berlin Institute of Health–Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Devakumar Sundaravinayagam
- Laboratory of DNA Repair and Maintenance of Genome Stability, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13092 Berlin, Germany
| | - Alexander Blume
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Andreas Marg
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Stefanie Grunwald
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Eric Metzler
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Helena Escobar
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Stefanie Müthel
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Haicui Wang
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Tobias Wollersheim
- Berlin Institute of Health–Universitätsmedizin Berlin, 10117 Berlin, Germany
- Charité Universitätsmedizin Berlin, Department of Anesthesiology and Operative Intensive Care Medicine, 13353 Berlin, Germany
| | - Steffen Weber-Carstens
- Charité Universitätsmedizin Berlin, Department of Anesthesiology and Operative Intensive Care Medicine, 13353 Berlin, Germany
| | - Altuna Akalin
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Michela Di Virgilio
- Laboratory of DNA Repair and Maintenance of Genome Stability, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13092 Berlin, Germany
| | - Baris Tursun
- Berlin Institute of Medical Systems Biology (BIMSB), Max Delbruck Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, A Joint Cooperation of the Charité Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Lindenberger Weg 80, 13125 Berlin, Germany
| |
Collapse
|
96
|
Love MW, Verna DF, Kothari SN, Scott JD. Outcomes of Bariatric Surgery With Concomitant Hiatal Hernia Repair Using an Absorbable Tissue Matrix. Am Surg 2023; 89:293-299. [PMID: 34058829 DOI: 10.1177/00031348211023450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Hiatal hernias are a common finding in patients who undergo bariatric surgery with an incidence of about 20% of all bariatric patients. Controversy exists on the utility of a biosynthetic tissue matrix (BTM) usage in combination with crural repair. This study was designed to explore the safety and benefits of the use of a BTM during concomitant hiatal hernia repair with bariatric surgical procedures. METHODS This was a retrospective chart review of bariatric surgical patients who underwent a concomitant hiatal hernia repair at a single practice at a tertiary academic medical center from January 2014 to February 2019. RESULTS A total of 420 patients were reviewed. Hiatal BTM reinforcement, recurrence, and postoperative proton pump inhibitor use were reported by type of operation. Recurrence was higher in gastric bypass patients who underwent hiatal hernia repair with suture cruroplasty alone vs. those who also underwent hiatal BTM reinforcement (7.1% vs. 3.7%, P = .52) and significantly higher in gastric sleeve patients who underwent hiatal hernia repair with suture cruroplasty alone vs. those who also underwent hiatal BTM reinforcement (7.1% vs. .5%, P = .01). No patient required reoperation for hiatal hernia recurrence. DISCUSSION Performing Roux-en-Y gastric bypass or vertical sleeve gastrectomy with concomitant hiatal hernia repair is safe and durable. Employing crural reinforcement with BTM may be of benefit in reducing recurrence rates of hiatal hernia, particularly in sleeve gastrectomy patients.
Collapse
Affiliation(s)
- Michael W Love
- Department of Surgery, Prisma Health, University of South Carolina School of Medicine - Greenville, Greenville, SC, USA
| | - Daniel F Verna
- Department of Surgery, Prisma Health, University of South Carolina School of Medicine - Greenville, Greenville, SC, USA
| | - Shanu N Kothari
- Department of Surgery, Prisma Health, University of South Carolina School of Medicine - Greenville, Greenville, SC, USA
| | - John D Scott
- Department of Surgery, Prisma Health, University of South Carolina School of Medicine - Greenville, Greenville, SC, USA
| |
Collapse
|
97
|
Kumar P, Liu C, Suliburk J, Hsu JW, Muthupillai R, Jahoor F, Minard CG, Taffet GE, Sekhar RV. Supplementing Glycine and N-Acetylcysteine (GlyNAC) in Older Adults Improves Glutathione Deficiency, Oxidative Stress, Mitochondrial Dysfunction, Inflammation, Physical Function, and Aging Hallmarks: A Randomized Clinical Trial. J Gerontol A Biol Sci Med Sci 2023; 78:75-89. [PMID: 35975308 PMCID: PMC9879756 DOI: 10.1093/gerona/glac135] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Elevated oxidative stress (OxS), mitochondrial dysfunction, and hallmarks of aging are identified as key contributors to aging, but improving/reversing these defects in older adults (OA) is challenging. In prior studies, we identified that deficiency of the intracellular antioxidant glutathione (GSH) could play a role and reported that supplementing GlyNAC (combination of glycine and N-acetylcysteine [NAC]) in aged mice improved GSH deficiency, OxS, mitochondrial fatty-acid oxidation (MFO), and insulin resistance (IR). To test whether GlyNAC supplementation in OA could improve GSH deficiency, OxS, mitochondrial dysfunction, IR, physical function, and aging hallmarks, we conducted a placebo-controlled randomized clinical trial. METHODS Twenty-four OA and 12 young adults (YA) were studied. OA was randomized to receive either GlyNAC (N = 12) or isonitrogenous alanine placebo (N = 12) for 16-weeks; YA (N = 12) received GlyNAC for 2-weeks. Participants were studied before, after 2-weeks, and after 16-weeks of supplementation to assess GSH concentrations, OxS, MFO, molecular regulators of energy metabolism, inflammation, endothelial function, IR, aging hallmarks, gait speed, muscle strength, 6-minute walk test, body composition, and blood pressure. RESULTS Compared to YA, OA had GSH deficiency, OxS, mitochondrial dysfunction (with defective molecular regulation), inflammation, endothelial dysfunction, IR, multiple aging hallmarks, impaired physical function, increased waist circumference, and systolic blood pressure. GlyNAC (and not placebo) supplementation in OA improved/corrected these defects. CONCLUSION GlyNAC supplementation in OA for 16-weeks was safe and well-tolerated. By combining the benefits of glycine, NAC and GSH, GlyNAC is an effective nutritional supplement that improves and reverses multiple age-associated abnormalities to promote health in aging humans. Clinical Trials Registration Number: NCT01870193.
Collapse
Affiliation(s)
- Premranjan Kumar
- Translational Metabolism Unit, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine
- Baylor College of Medicine, Houston, TX, USA
| | - Chun Liu
- Translational Metabolism Unit, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine
- Baylor College of Medicine, Houston, TX, USA
| | - James Suliburk
- Department of Surgery
- Baylor College of Medicine, Houston, TX, USA
| | - Jean W Hsu
- Baylor College of Medicine, Houston, TX, USA
- Baylor-St. Luke’s Medical Center
| | - Raja Muthupillai
- Baylor-St. Luke’s Medical Center
- Baylor College of Medicine, Houston, TX, USA
| | - Farook Jahoor
- USDA/ARS Children’s Nutrition Research Center
- Baylor College of Medicine, Houston, TX, USA
| | - Charles G Minard
- Institute of Clinical and Translational Research
- Baylor College of Medicine, Houston, TX, USA
| | - George E Taffet
- Section of Geriatrics, Department of Medicine
- Baylor College of Medicine, Houston, TX, USA
| | - Rajagopal V Sekhar
- Translational Metabolism Unit, Section of Endocrinology, Diabetes and Metabolism, Department of Medicine
- Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
98
|
Sastourné-Arrey Q, Mathieu M, Contreras X, Monferran S, Bourlier V, Gil-Ortega M, Murphy E, Laurens C, Varin A, Guissard C, Barreau C, André M, Juin N, Marquès M, Chaput B, Moro C, O'Gorman D, Casteilla L, Girousse A, Sengenès C. Adipose tissue is a source of regenerative cells that augment the repair of skeletal muscle after injury. Nat Commun 2023; 14:80. [PMID: 36604419 PMCID: PMC9816314 DOI: 10.1038/s41467-022-35524-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Fibro-adipogenic progenitors (FAPs) play a crucial role in skeletal muscle regeneration, as they generate a favorable niche that allows satellite cells to perform efficient muscle regeneration. After muscle injury, FAP content increases rapidly within the injured muscle, the origin of which has been attributed to their proliferation within the muscle itself. However, recent single-cell RNAseq approaches have revealed phenotype and functional heterogeneity in FAPs, raising the question of how this differentiation of regenerative subtypes occurs. Here we report that FAP-like cells residing in subcutaneous adipose tissue (ScAT), the adipose stromal cells (ASCs), are rapidly released from ScAT in response to muscle injury. Additionally, we find that released ASCs infiltrate the damaged muscle, via a platelet-dependent mechanism and thus contribute to the FAP heterogeneity. Moreover, we show that either blocking ASCs infiltration or removing ASCs tissue source impair muscle regeneration. Collectively, our data reveal that ScAT is an unsuspected physiological reservoir of regenerative cells that support skeletal muscle regeneration, underlining a beneficial relationship between muscle and fat.
Collapse
Affiliation(s)
- Quentin Sastourné-Arrey
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Maxime Mathieu
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Xavier Contreras
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Sylvie Monferran
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Virginie Bourlier
- Institute of Metabolic and Cardiovascular Diseases, INSERM /Paul Sabatier University UMR 1297, Team MetaDiab, Toulouse, France
| | - Marta Gil-Ortega
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Enda Murphy
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Claire Laurens
- Institute of Metabolic and Cardiovascular Diseases, INSERM /Paul Sabatier University UMR 1297, Team MetaDiab, Toulouse, France
| | - Audrey Varin
- RESTORE, Research Center, Team 2 FLAMES, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Christophe Guissard
- RESTORE, Research Center, Team 4 GOT-IT, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Corinne Barreau
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Mireille André
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Noémie Juin
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Marie Marquès
- Institute of Metabolic and Cardiovascular Diseases, INSERM /Paul Sabatier University UMR 1297, Team MetaDiab, Toulouse, France
| | - Benoit Chaput
- Department of Plastic and Reconstructive Surgery, Toulouse University Hospital, 31100, Toulouse, France
| | - Cédric Moro
- Institute of Metabolic and Cardiovascular Diseases, INSERM /Paul Sabatier University UMR 1297, Team MetaDiab, Toulouse, France
| | - Donal O'Gorman
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Louis Casteilla
- RESTORE, Research Center, Team 4 GOT-IT, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Amandine Girousse
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Coralie Sengenès
- RESTORE, Research Center, Team 1 STROMAGICS, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France.
| |
Collapse
|
99
|
Lahmann I, Birchmeier C. Visualizing MyoD Oscillations in Muscle Stem Cells. Methods Mol Biol 2023; 2640:259-276. [PMID: 36995601 DOI: 10.1007/978-1-0716-3036-5_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
The bHLH transcription factor MyoD is a master regulator of myogenic differentiation, and its sustained expression in fibroblasts suffices to differentiate them into muscle cells. MyoD expression oscillates in activated muscle stem cells of developing, postnatal and adult muscle under various conditions: when the stem cells are dispersed in culture, when they remain associated with single muscle fibers, or when they reside in muscle biopsies. The oscillatory period is around 3 h and thus much shorter than the cell cycle or circadian rhythm. Unstable MyoD oscillations and long periods of sustained MyoD expression are observed when stem cells undergo myogenic differentiation. The oscillatory expression of MyoD is driven by the oscillatory expression of the bHLH transcription factor Hes1 that periodically represses MyoD. Ablation of the Hes1 oscillator interferes with stable MyoD oscillations and leads to prolonged periods of sustained MyoD expression. This interferes with the maintenance of activated muscle stem cells and impairs muscle growth and repair. Thus, oscillations of MyoD and Hes1 control the balance between the proliferation and differentiation of muscle stem cells. Here, we describe time-lapse imaging methods using luciferase reporters, which can monitor dynamic MyoD gene expression in myogenic cells.
Collapse
Affiliation(s)
- Ines Lahmann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, Berlin, Germany
- Neurowissenschaftliches Forschungzentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Birchmeier
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Developmental Biology/Signal Transduction Group, Berlin, Germany.
- Neurowissenschaftliches Forschungzentrum, NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
100
|
Ikemoto-Uezumi M, Kurosawa T, Minato K, Uezumi A. Analyses of Mesenchymal Progenitors in Skeletal Muscle by Fluorescence-Activated Cell Sorting and Tissue Clearing. Methods Mol Biol 2023; 2640:117-127. [PMID: 36995591 DOI: 10.1007/978-1-0716-3036-5_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Mesenchymal progenitors, which are resident progenitor populations residing in skeletal muscle interstitial space, contribute to pathogeneses such as fat infiltration, fibrosis, and heterotopic ossification. In addition to their pathological roles, mesenchymal progenitors have also been shown to play important roles for successful muscle regeneration and homeostatic muscle maintenance. Therefore, detailed and accurate analyses of these progenitors are essential for the research on muscle diseases and health. Here, we describe a method for purification of mesenchymal progenitors based on the expression of PDGFRα, which is a specific and well-established marker for mesenchymal progenitors, using fluorescence-activated cell sorting (FACS). Purified cells can be used in several downstream experiments including cell culture, cell transplantation, and gene expression analysis. We also describe the method for whole-mount 3-dimensional imaging of mesenchymal progenitors by utilizing tissue clearing. The methods described herein provide a powerful platform for studying mesenchymal progenitors in skeletal muscle.
Collapse
Affiliation(s)
- Madoka Ikemoto-Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Tamaki Kurosawa
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Keitaro Minato
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Akiyoshi Uezumi
- Muscle Aging and Regenerative Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.
| |
Collapse
|