51
|
Transcriptome RNA Sequencing Reveals That Circular RNAs Are Abundantly Expressed in Embryonic Breast Muscle of Duck. Vet Sci 2023; 10:vetsci10020075. [PMID: 36851380 PMCID: PMC10004440 DOI: 10.3390/vetsci10020075] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/14/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Circular RNAs are widespread in various species and have important roles in myogenesis. However, the circular RNAs involved in breast muscle development in ducks have not yet been studied. Here, to identify circular RNAs during duck skeletal muscle development, three pectorales from Shan Ma ducks at E13 and E19, which represent undifferentiated and differentiated myoblasts, respectively, were collected and subjected to RNA sequencing. A total of 16,622 circular RNAs were identified, of which approximately 80% were exonic circular RNAs and 260 were markedly differentially expressed between E19 and E13. The parental genes of the differentially expressed circular RNAs were significantly enriched in muscle-related biological processes. Moreover, we found that the overexpression of circGAS2-2 promoted cell cycle progression and increased the proliferation viability of duck primary myoblasts; conversely, knockdown of circGAS2-2 retarded the cell cycle and reduced the proliferation viability of myoblasts. Taken together, our results demonstrate that circular RNAs are widespread and variously expressed during the development of duck skeletal muscle and that circGAS2-2 is involved in the regulation of myogenesis.
Collapse
|
52
|
Kametaka S, Isobe M, Komata K, Morinaga M, Nagahata K, Lee-Hotta S, Uchiyama Y, Shibata M, Sugiura H. Protective effects of hachimijiogan (HJG), a Japanese Kampo medicine, on cancer cachectic muscle wasting in mice. Biomed Res 2023; 44:199-207. [PMID: 37779032 DOI: 10.2220/biomedres.44.199] [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: 10/03/2023]
Abstract
Myogenesis is required to generate skeletal muscle tissue and to maintain skeletal muscle mass. Decreased myogenesis under various pathogenic conditions results in muscular atrophy. Through a small screening of Japanese traditional (Kampo) medicines, hachimijiogan (HJG) was shown to promote the myogenic differentiation of C2C12 myoblasts through the upregulation of myogenin. In tumor-bearing cancer-cachectic mice, HJG was also found to have a protective effect against cancer-cachectic muscle wasting. This effect was significant when HJG was administered in combination with aerobic exercise by treadmill running. Moreover, HJG ameliorated the cellular atrophy of C2C12 myotubes induced by treatment with conditioned medium derived from a colon-26 cancer cell culture. In addition, HJG suppressed H2O2-dependent myotube atrophy, suggesting that HJG could reverse the atrophic phenotypes by eliminating reactive oxygen species.
Collapse
Affiliation(s)
- Satoshi Kametaka
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1- 20 Daiko-Minami, Higashi-Ku, Nagoya, Aichi 461-0047, Japan
| | - Mari Isobe
- Division of Morphological Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Kenshin Komata
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1- 20 Daiko-Minami, Higashi-Ku, Nagoya, Aichi 461-0047, Japan
- Division of Home-visiting Nursing, Magokoronomori, Youmeikai Medical Corp. 17-10 Hatanocho, Atsutaku, Nagoya, Aichi 456-0077, Japan
| | - Makoto Morinaga
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1- 20 Daiko-Minami, Higashi-Ku, Nagoya, Aichi 461-0047, Japan
- Product development Center 2, R&D Institute, Morinaga & Co., Ltd
| | - Kazuma Nagahata
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1- 20 Daiko-Minami, Higashi-Ku, Nagoya, Aichi 461-0047, Japan
| | - Sachiko Lee-Hotta
- Division of Creative Physical Therapy, Department of Integrated Health Sciences, Gradu- ate School of Medicine, Nagoya University, 1-1-20 Daiko-Minami, Higashi-Ku, Nagoya, Aichi 461-0047, Japan
| | - Yasushi Uchiyama
- Division of Creative Physical Therapy, Department of Integrated Health Sciences, Gradu- ate School of Medicine, Nagoya University, 1-1-20 Daiko-Minami, Higashi-Ku, Nagoya, Aichi 461-0047, Japan
| | - Masahiro Shibata
- Division of Morphological Sciences, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Hideshi Sugiura
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1- 20 Daiko-Minami, Higashi-Ku, Nagoya, Aichi 461-0047, Japan
| |
Collapse
|
53
|
Cortés E, Pak JS, Özkan E. Structure and evolution of neuronal wiring receptors and ligands. Dev Dyn 2023; 252:27-60. [PMID: 35727136 PMCID: PMC10084454 DOI: 10.1002/dvdy.512] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 01/04/2023] Open
Abstract
One of the fundamental properties of a neuronal circuit is the map of its connections. The cellular and developmental processes that allow for the growth of axons and dendrites, selection of synaptic targets, and formation of functional synapses use neuronal surface receptors and their interactions with other surface receptors, secreted ligands, and matrix molecules. Spatiotemporal regulation of the expression of these receptors and cues allows for specificity in the developmental pathways that wire stereotyped circuits. The families of molecules controlling axon guidance and synapse formation are generally conserved across animals, with some important exceptions, which have consequences for neuronal connectivity. Here, we summarize the distribution of such molecules across multiple taxa, with a focus on model organisms, evolutionary processes that led to the multitude of such molecules, and functional consequences for the diversification or loss of these receptors.
Collapse
Affiliation(s)
- Elena Cortés
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Joseph S Pak
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
54
|
Dittmar T, Hass R. Extracellular Events Involved in Cancer Cell-Cell Fusion. Int J Mol Sci 2022; 23:16071. [PMID: 36555709 PMCID: PMC9784959 DOI: 10.3390/ijms232416071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Fusion among different cell populations represents a rare process that is mediated by both intrinsic and extracellular events. Cellular hybrid formation is relayed by orchestrating tightly regulated signaling pathways that can involve both normal and neoplastic cells. Certain important cell merger processes are often required during distinct organismal and tissue development, including placenta and skeletal muscle. In a neoplastic environment, however, cancer cell fusion can generate new cancer hybrid cells. Following survival during a subsequent post-hybrid selection process (PHSP), the new cancer hybrid cells express different tumorigenic properties. These can include elevated proliferative capacity, increased metastatic potential, resistance to certain therapeutic compounds, and formation of cancer stem-like cells, all of which characterize significantly enhanced tumor plasticity. However, many parts within this multi-step cascade are still poorly understood. Aside from intrinsic factors, cell fusion is particularly affected by extracellular conditions, including an inflammatory microenvironment, viruses, pH and ionic stress, hypoxia, and exosome signaling. Accordingly, the present review article will primarily highlight the influence of extracellular events that contribute to cell fusion in normal and tumorigenic tissues.
Collapse
Affiliation(s)
- Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Stockumer Str. 10, 58448 Witten, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
55
|
Tran V, Nahlé S, Robert A, Desanlis I, Killoran R, Ehresmann S, Thibault MP, Barford D, Ravichandran KS, Sauvageau M, Smith MJ, Kmita M, Côté JF. Biasing the conformation of ELMO2 reveals that myoblast fusion can be exploited to improve muscle regeneration. Nat Commun 2022; 13:7077. [PMID: 36400788 PMCID: PMC9674853 DOI: 10.1038/s41467-022-34806-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
Myoblast fusion is fundamental for the development of multinucleated myofibers. Evolutionarily conserved proteins required for myoblast fusion include RAC1 and its activator DOCK1. In the current study we analyzed the contribution of the DOCK1-interacting ELMO scaffold proteins to myoblast fusion. When Elmo1-/- mice underwent muscle-specific Elmo2 genetic ablation, they exhibited severe myoblast fusion defects. A mutation in the Elmo2 gene that reduced signaling resulted in a decrease in myoblast fusion. Conversely, a mutation in Elmo2 coding for a protein with an open conformation increased myoblast fusion during development and in muscle regeneration. Finally, we showed that the dystrophic features of the Dysferlin-null mice, a model of limb-girdle muscular dystrophy type 2B, were reversed when expressing ELMO2 in an open conformation. These data provide direct evidence that the myoblast fusion process could be exploited for regenerative purposes and improve the outcome of muscle diseases.
Collapse
Affiliation(s)
- Viviane Tran
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Sarah Nahlé
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Amélie Robert
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
| | - Inès Desanlis
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Ryan Killoran
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Sophie Ehresmann
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | | | - David Barford
- MRC Laboratory of Molecular Biology, Cambridge, CB2 OQH, UK
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, 22908, VA, USA
- VIB/UGent Inflammation Research Centre, Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Martin Sauvageau
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Department of Biochemistry, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Matthew J Smith
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, H3T 1J4, Canada
- Department of Pathology and Cell Biology, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Marie Kmita
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Department of Experimental Medicine, McGill University, Montréal, QC, H3G 2M1, Canada
| | - Jean-François Côté
- Montreal Clinical Research Institute (IRCM), Montreal, QC, H2W 1R7, Canada.
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Molecular Biology Programs, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
- Department of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC, H3A 0C7, Canada.
| |
Collapse
|
56
|
Abstract
Nuclear movement is crucial for the development of many cell types and organisms. Nuclear movement is highly conserved, indicating its necessity for cellular function and development. In addition to mononucleated cells, there are several examples of cells in which multiple nuclei exist within a shared cytoplasm. These multinucleated cells and syncytia have important functions for development and homeostasis. Here, we review a subset of the developmental contexts in which the regulation of the movement and positioning of multiple nuclei are well understood, including pronuclear migration, the Drosophila syncytial blastoderm, the Caenorhabditis elegans hypodermis, skeletal muscle and filamentous fungi. We apply the principles learned from these models to other systems.
Collapse
Affiliation(s)
- Jorel R. Padilla
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA
| | | | - Eric S. Folker
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA
| |
Collapse
|
57
|
Sinha S, Elbaz‐Alon Y, Avinoam O. Ca 2+ as a coordinator of skeletal muscle differentiation, fusion and contraction. FEBS J 2022; 289:6531-6542. [PMID: 35689496 PMCID: PMC9795905 DOI: 10.1111/febs.16552] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/05/2022] [Accepted: 06/09/2022] [Indexed: 12/30/2022]
Abstract
Muscle regeneration is essential for vertebrate muscle homeostasis and recovery after injury. During regeneration, muscle stem cells differentiate into myocytes, which then fuse with pre-existing muscle fibres. Hence, differentiation, fusion and contraction must be tightly regulated during regeneration to avoid the disastrous consequences of premature fusion of myocytes to actively contracting fibres. Cytosolic calcium (Ca2+ ), which is coupled to both induction of myogenic differentiation and contraction, has more recently been implicated in the regulation of myocyte-to-myotube fusion. In this viewpoint, we propose that Ca2+ -mediated coordination of differentiation, fusion and contraction is a feature selected in the amniotes to facilitate muscle regeneration.
Collapse
Affiliation(s)
- Sansrity Sinha
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Yael Elbaz‐Alon
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| | - Ori Avinoam
- Department of Biomolecular SciencesWeizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
58
|
Chakraborty M, Sivan A, Biswas A, Sinha B. Early tension regulation coupled to surface myomerger is necessary for the primary fusion of C2C12 myoblasts. Front Physiol 2022; 13. [PMID: 36277221 PMCID: PMC7613732 DOI: 10.3389/fphys.2022.976715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Here, we study the time-dependent regulation of fluctuation–tension during myogenesis and the role of the fusogen, myomerger. We measure nanometric height fluctuations of the basal membrane of C2C12 cells after triggering differentiation. Fusion of cells increases fluctuation–tension but prefers a transient lowering of tension (at ∼2–24 h). Cells fail to fuse if early tension is continuously enhanced by methyl-β-cyclodextrin (MβCD). Perturbing tension regulation also reduces fusion. During this pre-fusion window, cells that finally differentiate usually display lower tension than other non-fusing cells, validating early tension states to be linked to fate decision. Early tension reduction is accompanied by low but gradually increasing level of the surface myomerger. Locally too, regions with higher myomerger intensity display lower tension. However, this negative correlation is lost in the early phase by MβCD-based cholesterol depletion or later as differentiation progresses. We find that with tension and surface-myomerger’s enrichment under these conditions, myomerger clusters become pronouncedly diffused. We, therefore, propose that low tension aided by clustered surface-myomerger at the early phase is crucial for fusion and can be disrupted by cholesterol-reducing molecules, implying the potential to affect muscle health.
Collapse
|
59
|
Sun H, Shen L, Zhang P, Lin F, Ma J, Wu Y, Yu H, Sun L. Inhibition of High-Temperature Requirement Protein A2 Protease Activity Represses Myogenic Differentiation via UPRmt. Int J Mol Sci 2022; 23:ijms231911761. [PMID: 36233059 PMCID: PMC9569504 DOI: 10.3390/ijms231911761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 12/02/2022] Open
Abstract
Skeletal muscles require muscle satellite cell (MuSC) differentiation to facilitate the replenishment and repair of muscle fibers. A key step in this process is called myogenic differentiation. The differentiation ability of MuSCs decreases with age and can result in sarcopenia. Although mitochondria have been reported to be involved in myogenic differentiation by promoting a bioenergetic remodeling, little is known about the interplay of mitochondrial proteostasis and myogenic differentiation. High-temperature-requirement protein A2 (HtrA2/Omi) is a protease that regulates proteostasis in the mitochondrial intermembrane space (IMS). Mice deficient in HtrA2 protease activity show a distinct phenotype of sarcopenia. To investigate the role of IMS proteostasis during myogenic differentiation, we treated C2C12 myoblasts with UCF101, a specific inhibitor of HtrA2 during differentiation process. A key step in this process is called myogenic differentiation. The differentiation ability of MuSCs decreases with age and can result in sarcopenia. Further, CHOP, p-eIF2α, and other mitochondrial unfolded protein response (UPRmt)-related proteins are upregulated. Therefore, we suggest that imbalance of mitochondrial IMS proteostasis acts via a retrograde signaling pathway to inhibit myogenic differentiation via the UPRmt pathway. These novel mechanistic insights may have implications for the development of new strategies for the treatment of sarcopenia.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huimei Yu
- Correspondence: (H.Y.); (L.S.); Tel.: +86-0431-8561-9495 (H.Y. & L.S.)
| | - Liankun Sun
- Correspondence: (H.Y.); (L.S.); Tel.: +86-0431-8561-9495 (H.Y. & L.S.)
| |
Collapse
|
60
|
Wu P, Yong P, Zhang Z, Xu R, Shang R, Shi J, Zhang J, Bi P, Chen E, Du S. Loss of Myomixer Results in Defective Myoblast Fusion, Impaired Muscle Growth, and Severe Myopathy in Zebrafish. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:1023-1038. [PMID: 36083384 PMCID: PMC10112271 DOI: 10.1007/s10126-022-10159-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/25/2022] [Indexed: 06/15/2023]
Abstract
The development and growth of fish skeletal muscles require myoblast fusion to generate multinucleated myofibers. While zebrafish fast-twitch muscle can fuse to generate multinucleated fibers, the slow-twitch muscle fibers remain mononucleated in zebrafish embryos and larvae. The mechanism underlying the fiber-type-specific control of fusion remains elusive. Recent genetic studies using mice identified a long-sought fusion factor named Myomixer. To understand whether Myomixer is involved in the fiber-type specific fusion, we analyzed the transcriptional regulation of myomixer expression and characterized the muscle growth phenotype upon genetic deletion of myomixer in zebrafish. The data revealed that overexpression of Sonic Hedgehog (Shh) drastically inhibited myomixer expression and blocked myoblast fusion, recapitulating the phenotype upon direct genetic deletion of myomixer from zebrafish. The fusion defect in myomixer mutant embryos could be faithfully rescued upon re-expression of zebrafish myomixer gene or its orthologs from shark or human. Interestingly, myomixer mutant fish survived to adult stage though were notably smaller than wildtype siblings. Severe myopathy accompanied by the uncontrolled adipose infiltration was observed in both fast and slow muscle tissues of adult myomixer mutants. Collectively, our data highlight an indispensable role of myomixer gene for cell fusion during both embryonic muscle development and post-larval muscle growth.
Collapse
Affiliation(s)
- Ping Wu
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, China
| | - Pengzheng Yong
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA
| | - Zhanxiong Zhang
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA
| | - Rui Xu
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA
| | - Renjie Shang
- Center for Molecular Medicine & Department of Genetics, University of Georgia, Athens, USA
| | - Jun Shi
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, USA
- College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Jianshe Zhang
- Department of Bioengineering and Environmental Science, Changsha University, Changsha, China
| | - Pengpeng Bi
- Center for Molecular Medicine & Department of Genetics, University of Georgia, Athens, USA
| | - Elizabeth Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, USA
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, USA.
| |
Collapse
|
61
|
Singh AK, Rai A, Weber A, Posern G. miRNA mediated downregulation of cyclase-associated protein 1 (CAP1) is required for myoblast fusion. Front Cell Dev Biol 2022; 10:899917. [PMID: 36246999 PMCID: PMC9562714 DOI: 10.3389/fcell.2022.899917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Myoblast fusion is essential for the formation, growth, and regeneration of skeletal muscle, but the molecular mechanisms that govern fusion and myofiber formation remain poorly understood. Past studies have shown an important role of the actin cytoskeleton and actin regulators in myoblast fusion. The Cyclase-Associated Proteins (CAP) 1 and 2 recently emerged as critical regulators of actin treadmilling in higher eukaryotes including mammals. Whilst the role of CAP2 in skeletal muscle development and function is well characterized, involvement of CAP1 in this process remains elusive. Here we report that CAP1, plays a critical role in cytoskeletal remodeling during myoblast fusion and formation of myotubes. Cap1 mRNA and protein are expressed in both murine C2C12 and human LHCN-M2 myoblasts, but their abundance decreases during myogenic differentiation. Perturbing the temporally controlled expression of CAP1 by overexpression or CRISPR-Cas9 mediated knockout impaired actin rearrangement, myoblast alignment, expression of profusion molecules, differentiation into multinucleated myotubes, and myosin heavy chain expression. Endogenous Cap1 expression is post-transcriptionally downregulated during differentiation by canonical myomiRs miR-1, miR-133, and miR-206, which have conserved binding sites at the 3′ UTR of the Cap1 mRNA. Deletion of the endogenous 3′ UTR by CRISPR-Cas9 in C2C12 cells phenocopies overexpression of CAP1 by inhibiting myotube formation. Our findings implicates Cap1 and its myomiR-mediated downregulation in the myoblast fusion process and the generation of skeletal muscle.
Collapse
Affiliation(s)
- Anurag Kumar Singh
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Internal Medicine I, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- *Correspondence: Anurag Kumar Singh, ; Guido Posern,
| | - Amrita Rai
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Anja Weber
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- *Correspondence: Anurag Kumar Singh, ; Guido Posern,
| |
Collapse
|
62
|
Chen YF, Lee CW, Wu HH, Lin WT, Lee OK. Immunometabolism of macrophages regulates skeletal muscle regeneration. Front Cell Dev Biol 2022; 10:948819. [PMID: 36147742 PMCID: PMC9485946 DOI: 10.3389/fcell.2022.948819] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Sarcopenia is an age-related progressive loss of skeletal muscle mass, quality, and strength disease. In addition, sarcopenia is tightly correlated with age-associated pathologies, such as sarcopenic obesity and osteoporosis. Further understanding of disease mechanisms and the therapeutic strategies in muscle regeneration requires a deeper knowledge of the interaction of skeletal muscle and other cells in the muscle tissue. Skeletal muscle regeneration is a complex process that requires a series of highly coordinated events involving communication between muscle stem cells and niche cells, such as muscle fibro/adipogenic progenitors and macrophages. Macrophages play a critical role in tissue regeneration and the maintenance of muscle homeostasis by producing growth factors and cytokines that regulate muscle stem cells and myofibroblast activation. Furthermore, the aging-related immune dysregulation associated with the release of trophic factors and the polarization in macrophages transiently affect the inflammatory phase and impair muscle regeneration. In this review, we focus on the role and regulation of macrophages in skeletal muscle regeneration and homeostasis. The aim of this review is to highlight the important roles of macrophages as a therapeutic target in age-related sarcopenia and the increasing understanding of how macrophages are regulated will help to advance skeletal muscle regeneration.
Collapse
Affiliation(s)
- Yu-Fan Chen
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, Taiwan
| | - Chien-Wei Lee
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, Taiwan
| | - Hao-Hsiang Wu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Ting Lin
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Doctoral Degree Program of Translational Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Oscar K. Lee
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Stem Cell Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan
- *Correspondence: Oscar K. Lee,
| |
Collapse
|
63
|
Genetic Parameter Estimation and Genome-Wide Association Analysis of Social Genetic Effects on Average Daily Gain in Purebreds and Crossbreds. Animals (Basel) 2022; 12:ani12172300. [PMID: 36078021 PMCID: PMC9454713 DOI: 10.3390/ani12172300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/23/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Average daily gain (ADG) is influenced by both an individual’s direct genetic effect (DGE) and by a social genetic effect (SGE) derived from pen mates. Therefore, identifying the DGE and SGE on ADG is essential for a better understanding of pig breeding systems. We conducted this study to elucidate the genetic characteristics and relationships of DGE and SGE on ADG using purebred and crossbred pigs. We found that the DGE and SGE both contributed to ADG in both populations. In addition, the SGE of purebred pigs was highly correlated with the DGE of crossbred pigs. Furthermore, we identified several genomic regions that may be associated with the DGE and SGE on ADG. Our findings will contribute to future genomic evaluation studies of socially affected traits. Abstract Average daily gain (ADG) is an important growth trait in the pig industry. The direct genetic effect (DGE) has been studied mainly to assess the association between genetic information and economic traits. The social genetic effect (SGE) has been shown to affect ADG simultaneously with the DGE because of group housing systems. We conducted this study to elucidate the genetic characteristics and relationships of the DGE and SGE of purebred Korean Duroc and crossbred pigs by single-step genomic best linear unbiased prediction and a genome-wide association study. We used the genotype, phenotype, and pedigree data of 1779, 6022, and 7904 animals, respectively. Total heritabilities on ADG were 0.19 ± 0.04 and 0.39 ± 0.08 for purebred and crossbred pigs, respectively. The genetic correlation was the greatest (0.77 ± 0.12) between the SGE of purebred and DGE of crossbred pigs. We found candidate genes located in the quantitative trait loci (QTLs) for the SGE that were associated with behavior and neurodegenerative diseases, and candidate genes in the QTLs for DGE that were related to body mass, size of muscle fiber, and muscle hypertrophy. These results suggest that the genomic selection of purebred animals could be applied for crossbred performance.
Collapse
|
64
|
Fei W, Pang E, Hou L, Dai J, Liu M, Wang X, Xie B, Wang J. Synergistic Effect of Hydrogen and 5-Aza on Myogenic Differentiation through the p38 MAPK Signaling Pathway in Adipose-Derived Mesenchymal Stem Cells. Int J Stem Cells 2022; 16:78-92. [PMID: 36042011 PMCID: PMC9978834 DOI: 10.15283/ijsc21238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives This study aims to clarify the systems underlying regulation and regulatory roles of hydrogen combined with 5-Aza in the myogenic differentiation of adipose mesenchymal stem cells (ADSCs). Methods and Results In this study, ADSCs acted as an in vitro myogenic differentiating mode. First, the Alamar blue Staining and mitochondrial tracer technique were used to verify whether hydrogen combined with 5-Aza could promote cell proliferation. In addition, this study assessed myogenic differentiating markers (e.g., Myogenin, Mhc and Myod protein expressions) based on the Western blotting assay, analysis on cellular morphological characteristics (e.g., Myotube number, length, diameter and maturation index), RT-PCR (Myod, Myogenin and Mhc mRNA expression) and Immunofluorescence analysis (Desmin, Myosin and β-actin protein expression). Finally, to verify the mechanism of myogenic differentiation of hydrogen-bound 5-Aza, we performed bioinformatics analysis and Western blot to detect the expression of p-P38 protein. Hydrogen combined with 5-Aza significantly enhanced the proliferation and myogenic differentiation of ADSCs in vitro by increasing the number of single-cell mitochondria and upregulating the expression of myogenic biomarkers such as Myod, Mhc and myotube formation. The expressions of p-P38 was up-regulated by hydrogen combined with 5-Aza. The differentiating ability was suppressed when the cells were cultivated in combination with SB203580 (p38 MAPK signal pathway inhibitor). Conclusions Hydrogen alleviates the cytotoxicity of 5-Aza and synergistically promotes the myogenic differentiation capacity of adipose stem cells via the p38 MAPK pathway. Thus, the mentioned results present insights into myogenic differentiation and are likely to generate one potential alternative strategy for skeletal muscle related diseases.
Collapse
Affiliation(s)
- Wenyong Fei
- Department of Sports Medicine, Northern Jiangsu People’s Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Erkai Pang
- Department of Sports Medicine, Northern Jiangsu People’s Hospital, Dalian Medical University, Dalian, China
| | - Lei Hou
- Department of Sports Medicine, Northern Jiangsu People’s Hospital, Dalian Medical University, Dalian, China
| | - Jihang Dai
- Department of Sports Medicine, Northern Jiangsu People’s Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Mingsheng Liu
- Department of Sports Medicine, Northern Jiangsu People’s Hospital, Dalian Medical University, Dalian, China
| | - Xuanqi Wang
- Department of Sports Medicine, Northern Jiangsu People’s Hospital, Dalian Medical University, Dalian, China
| | - Bin Xie
- Department of Sports Medicine, Northern Jiangsu People’s Hospital, Dalian Medical University, Dalian, China
| | - Jingcheng Wang
- Department of Sports Medicine, Northern Jiangsu People’s Hospital, Clinical Medical College, Yangzhou University, Yangzhou, China,Correspondence to Jingcheng Wang, Department of Sports Medicine, Northern Jiangsu People’s Hospital, Clinical Medical College, Yangzhou University, 98# Nantong xi Road, Yangzhou 225001, China , Tel: +86-13909254888, Fax: +86-051487373425, E-mail:
| |
Collapse
|
65
|
Bai L, Tu WY, Xiao Y, Zhang K, Shen C. Motoneurons innervation determines the distinct gene expressions in multinucleated myofibers. Cell Biosci 2022; 12:140. [PMID: 36042463 PMCID: PMC9429338 DOI: 10.1186/s13578-022-00876-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 08/09/2022] [Indexed: 11/22/2022] Open
Abstract
Background Neuromuscular junctions (NMJs) are peripheral synapses connecting motoneurons and skeletal myofibers. At the postsynaptic side in myofibers, acetylcholine receptor (AChR) proteins are clustered by the neuronal agrin signal. Meanwhile, several nuclei in each myofiber are specially enriched around the NMJ for postsynaptic gene transcription. It remains mysterious that how gene expressions in these synaptic nuclei are systematically regulated, especially by motoneurons. Results We found that synaptic nuclei have a distinctive chromatin structure and gene expression profiling. Synaptic nuclei are formed during NMJ development and maintained by motoneuron innervation. Transcriptome analysis revealed that motoneuron innervation determines the distinct expression patterns in the synaptic region and non-synaptic region in each multinucleated myofiber, probably through epigenetic regulation. Myonuclei in synaptic and non-synaptic regions have different responses to denervation. Weighted gene co-expression network analysis revealed that the histone lysine demethylases Kdm1a is a negative regulator of synaptic gene expression. Inhibition of Kdm1a promotes AChR expression but impairs motor functions. Conclusion These results demonstrate that motoneurons innervation determines the distinct gene expressions in multinucleated myofibers. Thus, dysregulation of nerve-controlled chromatin structure and muscle gene expression might cause muscle weakness and atrophy in motoneuron degenerative disorders. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00876-6.
Collapse
|
66
|
Rujano MA, Briand D, Ðelić B, Marc J, Spéder P. An interplay between cellular growth and atypical fusion defines morphogenesis of a modular glial niche in Drosophila. Nat Commun 2022; 13:4999. [PMID: 36008397 PMCID: PMC9411534 DOI: 10.1038/s41467-022-32685-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Neural stem cells (NSCs) live in an intricate cellular microenvironment supporting their activity, the niche. Whilst shape and function are inseparable, the morphogenetic aspects of niche development are poorly understood. Here, we use the formation of a glial niche to investigate acquisition of architectural complexity. Cortex glia (CG) in Drosophila regulate neurogenesis and build a reticular structure around NSCs. We first show that individual CG cells grow tremendously to ensheath several NSC lineages, employing elaborate proliferative mechanisms which convert these cells into syncytia rich in cytoplasmic bridges. CG syncytia further undergo homotypic cell-cell fusion, using defined cell surface receptors and actin regulators. Cellular exchange is however dynamic in space and time. This atypical cell fusion remodels cellular borders, restructuring the CG syncytia. Ultimately, combined growth and fusion builds the multi-level architecture of the niche, and creates a modular, spatial partition of the NSC population. Our findings provide insights into how a niche forms and organises while developing intimate contacts with a stem cell population.
Collapse
Affiliation(s)
| | | | - Bojana Ðelić
- Institut Pasteur, CNRS UMR3738, Paris, France
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Cell Division and Neurogenesis, Ecole Normale Supérieure, CNRS, Inserm, PSL Université Paris, Paris, France
| | - Julie Marc
- Institut Pasteur, CNRS UMR3738, Paris, France
| | | |
Collapse
|
67
|
Oppen D, Grossmann L, Weiss J. Insights into characterizing and producing anisotropic food structures. Crit Rev Food Sci Nutr 2022; 64:1158-1176. [PMID: 35997311 DOI: 10.1080/10408398.2022.2113365] [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: 11/03/2022]
Abstract
Besides the flavor profile of food, texture plays a major role in terms of the acceptance and likeability of food products. In contrast to gel-like homogenous isotropic structures, where the characterization is established and structure-texture mechanisms are well understood, there is still a lack of knowledge in the field of anisotropic complex food matrices. Food systems that show anisotropic properties in terms of macroscopic mechanical anisotropy as in grown meat, or mixed complex systems where anisotropic shaped particles or fibers are embedded into an isotropic matrix are challenging to characterize, hence the structure-texture correlation is not trivial to understand. In this paper, we bring together the state of the art of different anisotropic structures as a source of food, their formation in terms of structured plant proteins, and consequently the structure-texture correlation of those. Characteristic terms and properties to differentiate between anisotropic systems are introduced with the purpose to facilitate characterization of those. Based on the here provided terms and characteristics, further studies toward understanding such systems and their perception can be conducted. Beyond that, a first opinion on crucial influencing factors on the perception of anisotropic systems and their mechanistic background is provided.
Collapse
Affiliation(s)
- Dominic Oppen
- Department of Food Material Science, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart, Germany
| | - Lutz Grossmann
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts, USA
| | - Jochen Weiss
- Department of Food Material Science, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
68
|
Matsumoto T. Implications of Polyploidy and Ploidy Alterations in Hepatocytes in Liver Injuries and Cancers. Int J Mol Sci 2022; 23:ijms23169409. [PMID: 36012671 PMCID: PMC9409051 DOI: 10.3390/ijms23169409] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Polyploidy, a condition in which more than two sets of chromosomes are present in a cell, is a characteristic feature of hepatocytes. A significant number of hepatocytes physiologically undergo polyploidization at a young age. Polyploidization of hepatocytes is enhanced with age and in a diseased liver. It is worth noting that polyploid hepatocytes can proliferate, in marked contrast to other types of polyploid cells, such as megakaryocytes and cardiac myocytes. Polyploid hepatocytes divide to maintain normal liver homeostasis and play a role in the regeneration of the damaged liver. Furthermore, polyploid hepatocytes have been shown to dynamically reduce ploidy during liver regeneration. Although it is still unclear why hepatocytes undergo polyploidization, accumulating evidence has revealed that alterations in the ploidy in hepatocytes are involved in the pathophysiology of liver cirrhosis and carcinogenesis. This review discusses the significance of hepatocyte ploidy in physiological liver function, liver injury, and liver cancer.
Collapse
Affiliation(s)
- Tomonori Matsumoto
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
69
|
ISOBE M, SUZUKI Y, SUGIURA H, SHIBATA M, OHSAKI Y, KAMETAKA S. Novel cell-based system to assay cell-cell fusion during myotube formation. Biomed Res 2022; 43:107-114. [DOI: 10.2220/biomedres.43.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Mari ISOBE
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University
| | - Yumika SUZUKI
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University
| | - Hideshi SUGIURA
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University
| | - Masahiro SHIBATA
- Division of Morphological Sciences, Kagoshima University Graduate School of Medical and Dental Sciences
| | - Yuki OHSAKI
- Department of Anatomy I, Sapporo Medical University School of Medicine
| | - Satoshi KAMETAKA
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University
| |
Collapse
|
70
|
Xu X, Alcocer HM, Gravely ME, Jackson AR, Gonzalez JM. Effects of in ovo injection of nicotinamide riboside on high-yield broiler myogenesis. J Anim Sci 2022; 100:6652313. [PMID: 35908780 PMCID: PMC9339335 DOI: 10.1093/jas/skac203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/02/2022] [Indexed: 11/12/2022] Open
Abstract
The objective of this study was to determine the effects of in ovo injection of high-yield broiler embryos with nicotinamide riboside (NR) on pectoralis major muscle (PMM) development, growth, and gene expression. Fertilized Cobb 700 broiler eggs were randomly assigned to one of four treatments within a 2 × 2 factorial design. Factor 1 consisted of NR dose (DOS) with eggs receiving 0 or 2.5 mM NR. Factor 2 consisted of injection location (LOC), with treatments injected into either the yolk sac or albumen. At day 10 of incubation, 100 μL of the assigned NR dose was injected into the yolk sac of the developing embryo and chicks were euthanized within 24 h of hatching. Chick PMM and individual fiber morphometrics, and expression of genes associated with cell cycle progression were analyzed. There were DOS × LOC interactions for hatched chick PM weight and length (P < 0.04). When NR was injected into the albumen, PMM weight decreased (P < 0.05); when NR was injected into the yolk, PMM weight increased (P < 0.05). Pectoralis major length was not affected (P > 0.05) when NR was injected into the albumen but was increased (P < 0.05) when NR was injected into the yolk. There was a DOS × LOC interaction (P = 0.04) for muscle fiber density and tended to be a DOS × LOC interaction (P = 0.07) for muscle fiber CSA. Pectoralis major muscle fiber density was not affected when NR was injected into the albumen (P > 0.05), but density increased when NR was injected into the yolk (P < 0.05). There were DOS × LOC interactions for hatched chick COXII, cyclin D, and SIRT1 expression (P ≤ 0.04), which may indicate NR improves skeletal muscle development and growth by enhancing myoblast proliferation during embryonic development.
Collapse
Affiliation(s)
- Xiaoxing Xu
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | - Hanna M Alcocer
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | - Morgan E Gravely
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | | | - John M Gonzalez
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
71
|
Chen WJ, Lin IH, Lee CW, Yoshioka K, Ono Y, Yan YT, Yen Y, Chen YF. Ribonucleotide reductase M2B in the myofibers modulates stem cell fate in skeletal muscle. NPJ Regen Med 2022; 7:37. [PMID: 35906243 PMCID: PMC9338274 DOI: 10.1038/s41536-022-00231-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
The balance among quiescence, differentiation, and self-renewal of skeletal muscle stem cells (MuSCs) is tightly regulated by their intrinsic and extrinsic properties from the niche. How the niche controls MuSC fate remains unclear. Ribonucleotide reductase M2B (Rrm2b) modulates MuSC quiescence/differentiation in muscle in response to injury. Rrm2b knockout in myofibers, but not in MuSCs, led to weakness of muscles, such as a loss of muscle mass and strength. After muscle injury, damaged myofibers were more efficiently repaired in the Rrm2b myofiber-specific knockout mice than the control mice, but these myofibers were thinner and showed weak functioning. Rrm2b-deleted myofibers released several myokines, which trigger MuSCs to differentiate but not re-enter the quiescent stage to replenish the stem cell pool. Overall, Rrm2b in the myofibers plays a critical role in modulating the MuSC fate by modifying the microenvironment, and it may lead to a possible strategy to treat muscle disorders.
Collapse
Affiliation(s)
- Wan-Jing Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11529, Taiwan
| | - I-Hsuan Lin
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, 11031, Taipei, Taiwan
| | - Chien-Wei Lee
- Center for Translational Genomics Research, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Kiyoshi Yoshioka
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Yu-Ting Yan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11529, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yun Yen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, 11031, Taipei, Taiwan. .,Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, 11031, Taipei, Taiwan. .,Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, 11031, Taipei, Taiwan. .,Cancer Center, Taipei Municipal WanFang Hospital, 116081, Taipei, Taiwan. .,Center for Cancer Translational Research, Tzu Chi University, Hualien, Taiwan.
| | - Yi-Fan Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, 11529, Taiwan. .,Graduate Institute of Translational Medicine, College of Medical Science and Technology, Taipei Medical University, 11031, Taipei, Taiwan. .,International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, 11031, Taipei, Taiwan. .,Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
72
|
Fukada SI, Higashimoto T, Kaneshige A. Differences in muscle satellite cell dynamics during muscle hypertrophy and regeneration. Skelet Muscle 2022; 12:17. [PMID: 35794679 PMCID: PMC9258228 DOI: 10.1186/s13395-022-00300-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/29/2022] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle homeostasis and function are ensured by orchestrated cellular interactions among several types of cells. A noticeable aspect of skeletal muscle biology is the drastic cell-cell communication changes that occur in multiple scenarios. The process of recovering from an injury, which is known as regeneration, has been relatively well investigated. However, the cellular interplay that occurs in response to mechanical loading, such as during resistance training, is poorly understood compared to regeneration. During muscle regeneration, muscle satellite cells (MuSCs) rebuild multinuclear myofibers through a stepwise process of proliferation, differentiation, fusion, and maturation, whereas during mechanical loading-dependent muscle hypertrophy, MuSCs do not undergo such stepwise processes (except in rare injuries) because the nuclei of MuSCs become directly incorporated into the mature myonuclei. In this review, six specific examples of such differences in MuSC dynamics between regeneration and hypertrophy processes are discussed.
Collapse
Affiliation(s)
- So-Ichiro Fukada
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Tatsuyoshi Higashimoto
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Akihiro Kaneshige
- Project for Muscle Stem Cell Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka, 565-0871, Japan
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, 569-1125, Japan
| |
Collapse
|
73
|
The mechanism of Megalobrama amblycephala muscle injury repair based on RNA-seq. Gene X 2022; 827:146455. [PMID: 35395368 DOI: 10.1016/j.gene.2022.146455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 01/19/2022] [Accepted: 03/25/2022] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle myogenesis and injury-induced muscle regeneration contribute to muscle formation. Skeletal muscle stem cells, termed satellite cells (SCs), proliferate to repair injured muscle. To identify the molecular mechanism of regeneration after muscle injury as well as the genes related to muscle development in fish, in this study, the immunohistochemistry and the high-throughput RNA sequencing (RNA-seq) analysis were performed after Megalobrama amblycephala muscle was injured by needle stab. The results showed that paired box7-positive (Pax7+) SCs increased, and peaked at 96 to 144 h-post injury (hpi). The 6729 differentially expressed genes (DEGs), including 2125 up-regulated and 4604 down-regulated genes were found. GO terms significantly enriched by DEGs contained intercellular connections, signaling transduction and enzyme activity. KEGG enrichment analysis showed that most of the pathways were related to immunity, metabolism and cells related molecules, including actin skeleton regulation, Epstein Barr virus infection and plaque adhesion. The WGCNA results revealed that actin cytoskeleton and lipid metabolism related genes probably played crucial roles during repair after muscle injury. Collectively, all these results will provide new insights into the molecular mechanisms underlying muscle injury repair of fish.
Collapse
|
74
|
Monocyte-Macrophage Lineage Cell Fusion. Int J Mol Sci 2022; 23:ijms23126553. [PMID: 35742997 PMCID: PMC9223484 DOI: 10.3390/ijms23126553] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 02/06/2023] Open
Abstract
Cell fusion (fusogenesis) occurs in natural and pathological conditions in prokaryotes and eukaryotes. Cells of monocyte–macrophage lineage are highly fusogenic. They create syncytial multinucleated giant cells (MGCs) such as osteoclasts (OCs), MGCs associated with the areas of infection/inflammation, and foreign body-induced giant cells (FBGCs). The fusion of monocytes/macrophages with tumor cells may promote cancer metastasis. We describe types and examples of monocyte–macrophage lineage cell fusion and the role of actin-based structures in cell fusion.
Collapse
|
75
|
Fu Y, Hao X, Shang P, Chamba Y, Zhang B, Zhang H. Functional Identification of Porcine DLK1 during Muscle Development. Animals (Basel) 2022; 12:ani12121523. [PMID: 35739860 PMCID: PMC9219491 DOI: 10.3390/ani12121523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Skeletal muscle is the largest tissue and serves as a protein reservoir and energy reservoir in the human and animal body. It also serves as the main metabolic activity site. The formation of skeletal muscle mainly depends on the differentiation and fusion of myocytes and other complex ordered processes; each step is regulated by various factors. In this study, we investigated the expression profiles, functional identification, and regulatory pathways of Delta-like 1 homolog (DLK1) in pigs and myocytes. We found that DLK1 was highly expressed in the muscle tissues of pigs. DLK1 promoted myocyte proliferation, migration, differentiation, fusion, and muscular hypertrophy, but suppressed muscle degradation. DLK1 also inhibited the Notch signaling pathway by regulating the expression of key factors in the pathway, thereby producing a phenotype in which DLK1 promotes muscle development. These findings provide valuable information to improve our understanding of the functional mechanisms of DLK1 that underly myogenesis to accelerate the process of animal genetic improvement. Abstract DLK1 is paternally expressed and is involved in metabolism switching, stem cell maintenance, cell proliferation, and differentiation. Porcine DLK1 was identified in our previous study as a candidate gene that regulates muscle development. In the present study, we characterized DLK1 expression in pigs, and the results showed that DLK1 was highly expressed in the muscles of pigs. In-vitro cellular tests showed that DLK1 promoted myoblast proliferation, migration, and muscular hypertrophy, and at the same time inhibited muscle degradation. The expression of myogenic and fusion markers and the formation of multinucleated myotubes were both upregulated in myoblasts with DLK1 overexpression. DLK1 levels in cultured myocytes were negatively correlated with the expression of key factors in the Notch pathway, suggesting that the suppression of Notch signaling pathways may mediate these processes. Collectively, our results suggest a biological function of DLK1 as an enhancer of muscle development by the inhibition of Notch pathways.
Collapse
Affiliation(s)
- Yu Fu
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.F.); (X.H.)
| | - Xin Hao
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.F.); (X.H.)
| | - Peng Shang
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi 860000, China; (P.S.); (Y.C.)
| | - Yangzom Chamba
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi 860000, China; (P.S.); (Y.C.)
| | - Bo Zhang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.F.); (X.H.)
- Correspondence: (B.Z.); (H.Z.); Tel.: +86-010-62734852 (H.Z.)
| | - Hao Zhang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (Y.F.); (X.H.)
- Correspondence: (B.Z.); (H.Z.); Tel.: +86-010-62734852 (H.Z.)
| |
Collapse
|
76
|
Nadkarni AV, Heald R. Reconstitution of muscle cell microtubule organization in vitro. Cytoskeleton (Hoboken) 2022; 78:492-502. [PMID: 35666041 DOI: 10.1002/cm.21710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/06/2022]
Abstract
Skeletal muscle differentiation occurs as muscle precursor cells (myoblasts) elongate and fuse to form multinucleated syncytial myotubes in which the highly-organized actomyosin sarcomeres of muscle fibers assemble. Although less well characterized, the microtubule cytoskeleton also undergoes dramatic rearrangement during myogenesis. The centrosome-nucleated microtubule array found in myoblasts is lost as the nuclear membrane acquires microtubule nucleating activity and microtubules emerge from multiple sites in the cell, eventually rearranging into a grid-like pattern in myotubes. In order to characterize perinuclear microtubule organization using a biochemically tractable system, we isolated nuclei from mouse C2C12 skeletal muscle cells during the course of differentiation and incubated them in cytoplasmic extracts prepared from eggs of the frog Xenopus laevis. Whereas centrosomes associated with myoblast nuclei gave rise to radial microtubule arrays in extracts, myotube nuclei produced a sun-like pattern with microtubules transiently nucleating from the entire nuclear envelope. Perinuclear microtubule growth was suppressed by inhibition of Aurora A kinase or by degradation of RNA, treatments that also inhibited microtubule growth from sperm centrosomes. Myotube nuclei displayed microtubule motor-based movements leading to their separation, as occurs in myotubes. This in vitro assay therefore recapitulates key features of microtubule organization and nuclear movement observed during muscle cell differentiation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ambika V Nadkarni
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, USA
| | - Rebecca Heald
- Department of Molecular & Cell Biology, University of California, Berkeley, CA, USA
| |
Collapse
|
77
|
Ramirez-Martinez A, Zhang Y, van den Boogaard MJ, McAnally JR, Rodriguez-Caycedo C, Chai AC, Chemello F, Massink MP, Cuppen I, Elferink MG, van Es RJ, Janssen NG, Walraven-van Oijen LP, Liu N, Bassel-Duby R, van Jaarsveld RH, Olson EN. Impaired activity of the fusogenic micropeptide Myomixer causes myopathy resembling Carey-Fineman-Ziter syndrome. J Clin Invest 2022; 132:e159002. [PMID: 35642635 PMCID: PMC9151691 DOI: 10.1172/jci159002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/21/2022] [Indexed: 01/19/2023] Open
Abstract
Skeletal muscle fibers contain hundreds of nuclei, which increase the overall transcriptional activity of the tissue and perform specialized functions. Multinucleation occurs through myoblast fusion, mediated by the muscle fusogens Myomaker (MYMK) and Myomixer (MYMX). We describe a human pedigree harboring a recessive truncating variant of the MYMX gene that eliminates an evolutionarily conserved extracellular hydrophobic domain of MYMX, thereby impairing fusogenic activity. Homozygosity of this human variant resulted in a spectrum of abnormalities that mimicked the clinical presentation of Carey-Fineman-Ziter syndrome (CFZS), caused by hypomorphic MYMK variants. Myoblasts generated from patient-derived induced pluripotent stem cells displayed defective fusion, and mice bearing the human MYMX variant died perinatally due to muscle abnormalities. In vitro assays showed that the human MYMX variant conferred minimal cell-cell fusogenicity, which could be restored with CRISPR/Cas9-mediated base editing, thus providing therapeutic potential for this disorder. Our findings identify MYMX as a recessive, monogenic human disease gene involved in CFZS, and provide new insights into the contribution of myoblast fusion to neuromuscular diseases.
Collapse
Affiliation(s)
- Andres Ramirez-Martinez
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yichi Zhang
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - John R. McAnally
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Cristina Rodriguez-Caycedo
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Andreas C. Chai
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Francesco Chemello
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | - Robert J.J. van Es
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Nard G. Janssen
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Ning Liu
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Eric N. Olson
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
78
|
Generation of Cancer Stem/Initiating Cells by Cell-Cell Fusion. Int J Mol Sci 2022; 23:ijms23094514. [PMID: 35562905 PMCID: PMC9101717 DOI: 10.3390/ijms23094514] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/10/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
CS/ICs have raised great expectations in cancer research and therapy, as eradication of this key cancer cell type is expected to lead to a complete cure. Unfortunately, the biology of CS/ICs is rather complex, since no common CS/IC marker has yet been identified. Certain surface markers or ALDH1 expression can be used for detection, but some studies indicated that cancer cells exhibit a certain plasticity, so CS/ICs can also arise from non-CS/ICs. Another problem is intratumoral heterogeneity, from which it can be inferred that different CS/IC subclones must be present in the tumor. Cell–cell fusion between cancer cells and normal cells, such as macrophages and stem cells, has been associated with the generation of tumor hybrids that can exhibit novel properties, such as an enhanced metastatic capacity and even CS/IC properties. Moreover, cell–cell fusion is a complex process in which parental chromosomes are mixed and randomly distributed among daughter cells, resulting in multiple, unique tumor hybrids. These, if they have CS/IC properties, may contribute to the heterogeneity of the CS/IC pool. In this review, we will discuss whether cell–cell fusion could also lead to the origin of different CS/ICs that may expand the overall CS/IC pool in a primary tumor.
Collapse
|
79
|
Zhang Y, Wang L, Kang H, Lin CY, Fan Y. Applying exercise-mimetic engineered skeletal muscle model to interrogate the adaptive response of irisin to mechanical force. iScience 2022; 25:104135. [PMID: 35434556 PMCID: PMC9010619 DOI: 10.1016/j.isci.2022.104135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/28/2022] [Accepted: 03/17/2022] [Indexed: 11/30/2022] Open
Abstract
Physical exercise induces the secretion of irisin from contractile muscle into circulation; however, the adaptive response of irisin to mechanical stimulus in skeletal muscle in vitro remains numerously unknown. In an effort to investigate whether irisin is inducible in vitro, we developed a bioreactor consisting of a retractable mechanical force controller and a conditional tissue culture system. Upon this model, a distinguished surge of irisin was detected in stretched myotubes as cyclic strain initiated, and the surge was able to be stalled by knocking out FNDC5. Intriguingly, increased irisin secretory is associated with the shifts of MyHC isoforms from anaerobic type to aerobic type in myotubes. We further revealed that PGC-1α1 and PGC-1α4 mRNAs expression, rather than PGC-1α2 and PGC-1α3, contributed to the generation of irisin in myotubes during cyclic strain. Lastly, combined with co-culturing MC3T3 osteoblasts, we demonstrated the bioactivity of generated irisin, promoting the osteogenic differentiation. Irisin is producible in an exercise-mimetic engineered skeletal muscle model Enhanced irisin production in response to a long-term cyclic stretch PGC-1α1 and PGC-1α4 mRNAs expression contributed to the generation of irisin Demonstration that induced irisin in our model regulating osteoblasts as native ways
Collapse
Affiliation(s)
- Yuwei Zhang
- Key laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Lizhen Wang
- Key laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Hongyan Kang
- Key laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Chia-Ying Lin
- Key laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.,Department of Biomedical, Chemical & Environmental Engineering, University of Cincinnati, Cincinnati, USA.,Department of Orthopaedic Surgery, University of Cincinnati, Cincinnati, USA.,Department of Neurosurgery, University of Cincinnati, Cincinnati, USA
| | - Yubo Fan
- Key laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.,School of Engineering Medicine, Beihang University, Beijing 100083, China
| |
Collapse
|
80
|
Patel A, Wu Y, Han X, Su Y, Maugel T, Shroff H, Roy S. Cytonemes coordinate asymmetric signaling and organization in the Drosophila muscle progenitor niche. Nat Commun 2022; 13:1185. [PMID: 35246530 PMCID: PMC8897416 DOI: 10.1038/s41467-022-28587-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/02/2022] [Indexed: 12/29/2022] Open
Abstract
Asymmetric signaling and organization in the stem-cell niche determine stem-cell fates. Here, we investigate the basis of asymmetric signaling and stem-cell organization using the Drosophila wing-disc that creates an adult muscle progenitor (AMP) niche. We show that AMPs extend polarized cytonemes to contact the disc epithelial junctions and adhere themselves to the disc/niche. Niche-adhering cytonemes localize FGF-receptor to selectively adhere to the FGF-producing disc and receive FGFs in a contact-dependent manner. Activation of FGF signaling in AMPs, in turn, reinforces disc-specific cytoneme polarity/adhesion, which maintains their disc-proximal positions. Loss of cytoneme-mediated adhesion promotes AMPs to lose niche occupancy and FGF signaling, occupy a disc-distal position, and acquire morphological hallmarks of differentiation. Niche-specific AMP organization and diversification patterns are determined by localized expression and presentation patterns of two different FGFs in the wing-disc and their polarized target-specific distribution through niche-adhering cytonemes. Thus, cytonemes are essential for asymmetric signaling and niche-specific AMP organization.
Collapse
Affiliation(s)
- Akshay Patel
- grid.164295.d0000 0001 0941 7177Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD USA
| | - Yicong Wu
- grid.94365.3d0000 0001 2297 5165Laboratory of High-Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD USA
| | - Xiaofei Han
- grid.94365.3d0000 0001 2297 5165Laboratory of High-Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD USA
| | - Yijun Su
- grid.94365.3d0000 0001 2297 5165Laboratory of High-Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD USA ,grid.94365.3d0000 0001 2297 5165Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD USA
| | - Tim Maugel
- grid.164295.d0000 0001 0941 7177Department of Biology, Laboratory for Biological Ultrastructure, University of Maryland, College Park, MD USA
| | - Hari Shroff
- grid.94365.3d0000 0001 2297 5165Laboratory of High-Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD USA ,grid.94365.3d0000 0001 2297 5165Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD USA
| | - Sougata Roy
- grid.164295.d0000 0001 0941 7177Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD USA
| |
Collapse
|
81
|
Gerber T, Loureiro C, Schramma N, Chen S, Jain A, Weber A, Weigert A, Santel M, Alim K, Treutlein B, Camp JG. Spatial transcriptomic and single-nucleus analysis reveals heterogeneity in a gigantic single-celled syncytium. eLife 2022; 11:e69745. [PMID: 35195068 PMCID: PMC8865844 DOI: 10.7554/elife.69745] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 02/07/2022] [Indexed: 11/25/2022] Open
Abstract
In multicellular organisms, the specification, coordination, and compartmentalization of cell types enable the formation of complex body plans. However, some eukaryotic protists such as slime molds generate diverse and complex structures while remaining in a multinucleate syncytial state. It is unknown if different regions of these giant syncytial cells have distinct transcriptional responses to environmental encounters and if nuclei within the cell diversify into heterogeneous states. Here, we performed spatial transcriptome analysis of the slime mold Physarum polycephalum in the plasmodium state under different environmental conditions and used single-nucleus RNA-sequencing to dissect gene expression heterogeneity among nuclei. Our data identifies transcriptome regionality in the organism that associates with proliferation, syncytial substructures, and localized environmental conditions. Further, we find that nuclei are heterogenous in their transcriptional profile and may process local signals within the plasmodium to coordinate cell growth, metabolism, and reproduction. To understand how nuclei variation within the syncytium compares to heterogeneity in single-nucleus cells, we analyzed states in single Physarum amoebal cells. We observed amoebal cell states at different stages of mitosis and meiosis, and identified cytokinetic features that are specific to nuclei divisions within the syncytium. Notably, we do not find evidence for predefined transcriptomic states in the amoebae that are observed in the syncytium. Our data shows that a single-celled slime mold can control its gene expression in a region-specific manner while lacking cellular compartmentalization and suggests that nuclei are mobile processors facilitating local specialized functions. More broadly, slime molds offer the extraordinary opportunity to explore how organisms can evolve regulatory mechanisms to divide labor, specialize, balance competition with cooperation, and perform other foundational principles that govern the logic of life.
Collapse
Affiliation(s)
- Tobias Gerber
- Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
| | - Cristina Loureiro
- Department of Biosystems Science and Engineering, ETH ZürichBaselSwitzerland
| | - Nico Schramma
- Max Planck Institute for Dynamics and Self-OrganizationGöttingenGermany
| | - Siyu Chen
- Max Planck Institute for Dynamics and Self-OrganizationGöttingenGermany
- Physics Department, Technical University of MunichMünchenGermany
| | - Akanksha Jain
- Department of Biosystems Science and Engineering, ETH ZürichBaselSwitzerland
| | - Anne Weber
- Max Planck Institute for Dynamics and Self-OrganizationGöttingenGermany
| | - Anne Weigert
- Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
| | - Malgorzata Santel
- Department of Biosystems Science and Engineering, ETH ZürichBaselSwitzerland
| | - Karen Alim
- Max Planck Institute for Dynamics and Self-OrganizationGöttingenGermany
- Physics Department, Technical University of MunichMünchenGermany
| | - Barbara Treutlein
- Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
- Department of Biosystems Science and Engineering, ETH ZürichBaselSwitzerland
| | - J Gray Camp
- Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
- Roche Institute for Translational Bioengineering (ITB), Roche Pharma Research and Early Development, Roche Innovation CenterBaselSwitzerland
- University of BaselBaselSwitzerland
| |
Collapse
|
82
|
Orioka M, Eguchi M, Mizui Y, Ikeda Y, Sakama A, Li Q, Yoshimura H, Ozawa T, Citterio D, Hiruta Y. A Series of Furimazine Derivatives for Sustained Live-Cell Bioluminescence Imaging and Application to the Monitoring of Myogenesis at the Single-Cell Level. Bioconjug Chem 2022; 33:496-504. [PMID: 35184558 DOI: 10.1021/acs.bioconjchem.2c00035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bioluminescence (BL) imaging, which utilizes light emitted through the enzymatic reaction of luciferase oxidizing its substrate luciferin, enables sensitive and noninvasive monitoring of life phenomena. Herein, we developed a series of caged furimazine (FMZ) derivatives by introducing a protective group at the C-3 position and a hydroxy group at the C-6 phenyl ring to realize long-term live-cell BL imaging based on the NanoLuc (NLuc)/NanoKAZ (NKAZ)-FMZ system. The membrane permeability and cytotoxicity of the substrates were evaluated and related to their hydrophobicity. Among the series, the derivative with the bulkiest protective group (adamantanecarbonyl group) and a hydroxy substituent (named Ad-FMZ-OH) showed significantly prolonged and constant BL signal in cells expressing NLuc compared to the native FMZ substrate. This derivative enabled continuous BL imaging at the single-cell level for 24 h. Furthermore, we applied Ad-FMZ-OH to BL imaging of myocyte fusion and succeeded in the consecutive and sensitive monitoring at a single-cell level over a day. In summary, NLuc/NKAZ-caged FMZ derivatives have the potential to be applied to live-cell BL imaging of various life phenomena that require long-term observation.
Collapse
Affiliation(s)
- Mariko Orioka
- Department of Applied Chemistry Faculty of Science and Technology, Keio University, Hiyoshi 3-14-1, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Masatoshi Eguchi
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuki Mizui
- Department of Applied Chemistry Faculty of Science and Technology, Keio University, Hiyoshi 3-14-1, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Yuma Ikeda
- Department of Applied Chemistry Faculty of Science and Technology, Keio University, Hiyoshi 3-14-1, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Akihiro Sakama
- Department of Applied Chemistry Faculty of Science and Technology, Keio University, Hiyoshi 3-14-1, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Qiaojing Li
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideaki Yoshimura
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Daniel Citterio
- Department of Applied Chemistry Faculty of Science and Technology, Keio University, Hiyoshi 3-14-1, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| | - Yuki Hiruta
- Department of Applied Chemistry Faculty of Science and Technology, Keio University, Hiyoshi 3-14-1, Kohoku-ku, Yokohama, Kanagawa 223-8522, Japan
| |
Collapse
|
83
|
Szabo K, Varga D, Vegh AG, Liu N, Xiao X, Xu L, Dux L, Erdelyi M, Rovo L, Keller-Pinter A. Syndecan-4 affects myogenesis via Rac1-mediated actin remodeling and exhibits copy-number amplification and increased expression in human rhabdomyosarcoma tumors. Cell Mol Life Sci 2022; 79:122. [PMID: 35128576 PMCID: PMC8818642 DOI: 10.1007/s00018-021-04121-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/14/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Skeletal muscle demonstrates a high degree of regenerative capacity repeating the embryonic myogenic program under strict control. Rhabdomyosarcoma is the most common sarcoma in childhood and is characterized by impaired muscle differentiation. In this study, we observed that silencing the expression of syndecan-4, the ubiquitously expressed transmembrane heparan sulfate proteoglycan, significantly enhanced myoblast differentiation, and fusion. During muscle differentiation, the gradually decreasing expression of syndecan-4 allows the activation of Rac1, thereby mediating myoblast fusion. Single-molecule localized superresolution direct stochastic optical reconstruction microscopy (dSTORM) imaging revealed nanoscale changes in actin cytoskeletal architecture, and atomic force microscopy showed reduced elasticity of syndecan-4-knockdown cells during fusion. Syndecan-4 copy-number amplification was observed in 28% of human fusion-negative rhabdomyosarcoma tumors and was accompanied by increased syndecan-4 expression based on RNA sequencing data. Our study suggests that syndecan-4 can serve as a tumor driver gene in promoting rabdomyosarcoma tumor development. Our results contribute to the understanding of the role of syndecan-4 in skeletal muscle development, regeneration, and tumorigenesis.
Collapse
|
84
|
Aase-Remedios ME, Coll-Lladó C, Ferrier DEK. Amphioxus muscle transcriptomes reveal vertebrate-like myoblast fusion genes and a highly conserved role of insulin signalling in the metabolism of muscle. BMC Genomics 2022; 23:93. [PMID: 35105312 PMCID: PMC8805411 DOI: 10.1186/s12864-021-08222-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/25/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The formation and functioning of muscles are fundamental aspects of animal biology, and the evolution of 'muscle genes' is central to our understanding of this tissue. Feeding-fasting-refeeding experiments have been widely used to assess muscle cellular and metabolic responses to nutrition. Though these studies have focused on vertebrate models and only a few invertebrate systems, they have found similar processes are involved in muscle degradation and maintenance. Motivation for these studies stems from interest in diseases whose pathologies involve muscle atrophy, a symptom also triggered by fasting, as well as commercial interest in the muscle mass of animals kept for consumption. Experimentally modelling atrophy by manipulating nutritional state causes muscle mass to be depleted during starvation and replenished with refeeding so that the genetic mechanisms controlling muscle growth and degradation can be understood. RESULTS Using amphioxus, the earliest branching chordate lineage, we address the gap in previous work stemming from comparisons between distantly related vertebrate and invertebrate models. Our amphioxus feeding-fasting-refeeding muscle transcriptomes reveal a highly conserved myogenic program and that the pro-orthologues of many vertebrate myoblast fusion genes were present in the ancestral chordate, despite these invertebrate chordates having unfused mononucleate myocytes. We found that genes differentially expressed between fed and fasted amphioxus were orthologous to the genes that respond to nutritional state in vertebrates. This response is driven in a large part by the highly conserved IGF/Akt/FOXO pathway, where depleted nutrient levels result in activation of FOXO, a transcription factor with many autophagy-related gene targets. CONCLUSION Reconstruction of these gene networks and pathways in amphioxus muscle provides a key point of comparison between the distantly related groups assessed thus far, significantly refining the reconstruction of the ancestral state for chordate myoblast fusion genes and identifying the extensive role of duplicated genes in the IGF/Akt/FOXO pathway across animals. Our study elucidates the evolutionary trajectory of muscle genes as they relate to the increased complexity of vertebrate muscles and muscle development.
Collapse
Affiliation(s)
- Madeleine E Aase-Remedios
- The Scottish Oceans Institute, Gatty Marine Laboratory, School of Biology, University of St Andrews, St Andrews, Fife, KY16 8LB, UK
| | - Clara Coll-Lladó
- The Scottish Oceans Institute, Gatty Marine Laboratory, School of Biology, University of St Andrews, St Andrews, Fife, KY16 8LB, UK
| | - David E K Ferrier
- The Scottish Oceans Institute, Gatty Marine Laboratory, School of Biology, University of St Andrews, St Andrews, Fife, KY16 8LB, UK.
| |
Collapse
|
85
|
Drosophila melanogaster: A Model System to Study Distinct Genetic Programs in Myoblast Fusion. Cells 2022; 11:cells11030321. [PMID: 35159130 PMCID: PMC8834112 DOI: 10.3390/cells11030321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 11/25/2022] Open
Abstract
Muscle fibers are multinucleated cells that arise during embryogenesis through the fusion of mononucleated myoblasts. Myoblast fusion is a lifelong process that is crucial for the growth and regeneration of muscles. Understanding the molecular mechanism of myoblast fusion may open the way for novel therapies in muscle wasting and weakness. Recent reports in Drosophila and mammals have provided new mechanistic insights into myoblast fusion. In Drosophila, muscle formation occurs twice: during embryogenesis and metamorphosis. A fundamental feature is the formation of a cell–cell communication structure that brings the apposing membranes into close proximity and recruits possible fusogenic proteins. However, genetic studies suggest that myoblast fusion in Drosophila is not a uniform process. The complexity of the players involved in myoblast fusion can be modulated depending on the type of muscle that is formed. In this review, we introduce the different types of multinucleated muscles that form during Drosophila development and provide an overview in advances that have been made to understand the mechanism of myoblast fusion. Finally, we will discuss conceptual frameworks in cell–cell fusion in Drosophila and mammals.
Collapse
|
86
|
Yoon JH, Lee SM, Lee Y, Kim MJ, Yang JW, Choi JY, Kwak JY, Lee KP, Yang YR, Kwon KS. Alverine citrate promotes myogenic differentiation and ameliorates muscle atrophy. Biochem Biophys Res Commun 2022; 586:157-162. [PMID: 34847441 DOI: 10.1016/j.bbrc.2021.11.076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/18/2021] [Indexed: 11/25/2022]
Abstract
Sarcopenia is the age-related loss of muscle mass and function and no pharmacological medication has been approved for its treatment. We established an atrogin-1/MAFbx promoter assay to find drug candidates that inhibit myotube atrophy. Alverine citrate (AC) was identified using high-throughput screening of an existing drug library. AC is an established medicine for stomach and intestinal spasms. AC treatment increased myotube diameter and inhibited atrophy signals induced by either C26-conditioned medium or dexamethasone in cultured C2C12 myoblasts. AC also enhanced myoblast fusion through the upregulation of fusion-related genes during C2C12 myoblast differentiation. Oral administration of AC improves muscle mass and physical performance in aged mice, as well as hindlimb-disused mice. Taken together, our data suggest that AC may be a novel therapeutic candidate for improving muscle weakness, including sarcopenia.
Collapse
Affiliation(s)
- Jong Hyeon Yoon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Seung-Min Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | | | - Min Ju Kim
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jae Won Yang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jeong Yi Choi
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ju Yeon Kwak
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Yong Ryoul Yang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea; Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea; Aventi Inc., Daejeon, Republic of Korea.
| |
Collapse
|
87
|
Wei Z, Ge F, Che Y, Wu S, Dong X, Song D. Metabolomics Coupled with Pathway Analysis Provides Insights into Sarco-Osteoporosis Metabolic Alterations and Estrogen Therapeutic Effects in Mice. Biomolecules 2021; 12:41. [PMID: 35053189 PMCID: PMC8773875 DOI: 10.3390/biom12010041] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
Abstract
Postmenopausal osteoporosis (PMOP) and sarcopenia are common diseases that predominantly affect postmenopausal women. In the occurrence and development of these two diseases, they are potentially pathologically connected with each other at various molecular levels. However, the application of metabolomics in sarco-osteoporosis and the metabolic rewiring happening throughout the estrogen loss-replenish process have not been reported. To investigate the metabolic alteration of sarco-osteoporosis and the possible therapeutical effects of estradiol, 24 mice were randomly divided into sham surgery, ovariectomy (OVX), and estradiol-treated groups. Three-dimensional reconstructions and histopathology examination showed significant bone loss after ovariectomy. Estrogen can well protect against OVX-induced bone loss deterioration. UHPLC-Q-TOF/MS was preformed to profile semi- polar metabolites of skeletal muscle samples from all groups. Metabolomics analysis revealed metabolic rewiring occurred in OVX group, most of which can be reversed by estrogen supplementation. In total, 65 differential metabolites were identified, and pathway analysis revealed that sarco-osteoporosis was related to the alterations in purine metabolism, glycerophospholipid metabolism, arginine biosynthesis, tryptophan metabolism, histidine metabolism, oxidative phosphorylation, and thermogenesis, which provided possible explanations for the metabolic mechanism of sarco-osteoporosis. This study indicates that an UHPLC-Q-TOF/MS-based metabolomics approach can elucidate the metabolic reprogramming mechanisms of sarco-osteoporosis and provide biological evidence of the therapeutical effects of estrogen on sarco-osteoporosis.
Collapse
Affiliation(s)
- Ziheng Wei
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China;
| | - Fei Ge
- School of Medicine, Shanghai University, Shanghai 200444, China; (F.G.); (Y.C.)
- College of Sciences, Shanghai University, Shanghai 200444, China
| | - Yanting Che
- School of Medicine, Shanghai University, Shanghai 200444, China; (F.G.); (Y.C.)
- College of Sciences, Shanghai University, Shanghai 200444, China
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Si Wu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xin Dong
- School of Medicine, Shanghai University, Shanghai 200444, China; (F.G.); (Y.C.)
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China;
| |
Collapse
|
88
|
Eigler T, Zarfati G, Amzallag E, Sinha S, Segev N, Zabary Y, Zaritsky A, Shakked A, Umansky KB, Schejter ED, Millay DP, Tzahor E, Avinoam O. ERK1/2 inhibition promotes robust myotube growth via CaMKII activation resulting in myoblast-to-myotube fusion. Dev Cell 2021; 56:3349-3363.e6. [PMID: 34932950 PMCID: PMC8693863 DOI: 10.1016/j.devcel.2021.11.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 07/28/2021] [Accepted: 11/21/2021] [Indexed: 11/19/2022]
Abstract
Myoblast fusion is essential for muscle development and regeneration. Yet, it remains poorly understood how mononucleated myoblasts fuse with preexisting fibers. We demonstrate that ERK1/2 inhibition (ERKi) induces robust differentiation and fusion of primary mouse myoblasts through a linear pathway involving RXR, ryanodine receptors, and calcium-dependent activation of CaMKII in nascent myotubes. CaMKII activation results in myotube growth via fusion with mononucleated myoblasts at a fusogenic synapse. Mechanistically, CaMKII interacts with and regulates MYMK and Rac1, and CaMKIIδ/γ knockout mice exhibit smaller regenerated myofibers following injury. In addition, the expression of a dominant negative CaMKII inhibits the formation of large multinucleated myotubes. Finally, we demonstrate the evolutionary conservation of the pathway in chicken myoblasts. We conclude that ERK1/2 represses a signaling cascade leading to CaMKII-mediated fusion of myoblasts to myotubes, providing an attractive target for the cultivated meat industry and regenerative medicine.
Collapse
Affiliation(s)
- Tamar Eigler
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Giulia Zarfati
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Emmanuel Amzallag
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sansrity Sinha
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Nadav Segev
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yishaia Zabary
- Department of Software & Information Systems Engineering, Ben Gurion University, Be'er Sheva, Israel
| | - Assaf Zaritsky
- Department of Software & Information Systems Engineering, Ben Gurion University, Be'er Sheva, Israel
| | - Avraham Shakked
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kfir-Baruch Umansky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal D Schejter
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
89
|
Hinkle ER, Essader TO, Gentile GM, Giudice J. ViaFuse: Fiji macros to calculate skeletal muscle cell viability and fusion index. Skelet Muscle 2021; 11:28. [PMID: 34915930 PMCID: PMC8675483 DOI: 10.1186/s13395-021-00284-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/24/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Measuring biological features of skeletal muscle cells is difficult because of their unique morphology and multinucleate nature upon differentiation. Here, we developed a new Fiji macro package called ViaFuse (that stands for viability and fusion) to measure skeletal muscle cell viability and differentiation. To test ViaFuse, we utilized immunofluorescence images of differentiated myotubes where the capping actin protein of muscle z-line subunit beta (CAPZB) was depleted in comparison with control cells. RESULTS We compared the values achieved using the ViaFuse macros first with manual quantification performed by researchers and second with those obtained utilizing the MATLAB muscle-centric software MyoCount. We observed a high degree of correlation between all methods of quantification. CONCLUSIONS ViaFuse can detect the borders of myotubes and identify nuclear clumps which have been limitations of previous muscle-centric imaging software. The ViaFuse macros require little computer power or space to run and user inputs to the ViaFuse macros are minimal, thereby automating the analysis process in a quick, easy, and accurate fashion. Additionally, the ViaFuse macros work with Fiji, an existing imaging software widely used by skeletal muscle researchers. Furthermore, ViaFuse is compatible with many computer systems, has a very intuitive interface, and does not require prior complex mathematical knowledge. Therefore, we propose ViaFuse as a robust and meticulous method to quantify skeletal muscle cell viability and differentiation.
Collapse
Affiliation(s)
- Emma Rose Hinkle
- Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Curriculum in Genetics and Molecular Biology (GMB), The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Tasneem Omar Essader
- Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Gabrielle Marie Gentile
- Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Curriculum in Genetics and Molecular Biology (GMB), The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jimena Giudice
- Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Curriculum in Genetics and Molecular Biology (GMB), The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
90
|
Mechanobiology of muscle and myofibril morphogenesis. Cells Dev 2021; 168:203760. [PMID: 34863916 DOI: 10.1016/j.cdev.2021.203760] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023]
Abstract
Muscles generate forces for animal locomotion. The contractile apparatus of muscles is the sarcomere, a highly regular array of large actin and myosin filaments linked by gigantic titin springs. During muscle development many sarcomeres assemble in series into long periodic myofibrils that mechanically connect the attached skeleton elements. Thus, ATP-driven myosin forces can power movement of the skeleton. Here we review muscle and myofibril morphogenesis, with a particular focus on their mechanobiology. We describe recent progress on the molecular structure of sarcomeres and their mechanical connections to the skeleton. We discuss current models predicting how tension coordinates the assembly of key sarcomeric components to periodic myofibrils that then further mature during development. This requires transcriptional feedback mechanisms that may help to coordinate myofibril assembly and maturation states with the transcriptional program. To fuel the varying energy demands of muscles we also discuss the close mechanical interactions of myofibrils with mitochondria and nuclei to optimally support powerful or enduring muscle fibers.
Collapse
|
91
|
Rodríguez-Fdez S, Bustelo XR. Rho GTPases in Skeletal Muscle Development and Homeostasis. Cells 2021; 10:cells10112984. [PMID: 34831205 PMCID: PMC8616218 DOI: 10.3390/cells10112984] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Rho guanosine triphosphate hydrolases (GTPases) are molecular switches that cycle between an inactive guanosine diphosphate (GDP)-bound and an active guanosine triphosphate (GTP)-bound state during signal transduction. As such, they regulate a wide range of both cellular and physiological processes. In this review, we will summarize recent work on the role of Rho GTPase-regulated pathways in skeletal muscle development, regeneration, tissue mass homeostatic balance, and metabolism. In addition, we will present current evidence that links the dysregulation of these GTPases with diseases caused by skeletal muscle dysfunction. Overall, this information underscores the critical role of a number of members of the Rho GTPase subfamily in muscle development and the overall metabolic balance of mammalian species.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
- Correspondence: or
| | - Xosé R. Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
92
|
Sladky VC, Eichin F, Reiberger T, Villunger A. Polyploidy control in hepatic health and disease. J Hepatol 2021; 75:1177-1191. [PMID: 34228992 DOI: 10.1016/j.jhep.2021.06.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/25/2021] [Accepted: 06/15/2021] [Indexed: 12/24/2022]
Abstract
A balanced increase in DNA content (ploidy) is observed in some human cell types, including bone-resorbing osteoclasts, platelet-producing megakaryocytes, cardiomyocytes or hepatocytes. The impact of increased hepatocyte ploidy on normal physiology and diverse liver pathologies is still poorly understood. Recent findings suggest swift genetic adaptation to hepatotoxic stress and the protection from malignant transformation as beneficial effects. Herein, we discuss the molecular mechanisms regulating hepatocyte polyploidisation and its implication for different liver diseases and hepatocellular carcinoma. We report on centrosomes' role in limiting polyploidy by activating the p53 signalling network (via the PIDDosome multiprotein complex) and we discuss the role of this pathway in liver disease. Increased hepatocyte ploidy is a hallmark of hepatic inflammation and may play a protective role against liver cancer. Our evolving understanding of hepatocyte ploidy is discussed from the perspective of its potential clinical application for risk stratification, prognosis, and novel therapeutic strategies in liver disease and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Valentina C Sladky
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Felix Eichin
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), 1090 Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria.
| |
Collapse
|
93
|
Physiological Roles of Apoptotic Cell Clearance: Beyond Immune Functions. Life (Basel) 2021; 11:life11111141. [PMID: 34833017 PMCID: PMC8621940 DOI: 10.3390/life11111141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
The clearance of apoptotic cells is known to be a critical step in maintaining tissue and organism homeostasis. This process is rapidly/promptly mediated by recruited or resident phagocytes. Phagocytes that engulf apoptotic cells have been closely linked to the release of anti-inflammatory cytokines to eliminate inflammatory responses. Defective clearance of apoptotic cells can cause severe inflammation and autoimmune responses due to secondary necrosis of apoptotic cells. Recently accumulated evidence indicates that apoptotic cells and their clearance have important physiological roles in addition to immune-related functions. Herein, we review the current understanding of the mechanisms and fundamental roles of apoptotic cell clearance and the beneficial roles of apoptotic cells in physiological processes such as differentiation and development.
Collapse
|
94
|
Dittmar T, Weiler J, Luo T, Hass R. Cell-Cell Fusion Mediated by Viruses and HERV-Derived Fusogens in Cancer Initiation and Progression. Cancers (Basel) 2021; 13:5363. [PMID: 34771528 PMCID: PMC8582398 DOI: 10.3390/cancers13215363] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 12/13/2022] Open
Abstract
Cell fusion is a well-known, but still scarcely understood biological phenomenon, which might play a role in cancer initiation, progression and formation of metastases. Although the merging of two (cancer) cells appears simple, the entire process is highly complex, energy-dependent and tightly regulated. Among cell fusion-inducing and -regulating factors, so-called fusogens have been identified as a specific type of proteins that are indispensable for overcoming fusion-associated energetic barriers and final merging of plasma membranes. About 8% of the human genome is of retroviral origin and some well-known fusogens, such as syncytin-1, are expressed by human (cancer) cells. Likewise, enveloped viruses can enable and facilitate cell fusion due to evolutionarily optimized fusogens, and are also capable to induce bi- and multinucleation underlining their fusion capacity. Moreover, multinucleated giant cancer cells have been found in tumors derived from oncogenic viruses. Accordingly, a potential correlation between viruses and fusogens of human endogenous retroviral origin in cancer cell fusion will be summarized in this review.
Collapse
Affiliation(s)
- Thomas Dittmar
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58448 Witten, Germany;
| | - Julian Weiler
- Institute of Immunology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, 58448 Witten, Germany;
| | - Tianjiao Luo
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany;
| | - Ralf Hass
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|
95
|
Hammers DW, Hart CC, Matheny MK, Heimsath EG, Lee YI, Hammer JA, Cheney RE, Sweeney HL. Filopodia powered by class x myosin promote fusion of mammalian myoblasts. eLife 2021; 10:e72419. [PMID: 34519272 PMCID: PMC8500716 DOI: 10.7554/elife.72419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/13/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle fibers are multinucleated cellular giants formed by the fusion of mononuclear myoblasts. Several molecules involved in myoblast fusion have been discovered, and finger-like projections coincident with myoblast fusion have also been implicated in the fusion process. The role of these cellular projections in muscle cell fusion was investigated herein. We demonstrate that these projections are filopodia generated by class X myosin (Myo10), an unconventional myosin motor protein specialized for filopodia. We further show that Myo10 is highly expressed by differentiating myoblasts, and Myo10 ablation inhibits both filopodia formation and myoblast fusion in vitro. In vivo, Myo10 labels regenerating muscle fibers associated with Duchenne muscular dystrophy and acute muscle injury. In mice, conditional loss of Myo10 from muscle-resident stem cells, known as satellite cells, severely impairs postnatal muscle regeneration. Furthermore, the muscle fusion proteins Myomaker and Myomixer are detected in myoblast filopodia. These data demonstrate that Myo10-driven filopodia facilitate multinucleated mammalian muscle formation.
Collapse
MESH Headings
- Animals
- Cell Differentiation
- Cell Fusion
- Cell Line
- Cell Proliferation
- Disease Models, Animal
- Humans
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Knockout
- Muscle Development
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/pathology
- Myosins/genetics
- Myosins/metabolism
- Pseudopodia/genetics
- Pseudopodia/metabolism
- Regeneration
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Time Factors
- Mice
Collapse
Affiliation(s)
- David W Hammers
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - Cora C Hart
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - Michael K Matheny
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - Ernest G Heimsath
- Department of Cell Biology & Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of MedicineChapel HillUnited States
| | - Young il Lee
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| | - John A Hammer
- Cell Biology and Physiology Center, National Heart, Lung and Blood InstituteBethesdaUnited States
| | - Richard E Cheney
- Department of Cell Biology & Physiology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of MedicineChapel HillUnited States
| | - H Lee Sweeney
- Department of Pharmacology & Therapeutics, University of Florida College of MedicineGainesvilleUnited States
- University of Florida Myology InstituteGainesvilleUnited States
| |
Collapse
|
96
|
Hass R, von der Ohe J, Dittmar T. Hybrid Formation and Fusion of Cancer Cells In Vitro and In Vivo. Cancers (Basel) 2021; 13:4496. [PMID: 34503305 PMCID: PMC8431460 DOI: 10.3390/cancers13174496] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/13/2022] Open
Abstract
The generation of cancer hybrid cells by intra-tumoral cell fusion opens new avenues for tumor plasticity to develop cancer stem cells with altered properties, to escape from immune surveillance, to change metastatic behavior, and to broaden drug responsiveness/resistance. Genomic instability and chromosomal rearrangements in bi- or multinucleated aneuploid cancer hybrid cells contribute to these new functions. However, the significance of cell fusion in tumorigenesis is controversial with respect to the low frequency of cancer cell fusion events and a clonal advantage of surviving cancer hybrid cells following a post-hybrid selection process. This review highlights alternative processes of cancer hybrid cell development such as entosis, emperipolesis, cannibalism, therapy-induced polyploidization/endoreduplication, horizontal or lateral gene transfer, and focusses on the predominant mechanisms of cell fusion. Based upon new properties of cancer hybrid cells the arising clinical consequences of the subsequent tumor heterogeneity after cancer cell fusion represent a major therapeutic challenge.
Collapse
Affiliation(s)
- Ralf Hass
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany;
| | - Juliane von der Ohe
- Biochemistry and Tumor Biology Laboratory, Department of Obstetrics and Gynecology, Hannover Medical School, 30625 Hannover, Germany;
| | - Thomas Dittmar
- Institute of Immunology, Center of Biomedical Education and Research (ZABF), Witten/Herdecke University, 58448 Witten, Germany
| |
Collapse
|
97
|
Ng DCH, Ho UY, Grounds MD. Cilia, Centrosomes and Skeletal Muscle. Int J Mol Sci 2021; 22:9605. [PMID: 34502512 PMCID: PMC8431768 DOI: 10.3390/ijms22179605] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022] Open
Abstract
Primary cilia are non-motile, cell cycle-associated organelles that can be found on most vertebrate cell types. Comprised of microtubule bundles organised into an axoneme and anchored by a mature centriole or basal body, primary cilia are dynamic signalling platforms that are intimately involved in cellular responses to their extracellular milieu. Defects in ciliogenesis or dysfunction in cilia signalling underlie a host of developmental disorders collectively referred to as ciliopathies, reinforcing important roles for cilia in human health. Whilst primary cilia have long been recognised to be present in striated muscle, their role in muscle is not well understood. However, recent studies indicate important contributions, particularly in skeletal muscle, that have to date remained underappreciated. Here, we explore recent revelations that the sensory and signalling functions of cilia on muscle progenitors regulate cell cycle progression, trigger differentiation and maintain a commitment to myogenesis. Cilia disassembly is initiated during myoblast fusion. However, the remnants of primary cilia persist in multi-nucleated myotubes, and we discuss their potential role in late-stage differentiation and myofiber formation. Reciprocal interactions between cilia and the extracellular matrix (ECM) microenvironment described for other tissues may also inform on parallel interactions in skeletal muscle. We also discuss emerging evidence that cilia on fibroblasts/fibro-adipogenic progenitors and myofibroblasts may influence cell fate in both a cell autonomous and non-autonomous manner with critical consequences for skeletal muscle ageing and repair in response to injury and disease. This review addresses the enigmatic but emerging role of primary cilia in satellite cells in myoblasts and myofibers during myogenesis, as well as the wider tissue microenvironment required for skeletal muscle formation and homeostasis.
Collapse
Affiliation(s)
- Dominic C. H. Ng
- School of Biomedical Science, Faculty of Medicine, University of Queensland, St Lucia, Brisbane, QLD 4072, Australia;
| | - Uda Y. Ho
- School of Biomedical Science, Faculty of Medicine, University of Queensland, St Lucia, Brisbane, QLD 4072, Australia;
| | - Miranda D. Grounds
- School of Human Sciences, Faculty of Medicine, University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
98
|
Blake C, Massey O, Boyd-Moss M, Firipis K, Rifai A, Franks S, Quigley A, Kapsa R, Nisbet DR, Williams RJ. Replace and repair: Biomimetic bioprinting for effective muscle engineering. APL Bioeng 2021; 5:031502. [PMID: 34258499 PMCID: PMC8270648 DOI: 10.1063/5.0040764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
The debilitating effects of muscle damage, either through ischemic injury or volumetric muscle loss (VML), can have significant impacts on patients, and yet there are few effective treatments. This challenge arises when function is degraded due to significant amounts of skeletal muscle loss, beyond the regenerative ability of endogenous repair mechanisms. Currently available surgical interventions for VML are quite invasive and cannot typically restore function adequately. In response to this, many new bioengineering studies implicate 3D bioprinting as a viable option. Bioprinting for VML repair includes three distinct phases: printing and seeding, growth and maturation, and implantation and application. Although this 3D bioprinting technology has existed for several decades, the advent of more advanced and novel printing techniques has brought us closer to clinical applications. Recent studies have overcome previous limitations in diffusion distance with novel microchannel construct architectures and improved myotubule alignment with highly biomimetic nanostructures. These structures may also enhance angiogenic and nervous ingrowth post-implantation, though further research to improve these parameters has been limited. Inclusion of neural cells has also shown to improve myoblast maturation and development of neuromuscular junctions, bringing us one step closer to functional, implantable skeletal muscle constructs. Given the current state of skeletal muscle 3D bioprinting, the most pressing future avenues of research include furthering our understanding of the physical and biochemical mechanisms of myotube development and expanding our control over macroscopic and microscopic construct structures. Further to this, current investigation needs to be expanded from immunocompromised rodent and murine myoblast models to more clinically applicable human cell lines as we move closer to viable therapeutic implementation.
Collapse
Affiliation(s)
- Cooper Blake
- Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Oliver Massey
- Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | | | | | | | - Stephanie Franks
- Laboratory of Advanced Biomaterials, The Australian National University, Canberra, ACT 2601, Australia
| | | | | | | | | |
Collapse
|
99
|
Jirka C, Pak JH, Grosgogeat CA, Marchetii MM, Gupta VA. Dysregulation of NRAP degradation by KLHL41 contributes to pathophysiology in nemaline myopathy. Hum Mol Genet 2021; 28:2549-2560. [PMID: 30986853 DOI: 10.1093/hmg/ddz078] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/29/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022] Open
Abstract
Nemaline myopathy (NM) is the most common form of congenital myopathy that results in hypotonia and muscle weakness. This disease is clinically and genetically heterogeneous, but three recently discovered genes in NM encode for members of the Kelch family of proteins. Kelch proteins act as substrate-specific adaptors for Cullin 3 (CUL3) E3 ubiquitin ligase to regulate protein turnover through the ubiquitin-proteasome machinery. Defects in thin filament formation and/or stability are key molecular processes that underlie the disease pathology in NM; however, the role of Kelch proteins in these processes in normal and diseases conditions remains elusive. Here, we describe a role of NM causing Kelch protein, KLHL41, in premyofibil-myofibil transition during skeletal muscle development through a regulation of the thin filament chaperone, nebulin-related anchoring protein (NRAP). KLHL41 binds to the thin filament chaperone NRAP and promotes ubiquitination and subsequent degradation of NRAP, a process that is critical for the formation of mature myofibrils. KLHL41 deficiency results in abnormal accumulation of NRAP in muscle cells. NRAP overexpression in transgenic zebrafish resulted in a severe myopathic phenotype and absence of mature myofibrils demonstrating a role in disease pathology. Reducing Nrap levels in KLHL41 deficient zebrafish rescues the structural and function defects associated with disease pathology. We conclude that defects in KLHL41-mediated ubiquitination of sarcomeric proteins contribute to structural and functional deficits in skeletal muscle. These findings further our understanding of how the sarcomere assembly is regulated by disease-causing factors in vivo, which will be imperative for developing mechanism-based specific therapeutic interventions.
Collapse
Affiliation(s)
- Caroline Jirka
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jasmine H Pak
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Claire A Grosgogeat
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Vandana A Gupta
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
100
|
Archacka K, Grabowska I, Mierzejewski B, Graffstein J, Górzyńska A, Krawczyk M, Różycka AM, Kalaszczyńska I, Muras G, Stremińska W, Jańczyk-Ilach K, Walczak P, Janowski M, Ciemerych MA, Brzoska E. Hypoxia preconditioned bone marrow-derived mesenchymal stromal/stem cells enhance myoblast fusion and skeletal muscle regeneration. Stem Cell Res Ther 2021; 12:448. [PMID: 34372911 PMCID: PMC8351116 DOI: 10.1186/s13287-021-02530-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
Background The skeletal muscle reconstruction occurs thanks to unipotent stem cells, i.e., satellite cells. The satellite cells remain quiescent and localized between myofiber sarcolemma and basal lamina. They are activated in response to muscle injury, proliferate, differentiate into myoblasts, and recreate myofibers. The stem and progenitor cells support skeletal muscle regeneration, which could be disturbed by extensive damage, sarcopenia, cachexia, or genetic diseases like dystrophy. Many lines of evidence showed that the level of oxygen regulates the course of cell proliferation and differentiation. Methods In the present study, we analyzed hypoxia impact on human and pig bone marrow-derived mesenchymal stromal cell (MSC) and mouse myoblast proliferation, differentiation, and fusion. Moreover, the influence of the transplantation of human bone marrow-derived MSCs cultured under hypoxic conditions on skeletal muscle regeneration was studied. Results We showed that bone marrow-derived MSCs increased VEGF expression and improved myogenesis under hypoxic conditions in vitro. Transplantation of hypoxia preconditioned bone marrow-derived MSCs into injured muscles resulted in the improved cell engraftment and formation of new vessels. Conclusions We suggested that SDF-1 and VEGF secreted by hypoxia preconditioned bone marrow-derived MSCs played an essential role in cell engraftment and angiogenesis. Importantly, hypoxia preconditioned bone marrow-derived MSCs more efficiently engrafted injured muscles; however, they did not undergo myogenic differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02530-3.
Collapse
Affiliation(s)
- Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Bartosz Mierzejewski
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Joanna Graffstein
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Alicja Górzyńska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Marta Krawczyk
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Anna M Różycka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Ilona Kalaszczyńska
- Department of Histology and Embryology, Medical University of Warsaw, 02-004, Warsaw, Poland.,Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097, Warsaw, Poland
| | - Gabriela Muras
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Władysława Stremińska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Katarzyna Jańczyk-Ilach
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Piotr Walczak
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Warszawska 30 St, 10-082, Olsztyn, Poland.,Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mirosław Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, 21201, USA.,NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5 St, 02-106, Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland.
| |
Collapse
|