1
|
Tobias IC, Moorthy SD, Shchuka VM, Langroudi L, Cherednychenko M, Gillespie ZE, Duncan AG, Tian R, Gajewska NA, Di Roberto RB, Mitchell JA. A Sox2 enhancer cluster regulates region-specific neural fates from mouse embryonic stem cells. G3 (BETHESDA, MD.) 2025; 15:jkaf012. [PMID: 39849901 PMCID: PMC12005160 DOI: 10.1093/g3journal/jkaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
Sex-determining region Y box 2 (Sox2) is a critical transcription factor for embryogenesis and neural stem and progenitor cell (NSPC) maintenance. While distal enhancers control Sox2 in embryonic stem cells (ESCs), enhancers closer to the gene are implicated in Sox2 transcriptional regulation in neural development. We hypothesize that a downstream enhancer cluster, termed Sox2 regulatory regions 2-18 (SRR2-18), regulates Sox2 transcription in neural stem cells and we investigate this in NSPCs derived from mouse ESCs. Using functional genomics and CRISPR-Cas9-mediated deletion analyses, we investigate the role of SRR2-18 in Sox2 regulation during neural differentiation. Transcriptome analyses demonstrate that the loss of even 1 copy of SRR2-18 disrupts the region-specific identity of NSPCs, reducing the expression of genes associated with more anterior regions of the embryonic nervous system. Homozygous deletion of this Sox2 neural enhancer cluster causes reduced SOX2 protein, less frequent interaction with transcriptional machinery, and leads to perturbed chromatin accessibility genome-wide further affecting the expression of neurodevelopmental and anterior-posterior regionalization genes. Furthermore, homozygous NSPC deletants exhibit self-renewal defects and impaired differentiation into cell types found in the brain. Altogether, our data define a cis-regulatory enhancer cluster controlling Sox2 transcription in NSPCs and highlight the sensitivity of neural differentiation processes to decreased Sox2 transcription, which causes differentiation into posterior neural fates, specifically the caudal neural tube. This study highlights the importance of precise Sox2 regulation by SRR2-18 in neural differentiation.
Collapse
Affiliation(s)
- Ian C Tobias
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Sakthi D Moorthy
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Virlana M Shchuka
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Lida Langroudi
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Mariia Cherednychenko
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Zoe E Gillespie
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Andrew G Duncan
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Ruxiao Tian
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Natalia A Gajewska
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Raphaël B Di Roberto
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| |
Collapse
|
2
|
Wang W, Wang Z, Li R, Huang W, Ling Q, Li X, Li Z, Cao M, Zhang Z, Sun Q, Liang Z, Zhang HA, Jiang X, Lin C, Chen Y, Zhao B, Zhao Y, Pan JA, Peng X. RNF138 regulates skeletal muscle differentiation via the Wnt/β-catenin signaling pathway. Theranostics 2025; 15:4446-4464. [PMID: 40225576 PMCID: PMC11984406 DOI: 10.7150/thno.110925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
Rationale: Myogenesis is a strictly regulated process driven by signaling pathways activating muscle-specific gene expression. During myogenesis, muscle stem cells exhibit DNA damage response (DDR) features, which are essential for myoblast differentiation and skeletal muscle regeneration. However, the specific roles of DDR-associated proteins in these processes are not yet fully understood. Methods: Gene knockdown and knockout were used in cell and animal models to study RNF138's function in myoblast differentiation and skeletal muscle regeneration. Multi-omics profiling, including transcriptomics and proteomics, was conducted to identify the key proteins regulated by RNF138 in myogenesis. Protein turnover assays were utilized to investigate RNF138's role in APC protein turnover. Immunofluorescence microscopy was performed to confirm the protein colocalization and subcellular localization. Results: RNF138 expression increases during myoblast differentiation and in regenerating myofibers following muscle injury. Knockdown of RNF138 in C2C12 myoblasts impairs myogenic differentiation and fusion. Additionally, Rnf138-deficient mice exhibit delayed muscle regeneration following cardiotoxin-induced injury. Multi-omics profiling, including transcriptomics and proteomics, reveals that Wnt/β-catenin signaling, a key driver of myogenic differentiation, is enhanced by RNF138. Mechanistically, RNF138 stabilizes β-catenin and enhances its nuclear localization by facilitating lysosomal degradation of APC, a component of the β-catenin degradation complex responsible for mediating the export of β-catenin from the nucleus to the cytoplasm for further ubiquitin-proteasome degradation. Conclusions: We reveal a noncanonical role for RNF138, an E3 ubiquitin ligase, as a positive regulator of myoblast differentiation and skeletal muscle regeneration via the Wnt/β-catenin pathway. This finding highlights the noncanonical function of RNF138 beyond its known roles in DDR and other cellular processes. Therefore, RNF138 provides a potential link between DDR and myoblast differentiation, offering new insights into the molecular regulation of muscle regeneration.
Collapse
Affiliation(s)
- Wenhao Wang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Zhuohua Wang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Rourong Li
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Weiyi Huang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Qiao Ling
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Xiaoxiao Li
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Zan Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Manqi Cao
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Zhihui Zhang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Qingrong Sun
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Zhijuan Liang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Hua-an Zhang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Xuan Jiang
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Chuwen Lin
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Yaoqing Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Bo Zhao
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Yu Zhao
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Ji-An Pan
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
- The Center for Infection and Immunity Study, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| | - Xiaoxue Peng
- The Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
- The Center for Infection and Immunity Study, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, No. 66, Gongchang Road, Guangming District, Shenzhen, Guangdong 518107, China
| |
Collapse
|
3
|
Wei Z, Babkirk K, Chen S, Pei M. Epithelial-to-mesenchymal transition transcription factors: New strategies for mesenchymal tissue regeneration. Cytokine Growth Factor Rev 2025:S1359-6101(25)00032-2. [PMID: 40011185 DOI: 10.1016/j.cytogfr.2025.02.001] [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: 01/03/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
The epithelial-mesenchymal transition transcription factors (EMT-TFs)-ZEB, SNAI, and TWIST families-have been extensively studied in embryonic development and tumor metastasis, providing valuable insight into their roles in cell behavior and transformation. These EMT-TFs have garnered increasing attention in the context of mesenchymal tissue regeneration, potentially contributing an approach for cell therapy. Given that dysregulated EMT-TF expression can impair cell survival and lineage differentiation, controlled regulation of their expression could offer significant advantages for tissue regeneration. However, there is a lack of comprehensive reviews to summarize the influence of the EMT-TFs on mesenchymal tissue regeneration and potential molecular mechanisms. This review explores the regulatory roles of ZEB, SNAI, and TWIST in the regeneration of bone, adipose, cartilage, muscle, and other mesenchymal tissues, with a focus on the underlying molecular signaling mechanisms. Gaining a deeper understanding of how EMT-TFs regulate cell proliferation, apoptosis, migration, and differentiation may offer new insights into the management of mesenchymal tissue repair and open novel avenues for enhancing tissue regeneration.
Collapse
Affiliation(s)
- Zhixin Wei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Kiya Babkirk
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
4
|
Gustafson AL, Durbin AD, Artinger KB, Ford HL. Myogenesis gone awry: the role of developmental pathways in rhabdomyosarcoma. Front Cell Dev Biol 2025; 12:1521523. [PMID: 39902277 PMCID: PMC11788348 DOI: 10.3389/fcell.2024.1521523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/23/2024] [Indexed: 02/05/2025] Open
Abstract
Rhabdomyosarcoma is a soft-tissue sarcoma that occurs most frequently in pediatric patients and has poor survival rates in patients with recurrent or metastatic disease. There are two major sub-types of RMS: fusion-positive (FP-RMS) and fusion-negative (FN-RMS); with FP-RMS typically containing chromosomal translocations between the PAX3/7-FOXO1 loci. Regardless of subtype, RMS resembles embryonic skeletal muscle as it expresses the myogenic regulatory factors (MRFs), MYOD1 and MYOG. During normal myogenesis, these developmental transcription factors (TFs) orchestrate the formation of terminally differentiated, striated, and multinucleated skeletal muscle. However, in RMS these TFs become dysregulated such that they enable the sustained properties of malignancy. In FP-RMS, the PAX3/7-FOXO1 chromosomal translocation results in restructured chromatin, altering the binding of many MRFs and driving an oncogenic state. In FN-RMS, re-expression of MRFs, as well as other myogenic TFs, blocks terminal differentiation and holds cells in a proliferative, stem-cell-like state. In this review, we delve into the myogenic transcriptional networks that are dysregulated in and contribute to RMS progression. Advances in understanding the mechanisms through which myogenesis becomes stalled in RMS will lead to new tumor-specific therapies that target these aberrantly expressed developmental transcriptional pathways.
Collapse
Affiliation(s)
- Annika L. Gustafson
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Adam D. Durbin
- Division of Molecular Oncology, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Kristin B. Artinger
- Department of Diagnostic and Biological Sciences, University of Minnesota School of Dentistry, Minneapolis, MN, United States
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
5
|
Wenlun W, Chaohang Y, Yan H, Wenbin L, Nanqing Z, Qianmin H, Shengcai W, Qing Y, Shirui Y, Feng Z, Lingyun Z. Developing a ceRNA-based lncRNA-miRNA-mRNA regulatory network to uncover roles in skeletal muscle development. FRONTIERS IN BIOINFORMATICS 2025; 4:1494717. [PMID: 39882307 PMCID: PMC11774864 DOI: 10.3389/fbinf.2024.1494717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
The precise role of lncRNAs in skeletal muscle development and atrophy remain elusive. We conducted a bioinformatic analysis of 26 GEO datasets from mouse studies, encompassing embryonic development, postnatal growth, regeneration, cell proliferation, and differentiation, using R and relevant packages (limma et al.). LncRNA-miRNA relationships were predicted using miRcode and lncBaseV2, with miRNA-mRNA pairs identified via miRcode, miRDB, and Targetscan7. Based on the ceRNA theory, we constructed and visualized the lncRNA-miRNA-mRNA regulatory network using ggalluvial among other R packages. GO, Reactome, KEGG, and GSEA explored interactions in muscle development and regeneration. We identified five candidate lncRNAs (Xist, Gas5, Pvt1, Airn, and Meg3) as potential mediators in these processes and microgravity-induced muscle wasting. Additionally, we created a detailed lncRNA-miRNA-mRNA regulatory network, including interactions such as lncRNA Xist/miR-126/IRS1, lncRNA Xist/miR-486-5p/GAB2, lncRNA Pvt1/miR-148/RAB34, and lncRNA Gas5/miR-455-5p/SOCS3. Significant signaling pathway changes (PI3K/Akt, MAPK, NF-κB, cell cycle, AMPK, Hippo, and cAMP) were observed during muscle development, regeneration, and atrophy. Despite bioinformatics challenges, our research underscores the significant roles of lncRNAs in muscle protein synthesis, degradation, cell proliferation, differentiation, function, and metabolism under both normal and microgravity conditions. This study offers new insights into the molecular mechanisms governing skeletal muscle development and regeneration.
Collapse
Affiliation(s)
- Wang Wenlun
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Yu Chaohang
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Huang Yan
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Li Wenbin
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Zhou Nanqing
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Hu Qianmin
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Wu Shengcai
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Yuan Qing
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Yu Shirui
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Zhang Feng
- Department of Food Science and Engineering, Moutai Institute, Renhuai, Guizhou, China
| | - Zhu Lingyun
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| |
Collapse
|
6
|
Sommers O, Tomsine RA, Khacho M. Mitochondrial Dynamics Drive Muscle Stem Cell Progression from Quiescence to Myogenic Differentiation. Cells 2024; 13:1773. [PMID: 39513880 PMCID: PMC11545319 DOI: 10.3390/cells13211773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
From quiescence to activation and myogenic differentiation, muscle stem cells (MuSCs) experience drastic alterations in their signaling activity and metabolism. Through balanced cycles of fission and fusion, mitochondria alter their morphology and metabolism, allowing them to affect their decisive role in modulating MuSC activity and fate decisions. This tightly regulated process contributes to MuSC regulation by mediating changes in redox signaling pathways, cell cycle progression, and cell fate decisions. In this review, we discuss the role of mitochondrial dynamics as an integral modulator of MuSC activity, fate, and maintenance. Understanding the influence of mitochondrial dynamics in MuSCs in health and disease will further the development of therapeutics that support MuSC integrity and thus may aid in restoring the regenerative capacity of skeletal muscle.
Collapse
Affiliation(s)
- Olivia Sommers
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Rholls A. Tomsine
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Mireille Khacho
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Center for Neuromuscular Disease (CNMD), University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Ottawa Institute of Systems Biology (OISB), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
7
|
Price FD, Matyas MN, Gehrke AR, Chen W, Wolin EA, Holton KM, Gibbs RM, Lee A, Singu PS, Sakakeeny JS, Poteracki JM, Goune K, Pfeiffer IT, Boswell SA, Sorger PK, Srivastava M, Pfaff KL, Gussoni E, Buchanan SM, Rubin LL. Organoid culture promotes dedifferentiation of mouse myoblasts into stem cells capable of complete muscle regeneration. Nat Biotechnol 2024:10.1038/s41587-024-02344-7. [PMID: 39261590 DOI: 10.1038/s41587-024-02344-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/27/2024] [Indexed: 09/13/2024]
Abstract
Experimental cell therapies for skeletal muscle conditions have shown little success, primarily because they use committed myogenic progenitors rather than true muscle stem cells, known as satellite cells. Here we present a method to generate in vitro-derived satellite cells (idSCs) from skeletal muscle tissue. When transplanted in small numbers into mouse muscle, mouse idSCs fuse into myofibers, repopulate the satellite cell niche, self-renew, support multiple rounds of muscle regeneration and improve force production on par with freshly isolated satellite cells in damaged skeletal muscle. We compared the epigenomic and transcriptional signatures between idSCs, myoblasts and satellite cells and used these signatures to identify core signaling pathways and genes that confer idSC functionality. Finally, from human muscle biopsies, we successfully generated satellite cell-like cells in vitro. After further development, idSCs may provide a scalable source of cells for the treatment of genetic muscle disorders, trauma-induced muscle damage and age-related muscle weakness.
Collapse
Affiliation(s)
- Feodor D Price
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Systems Biology and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA.
| | - Mark N Matyas
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Andrew R Gehrke
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - William Chen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Erica A Wolin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kristina M Holton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rebecca M Gibbs
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Alice Lee
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Pooja S Singu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jeffrey S Sakakeeny
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - James M Poteracki
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Kelsey Goune
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Isabella T Pfeiffer
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Sarah A Boswell
- Department of Systems Biology and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Peter K Sorger
- Department of Systems Biology and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Mansi Srivastava
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Kathleen Lindahl Pfaff
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Emanuela Gussoni
- Division of Genetics and Genomics and the Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Sean M Buchanan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
8
|
Xia Y, Zhang X, Zhang X, Zhu H, Zhong X, Song W, Yuan J, Sha Z, Li F. Gene structure, expression and function analysis of the MyoD gene in the Pacific white shrimp Litopenaeus vannamei. Gene 2024; 921:148523. [PMID: 38703863 DOI: 10.1016/j.gene.2024.148523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
The Pacific white shrimp Litopenaeus vannamei is a representative species of decapod crustacean and an economically important marine aquaculture species worldwide. However, research on the genes involved in muscle growth and development in shrimp is still lacking. MyoD is recognized as a crucial regulator of myogenesis and plays an essential role in muscle growth and differentiation in various animals. Nonetheless, little information is available concerning the function of this gene among crustaceans. In this study, we identified a sequence of the MyoD gene (LvMyoD) with a conserved bHLH domain in the L. vannamei genome. Phylogenetic analysis revealed that both the overall protein sequence and specific functional sites of LvMyoD are highly conserved with those of other crustacean species and that they are evolutionarily closely related to vertebrate MyoD and Myf5. LvMyoD expression is initially high during early muscle development in shrimp and gradually decreases after 40 days post-larval development. In adults, the muscle-specific expression of LvMyoD was confirmed through RT-qPCR analysis. Knockdown of LvMyoD inhibited the growth of the shrimp in body length and weight. Histological observation and transcriptome sequencing of muscle samples after RNA interference (RNAi) revealed nuclear agglutination and looseness in muscle fibers. Additionally, we observed significant effects on the expression of genes involved in heat shock proteins, myosins, actins, protein synthesis, and glucose metabolism. These findings suggest that LvMyoD plays a critical role in regulating muscle protein synthesis and muscle cell differentiation. Overall, this study highlights the involvement of LvMyoD in myogenesis and muscle growth, suggesting that it is a potentially important regulatory target for shrimp breeding efforts.
Collapse
Affiliation(s)
- Yanting Xia
- Chinese Academy of Sciences (CAS) and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Institute of Aquatic Biotechnology, Collage of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Xiaojun Zhang
- Chinese Academy of Sciences (CAS) and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Institute of Aquatic Biotechnology, Collage of Life Sciences, Qingdao University, Qingdao 266071, China; College of Earth Science, University of Chinese Academy of Sciences, Beijing, 100049, China; Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Xiaoxi Zhang
- Chinese Academy of Sciences (CAS) and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Haochen Zhu
- Chinese Academy of Sciences (CAS) and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Institute of Aquatic Biotechnology, Collage of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Xiaoyun Zhong
- Chinese Academy of Sciences (CAS) and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; College of Earth Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Jianbo Yuan
- Chinese Academy of Sciences (CAS) and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; College of Earth Science, University of Chinese Academy of Sciences, Beijing, 100049, China; Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan 430072, China
| | - Zhenxia Sha
- Institute of Aquatic Biotechnology, Collage of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Fuhua Li
- Chinese Academy of Sciences (CAS) and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; College of Earth Science, University of Chinese Academy of Sciences, Beijing, 100049, China; Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
9
|
Blackburn DM, Sahinyan K, Hernández-Corchado A, Lazure F, Richard V, Raco L, Perron G, Zahedi RP, Borchers CH, Lepper C, Kawabe H, Jahani-Asl A, Najafabadi HS, Soleimani VD. The E3 ubiquitin ligase Nedd4L preserves skeletal muscle stem cell quiescence by inhibiting their activation. iScience 2024; 27:110241. [PMID: 39015146 PMCID: PMC11250905 DOI: 10.1016/j.isci.2024.110241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/14/2023] [Accepted: 06/07/2024] [Indexed: 07/18/2024] Open
Abstract
Adult stem cells play a critical role in tissue repair and maintenance. In tissues with slow turnover, including skeletal muscle, these cells are maintained in a mitotically quiescent state yet remain poised to re-enter the cell cycle to replenish themselves and regenerate the tissue. Using a panomics approach we show that the PAX7/NEDD4L axis acts against muscle stem cell activation in homeostatic skeletal muscle. Our findings suggest that PAX7 transcriptionally activates the E3 ubiquitin ligase Nedd4L and that the conditional genetic deletion of Nedd4L impairs muscle stem cell quiescence, with an upregulation of cell cycle and myogenic differentiation genes. Loss of Nedd4L in muscle stem cells results in the expression of doublecortin (DCX), which is exclusively expressed during their in vivo activation. Together, these data establish that the ubiquitin proteasome system, mediated by Nedd4L, is a key contributor to the muscle stem cell quiescent state in adult mice.
Collapse
Affiliation(s)
- Darren M. Blackburn
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC H3A 0C7, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte- Sainte-Catherine, Montréal, QC H3T 1E2, Canada
| | - Korin Sahinyan
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC H3A 0C7, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte- Sainte-Catherine, Montréal, QC H3T 1E2, Canada
| | - Aldo Hernández-Corchado
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC H3A 0C7, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC H3A 0C7, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte- Sainte-Catherine, Montréal, QC H3T 1E2, Canada
| | - Vincent Richard
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC H3T 1E2, Canada
| | - Laura Raco
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte- Sainte-Catherine, Montréal, QC H3T 1E2, Canada
| | - Gabrielle Perron
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC H3A 0C7, Canada
| | - René P. Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC H3T 1E2, Canada
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB R3E 3P4, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Christoph H. Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC H3T 1E2, Canada
- Gerald Bronfman Department of Oncology, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC H3T 1E2, Canada
- Division of Experimental Medicine, McGill University, Montréal, QC H4A 3J1, Canada
- Department of Pathology, McGill University, Montréal, QC H3A 2B4, Canada
| | - Christoph Lepper
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Hiroshi Kawabe
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine 37075 Göttingen, Germany
| | - Arezu Jahani-Asl
- Department of Cellular and Molecular Medicine and University of Ottawa Brain and Mind Research Institute, 451 Smyth Rd, Ottawa, ON K1H 8M5, Canada
| | - Hamed S. Najafabadi
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC H3A 0C7, Canada
| | - Vahab D. Soleimani
- Department of Human Genetics, McGill University, 3640 rue University, Montréal, QC H3A 0C7, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte- Sainte-Catherine, Montréal, QC H3T 1E2, Canada
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, 451 Smyth Rd, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
10
|
Chen J, Li Q. Emerging role of HDAC11 in skeletal muscle biology. Front Cell Dev Biol 2024; 12:1368171. [PMID: 38859964 PMCID: PMC11163118 DOI: 10.3389/fcell.2024.1368171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/07/2024] [Indexed: 06/12/2024] Open
Abstract
HDAC11 is an epigenetic repressor of gene transcription, acting through its deacetylase activity to remove functional acetyl groups from the lysine residues of histones at genomic loci. It has been implicated in the regulation of different immune responses, metabolic activities, as well as cell cycle progression. Recent studies have also shed lights on the impact of HDAC11 on myogenic differentiation and muscle development, indicating that HDAC11 is important for histone deacetylation at the promoters to inhibit transcription of cell cycle related genes, thereby permitting myogenic activation at the onset of myoblast differentiation. Interestingly, the upstream networks of HDAC11 target genes are mainly associated with cell cycle regulators and the acetylation of histones at the HDAC11 target promoters appears to be residue specific. As such, selective inhibition, or activation of HDAC11 presents a potential therapeutic approach for targeting distinct epigenetic pathways in clinical applications.
Collapse
Affiliation(s)
- Jihong Chen
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Qiao Li
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
11
|
Lyu P, Jiang H. Chromatin profiling reveals TFAP4 as a critical transcriptional regulator of bovine satellite cell differentiation. BMC Genomics 2024; 25:272. [PMID: 38475725 DOI: 10.1186/s12864-024-10189-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Satellite cells are myogenic precursor cells in adult skeletal muscle and play a crucial role in skeletal muscle regeneration, maintenance, and growth. Like embryonic myoblasts, satellite cells have the ability to proliferate, differentiate, and fuse to form multinucleated myofibers. In this study, we aimed to identify additional transcription factors that control gene expression during bovine satellite cell proliferation and differentiation. RESULTS Using chromatin immunoprecipitation followed by sequencing, we identified 56,973 and 54,470 genomic regions marked with both the histone modifications H3K4me1 and H3K27ac, which were considered active enhancers, and 50,956 and 59,174 genomic regions marked with H3K27me3, which were considered repressed enhancers, in proliferating and differentiating bovine satellite cells, respectively. In addition, we identified 1,216 and 1,171 super-enhancers in proliferating and differentiating bovine satellite cells, respectively. Analyzing these enhancers showed that in proliferating bovine satellite cells, active enhancers were associated with genes stimulating cell proliferation or inhibiting myoblast differentiation whereas repressed enhancers were associated with genes essential for myoblast differentiation, and that in differentiating satellite cells, active enhancers were associated with genes essential for myoblast differentiation or muscle contraction whereas repressed enhancers were associated with genes stimulating cell proliferation or inhibiting myoblast differentiation. Active enhancers in proliferating bovine satellite cells were enriched with binding sites for many transcription factors such as MYF5 and the AP-1 family transcription factors; active enhancers in differentiating bovine satellite cells were enriched with binding sites for many transcription factors such as MYOG and TFAP4; and repressed enhancers in both proliferating and differentiating bovine satellite cells were enriched with binding sites for NF-kB, ZEB-1, and several other transcription factors. The role of TFAP4 in satellite cell or myoblast differentiation was previously unknown, and through gene knockdown and overexpression, we experimentally validated a critical role for TFAP4 in the differentiation and fusion of bovine satellite cells into myofibers. CONCLUSIONS Satellite cell proliferation and differentiation are controlled by many transcription factors such as AP-1, TFAP4, NF-kB, and ZEB-1 whose roles in these processes were previously unknown in addition to those transcription factors such as MYF5 and MYOG whose roles in these processes are widely known.
Collapse
Affiliation(s)
- Pengcheng Lyu
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Honglin Jiang
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA.
| |
Collapse
|
12
|
Luo L, Xu N, Fan W, Wu Y, Chen P, Li Z, He Z, Liu H, Lin Y, Zheng G. The TGFβ2-Snail1-miRNA TGFβ2 Circuitry is Critical for the Development of Aggressive Functions in Breast Cancer. Clin Transl Med 2024; 14:e1558. [PMID: 38299307 PMCID: PMC10831563 DOI: 10.1002/ctm2.1558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 02/02/2024] Open
Abstract
There have been contradictory reports on the biological role of transforming growth factor-βs (TGFβs) in breast cancer (BC), especially with regard to their ability to promote epithelial-mesenchymal transition (EMT). Here, we show that TGFβ2 is preferentially expressed in mesenchymal-like BCs and maintains the EMT phenotype, correlating with cancer stem cell-like characteristics, growth, metastasis and chemo-resistance and predicting worse clinical outcomes. However, this is only true in ERα- BC. In ERα+ luminal-type BC, estrogen receptor interacts with p-Smads to block TGFβ signalling. Furthermore, we also identify a microRNAs (miRNAs) signature (miRNAsTGFβ2 ) that is weakened in TGFβ2-overexpressing BC cells. We discover that TGFβ2-Snail1 recruits enhancer of zeste homolog-2 to convert miRNAsTGFβ2 promoters from an active to repressive chromatin configuration and then repress miRNAsTGFβ2 transcription, forming a negative feedback loop. On the other hand, miRNAsTGFβ2 overexpression reverses the mesenchymal-like traits in agreement with the inhibition of TGFβ2-Snail1 signalling in BC cells. These findings clarify the roles of TGFβ2 in BC and suggest novel therapeutic strategies based on the TGFβ2-Snail1-miRNAsTGFβ2 loop for a subset type of human BCs.
Collapse
Affiliation(s)
- Liyun Luo
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Ning Xu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Weina Fan
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Yixuan Wu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Pingping Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Zhihui Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Zhimin He
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Hao Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Ying Lin
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| | - Guopei Zheng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseGuangzhouChina
| |
Collapse
|
13
|
Sreenivas P, Wang L, Wang M, Challa A, Modi P, Hensch NR, Gryder B, Chou HC, Zhao XR, Sunkel B, Moreno-Campos R, Khan J, Stanton BZ, Ignatius MS. A SNAI2/CTCF Interaction is Required for NOTCH1 Expression in Rhabdomyosarcoma. Mol Cell Biol 2023; 43:547-565. [PMID: 37882064 PMCID: PMC10761179 DOI: 10.1080/10985549.2023.2256640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 08/30/2023] [Indexed: 10/27/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a pediatric malignancy of the muscle with characteristics of cells blocked in differentiation. NOTCH1 is an oncogene that promotes self-renewal and blocks differentiation in the fusion negative-RMS sub-type. However, how NOTCH1 expression is transcriptionally maintained in tumors is unknown. Analyses of SNAI2 and CTCF chromatin binding and HiC analyses revealed a conserved SNAI2/CTCF overlapping peak downstream of the NOTCH1 locus marking a sub-topologically associating domain (TAD) boundary. Deletion of the SNAI2-CTCF peak showed that it is essential for NOTCH1 expression and viability of FN-RMS cells. Reintroducing constitutively activated NOTCH1-ΔE in cells with the SNAI2-CTCF peak deleted restored cell-viability. Ablation of SNAI2 using CRISPR/Cas9 reagents resulted in the loss of majority of RD and SMS-CTR FN-RMS cells. However, the few surviving clones that repopulate cultures have recovered NOTCH1. Cells that re-establish NOTCH1 expression after SNAI2 ablation are unable to differentiate robustly as SNAI2 shRNA knockdown cells; yet, SNAI2-ablated cells continued to be exquisitely sensitive to ionizing radiation. Thus, we have uncovered a novel mechanism by which SNAI2 and CTCF maintenance of a sub-TAD boundary promotes rather than represses NOTCH1 expression. Further, we demonstrate that SNAI2 suppression of apoptosis post-radiation is independent of SNAI2/NOTCH1 effects on self-renewal and differentiation.
Collapse
Affiliation(s)
- Prethish Sreenivas
- Greehey Children’s Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Long Wang
- Greehey Children’s Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Meng Wang
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Anil Challa
- Greehey Children’s Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Paulomi Modi
- Greehey Children’s Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Nicole Rae Hensch
- Greehey Children’s Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Berkley Gryder
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Xiang R. Zhao
- Greehey Children’s Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Benjamin Sunkel
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Rodrigo Moreno-Campos
- Greehey Children’s Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Javed Khan
- Pediatric Oncology Branch, NCI, NIH, Bethesda, Maryland, USA
| | - Benjamin Z. Stanton
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children’s Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Myron S. Ignatius
- Greehey Children’s Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| |
Collapse
|
14
|
Li Q, Mach YZ, Hamed M, Khilji S, Chen J. Regulation of HDAC11 gene expression in early myogenic differentiation. PeerJ 2023; 11:e15961. [PMID: 37663282 PMCID: PMC10474826 DOI: 10.7717/peerj.15961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023] Open
Abstract
Histone acetylation and deacetylation affect the patterns of gene expression in cellular differentiation, playing pivotal roles in tissue development and maintenance. For example, the intrinsic histone acetyltransferase activity of transcriptional coactivator p300 is especially required for the expression of myogenic regulatory factors including Myf5 and MyoD, and consequently for skeletal myogenesis. On the other hand, histone deacetylases (HDACs) remove the acetyl group from histones, which is critical for gene repression in stem cell fate transition. Through integrative omic analyses, we found that while some HDACs were differentially expressed at the early stage of skeletal myoblast differentiation, Hdac11 gene expression was significantly enhanced by nuclear receptor signaling. In addition, p300 and MyoD control Hdac11 expression in milieu of normal and signal-enhanced myoblast differentiation. Thus, HDAC11 may be essential to differential gene expression at the onset of myoblast differentiation.
Collapse
Affiliation(s)
- Qiao Li
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Yan Z. Mach
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Munerah Hamed
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Saadia Khilji
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jihong Chen
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
15
|
Qabrati X, Kim I, Ghosh A, Bundschuh N, Noé F, Palmer AS, Bar-Nur O. Transgene-free direct conversion of murine fibroblasts into functional muscle stem cells. NPJ Regen Med 2023; 8:43. [PMID: 37553383 PMCID: PMC10409758 DOI: 10.1038/s41536-023-00317-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 07/21/2023] [Indexed: 08/10/2023] Open
Abstract
Transcription factor-based cellular reprogramming provides an attractive approach to produce desired cell types for regenerative medicine purposes. Such cellular conversions are widely dependent on viral vectors to efficiently deliver and express defined factors in target cells. However, use of viral vectors is associated with unfavorable genomic integrations that can trigger deleterious molecular consequences, rendering this method a potential impediment to clinical applications. Here, we report on a highly efficient transgene-free approach to directly convert mouse fibroblasts into induced myogenic progenitor cells (iMPCs) by overexpression of synthetic MyoD-mRNA in concert with an enhanced small molecule cocktail. First, we performed a candidate compound screen and identified two molecules that enhance fibroblast reprogramming into iMPCs by suppression of the JNK and JAK/STAT pathways. Simultaneously, we developed an optimal transfection protocol to transiently overexpress synthetic MyoD-mRNA in fibroblasts. Combining these two techniques enabled robust and rapid reprogramming of fibroblasts into Pax7 positive iMPCs in as little as 10 days. Nascent transgene-free iMPCs proliferated extensively in vitro, expressed a suite of myogenic stem cell markers, and could differentiate into highly multinucleated and contractile myotubes. Furthermore, using global and single-cell transcriptome assays, we delineated gene expression changes associated with JNK and JAK/STAT pathway inhibition during reprogramming, and identified in iMPCs a Pax7+ stem cell subpopulation resembling satellite cells. Last, transgene-free iMPCs robustly engrafted skeletal muscles of a Duchenne muscular dystrophy mouse model, restoring dystrophin expression in hundreds of myofibers. In summary, this study reports on an improved and clinically safer approach to convert fibroblasts into myogenic stem cells that can efficiently contribute to muscle regeneration in vivo.
Collapse
Affiliation(s)
- Xhem Qabrati
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Inseon Kim
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Adhideb Ghosh
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Nicola Bundschuh
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Falko Noé
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Andrew S Palmer
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
- Institute for Health and Sport, Victoria University, Footscray, VIC, Australia
| | - Ori Bar-Nur
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
16
|
Loder S, Patel N, Morgani S, Sambon M, Leucht P, Levi B. Genetic models for lineage tracing in musculoskeletal development, injury, and healing. Bone 2023; 173:116777. [PMID: 37156345 PMCID: PMC10860167 DOI: 10.1016/j.bone.2023.116777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/07/2023] [Accepted: 04/17/2023] [Indexed: 05/10/2023]
Abstract
Musculoskeletal development and later post-natal homeostasis are highly dynamic processes, marked by rapid structural and functional changes across very short periods of time. Adult anatomy and physiology are derived from pre-existing cellular and biochemical states. Consequently, these early developmental states guide and predict the future of the system as a whole. Tools have been developed to mark, trace, and follow specific cells and their progeny either from one developmental state to the next or between circumstances of health and disease. There are now many such technologies alongside a library of molecular markers which may be utilized in conjunction to allow for precise development of unique cell 'lineages'. In this review, we first describe the development of the musculoskeletal system beginning as an embryonic germ layer and at each of the key developmental stages that follow. We then discuss these structures in the context of adult tissues during homeostasis, injury, and repair. Special focus is given in each of these sections to the key genes involved which may serve as markers of lineage or later in post-natal tissues. We then finish with a technical assessment of lineage tracing and the techniques and technologies currently used to mark cells, tissues, and structures within the musculoskeletal system.
Collapse
Affiliation(s)
- Shawn Loder
- Department of Plastic Surgery, University of Pittsburgh, Scaife Hall, Suite 6B, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Nicole Patel
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
17
|
Pomella S, Danielli SG, Alaggio R, Breunis WB, Hamed E, Selfe J, Wachtel M, Walters ZS, Schäfer BW, Rota R, Shipley JM, Hettmer S. Genomic and Epigenetic Changes Drive Aberrant Skeletal Muscle Differentiation in Rhabdomyosarcoma. Cancers (Basel) 2023; 15:2823. [PMID: 37345159 DOI: 10.3390/cancers15102823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma in children and adolescents, represents an aberrant form of skeletal muscle differentiation. Both skeletal muscle development, as well as regeneration of adult skeletal muscle are governed by members of the myogenic family of regulatory transcription factors (MRFs), which are deployed in a highly controlled, multi-step, bidirectional process. Many aspects of this complex process are deregulated in RMS and contribute to tumorigenesis. Interconnected loops of super-enhancers, called core regulatory circuitries (CRCs), define aberrant muscle differentiation in RMS cells. The transcriptional regulation of MRF expression/activity takes a central role in the CRCs active in skeletal muscle and RMS. In PAX3::FOXO1 fusion-positive (PF+) RMS, CRCs maintain expression of the disease-driving fusion oncogene. Recent single-cell studies have revealed hierarchically organized subsets of cells within the RMS cell pool, which recapitulate developmental myogenesis and appear to drive malignancy. There is a large interest in exploiting the causes of aberrant muscle development in RMS to allow for terminal differentiation as a therapeutic strategy, for example, by interrupting MEK/ERK signaling or by interfering with the epigenetic machinery controlling CRCs. In this review, we provide an overview of the genetic and epigenetic framework of abnormal muscle differentiation in RMS, as it provides insights into fundamental mechanisms of RMS malignancy, its remarkable phenotypic diversity and, ultimately, opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Silvia Pomella
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS Istituto Ospedale Pediatrico Bambino Gesu, Viale San Paolo 15, 00146 Rome, Italy
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Sara G Danielli
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, 8032 Zürich, Switzerland
| | - Rita Alaggio
- Department of Pathology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy
| | - Willemijn B Breunis
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, 8032 Zürich, Switzerland
| | - Ebrahem Hamed
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, 79106 Freiburg, Germany
| | - Joanna Selfe
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London SM2 FNG, UK
| | - Marco Wachtel
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, 8032 Zürich, Switzerland
| | - Zoe S Walters
- Translational Epigenomics Team, Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Beat W Schäfer
- Department of Oncology and Children's Research Center, University Children's Hospital of Zurich, 8032 Zürich, Switzerland
| | - Rossella Rota
- Department of Hematology/Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS Istituto Ospedale Pediatrico Bambino Gesu, Viale San Paolo 15, 00146 Rome, Italy
| | - Janet M Shipley
- Sarcoma Molecular Pathology Team, Divisions of Molecular Pathology and Cancer Therapeutics, The Institute of Cancer Research, London SM2 FNG, UK
| | - Simone Hettmer
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, University Medical Center Freiburg, University of Freiburg, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), 79104 Freiburg, Germany
- Comprehensive Cancer Centre Freiburg (CCCF), University Medical Center Freiburg, 790106 Freiburg, Germany
| |
Collapse
|
18
|
Sahinyan K, Lazure F, Blackburn DM, Soleimani VD. Decline of regenerative potential of old muscle stem cells: contribution to muscle aging. FEBS J 2023; 290:1267-1289. [PMID: 35029021 DOI: 10.1111/febs.16352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/23/2021] [Accepted: 01/11/2022] [Indexed: 01/01/2023]
Abstract
Muscle stem cells (MuSCs) are required for life-long muscle regeneration. In general, aging has been linked to a decline in the numbers and the regenerative potential of MuSCs. Muscle regeneration depends on the proper functioning of MuSCs, which is itself dependent on intricate interactions with its niche components. Aging is associated with both cell-intrinsic and niche-mediated changes, which can be the result of transcriptional, posttranscriptional, or posttranslational alterations in MuSCs or in the components of their niche. The interplay between cell intrinsic alterations in MuSCs and changes in the stem cell niche environment during aging and its impact on the number and the function of MuSCs is an important emerging area of research. In this review, we discuss whether the decline in the regenerative potential of MuSCs with age is the cause or the consequence of aging skeletal muscle. Understanding the effect of aging on MuSCs and the individual components of their niche is critical to develop effective therapeutic approaches to diminish or reverse the age-related defects in muscle regeneration.
Collapse
Affiliation(s)
- Korin Sahinyan
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Darren M Blackburn
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| |
Collapse
|
19
|
Chatterjee D, Cong F, Wang XF, Machado Costa CA, Huang YC, Deng WM. Cell polarity opposes Jak/STAT-mediated Escargot activation that drives intratumor heterogeneity in a Drosophila tumor model. Cell Rep 2023; 42:112061. [PMID: 36709425 PMCID: PMC10374876 DOI: 10.1016/j.celrep.2023.112061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 11/28/2022] [Accepted: 01/18/2023] [Indexed: 01/30/2023] Open
Abstract
In proliferating neoplasms, microenvironment-derived selective pressures promote tumor heterogeneity by imparting diverse capacities for growth, differentiation, and invasion. However, what makes a tumor cell respond to signaling cues differently from a normal cell is not well understood. In the Drosophila ovarian follicle cells, apicobasal-polarity loss induces heterogeneous epithelial multilayering. When exacerbated by oncogenic-Notch expression, this multilayer displays an increased consistency in the occurrence of morphologically distinguishable cells adjacent to the polar follicle cells. Polar cells release the Jak/STAT ligand Unpaired (Upd), in response to which neighboring polarity-deficient cells exhibit a precursor-like transcriptomic state. Among the several regulons active in these cells, we could detect and further validate the expression of Snail family transcription factor Escargot (Esg). We also ascertain a similar relationship between Upd and Esg in normally developing ovaries, where establishment of polarity determines early follicular differentiation. Overall, our results indicate that epithelial-cell polarity acts as a gatekeeper against microenvironmental selective pressures that drive heterogeneity.
Collapse
Affiliation(s)
- Deeptiman Chatterjee
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, USA
| | - Fei Cong
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, USA
| | - Xian-Feng Wang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, USA
| | - Caique Almeida Machado Costa
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, USA
| | - Yi-Chun Huang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, USA
| | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, LA 70112, USA.
| |
Collapse
|
20
|
Cai S, Hu B, Wang X, Liu T, Lin Z, Tong X, Xu R, Chen M, Duo T, Zhu Q, Liang Z, Li E, Chen Y, Li J, Liu X, Mo D. Integrative single-cell RNA-seq and ATAC-seq analysis of myogenic differentiation in pig. BMC Biol 2023; 21:19. [PMID: 36726129 PMCID: PMC9893630 DOI: 10.1186/s12915-023-01519-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Skeletal muscle development is a multistep process whose understanding is central in a broad range of fields and applications, from the potential medical value to human society, to its economic value associated with improvement of agricultural animals. Skeletal muscle initiates in the somites, with muscle precursor cells generated in the dermomyotome and dermomyotome-derived myotome before muscle differentiation ensues, a developmentally regulated process that is well characterized in model organisms. However, the regulation of skeletal muscle ontogeny during embryonic development remains poorly defined in farm animals, for instance in pig. Here, we profiled gene expression and chromatin accessibility in developing pig somites and myotomes at single-cell resolution. RESULTS We identified myogenic cells and other cell types and constructed a differentiation trajectory of pig skeletal muscle ontogeny. Along this trajectory, the dynamic changes in gene expression and chromatin accessibility coincided with the activities of distinct cell type-specific transcription factors. Some novel genes upregulated along the differentiation trajectory showed higher expression levels in muscular dystrophy mice than that in healthy mice, suggesting their involvement in myogenesis. Integrative analysis of chromatin accessibility, gene expression data, and in vitro experiments identified EGR1 and RHOB as critical regulators of pig embryonic myogenesis. CONCLUSIONS Collectively, our results enhance our understanding of the molecular and cellular dynamics in pig embryonic myogenesis and offer a high-quality resource for the further study of pig skeletal muscle development and human muscle disease.
Collapse
Affiliation(s)
- Shufang Cai
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
- Guangdong Key Laboratory of Animal Breeding and Nutrition, State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640 Guangdong China
| | - Bin Hu
- Guangdong Key Laboratory of Animal Breeding and Nutrition, State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640 Guangdong China
| | - Xiaoyu Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Tongni Liu
- Faculty of Forestry, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Zhuhu Lin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Xian Tong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Rong Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Meilin Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Tianqi Duo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Qi Zhu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Ziyun Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Enru Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Jianhao Li
- Guangdong Key Laboratory of Animal Breeding and Nutrition, State Key Laboratory of Livestock and Poultry Breeding, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640 Guangdong China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510006 Guangdong China
| |
Collapse
|
21
|
Kantzer CG, Yang W, Grommisch D, Patil KV, Mak KHM, Shirokova V, Genander M. ID1 and CEBPA coordinate epidermal progenitor cell differentiation. Development 2022; 149:282464. [PMID: 36330928 PMCID: PMC9845743 DOI: 10.1242/dev.201262] [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: 08/26/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
The regulatory circuits that coordinate epidermal differentiation during development are still not fully understood. Here, we report that the transcriptional regulator ID1 is enriched in mouse basal epidermal progenitor cells and find ID1 expression to be diminished upon differentiation. In utero silencing of Id1 impairs progenitor cell proliferation, leads to precocious delamination of targeted progenitor cells and enables differentiated keratinocytes to retain progenitor markers and characteristics. Transcriptional profiling suggests that ID1 acts by mediating adhesion to the basement membrane while inhibiting spinous layer differentiation. Co-immunoprecipitation reveals ID1 binding to transcriptional regulators of the class I bHLH family. We localize bHLH Tcf3, Tcf4 and Tcf12 to epidermal progenitor cells during epidermal stratification and establish TCF3 as a downstream effector of ID1-mediated epidermal proliferation. Finally, we identify crosstalk between CEBPA, a known mediator of epidermal differentiation, and Id1, and demonstrate that CEBPA antagonizes BMP-induced activation of Id1. Our work establishes ID1 as a key coordinator of epidermal development, acting to balance progenitor proliferation with differentiation and unveils how functional crosstalk between CEBPA and Id1 orchestrates epidermal lineage progression.
Collapse
Affiliation(s)
| | - Wei Yang
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - David Grommisch
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Kim Vikhe Patil
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Kylie Hin-Man Mak
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Vera Shirokova
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Maria Genander
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77, Stockholm, Sweden,Author for correspondence ()
| |
Collapse
|
22
|
Battistelli C, Garbo S, Maione R. MyoD-Induced Trans-Differentiation: A Paradigm for Dissecting the Molecular Mechanisms of Cell Commitment, Differentiation and Reprogramming. Cells 2022; 11:3435. [PMID: 36359831 PMCID: PMC9654159 DOI: 10.3390/cells11213435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 10/20/2023] Open
Abstract
The discovery of the skeletal muscle-specific transcription factor MyoD represents a milestone in the field of transcriptional regulation during differentiation and cell-fate reprogramming. MyoD was the first tissue-specific factor found capable of converting non-muscle somatic cells into skeletal muscle cells. A unique feature of MyoD, with respect to other lineage-specific factors able to drive trans-differentiation processes, is its ability to dramatically change the cell fate even when expressed alone. The present review will outline the molecular strategies by which MyoD reprograms the transcriptional regulation of the cell of origin during the myogenic conversion, focusing on the activation and coordination of a complex network of co-factors and epigenetic mechanisms. Some molecular roadblocks, found to restrain MyoD-dependent trans-differentiation, and the possible ways for overcoming these barriers, will also be discussed. Indeed, they are of critical importance not only to expand our knowledge of basic muscle biology but also to improve the generation skeletal muscle cells for translational research.
Collapse
Affiliation(s)
| | | | - Rossella Maione
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| |
Collapse
|
23
|
Lv W, Jiang W, Luo H, Tong Q, Niu X, Liu X, Miao Y, Wang J, Guo Y, Li J, Zhan X, Hou Y, Peng Y, Wang J, Zhao S, Xu Z, Zuo B. Long noncoding RNA lncMREF promotes myogenic differentiation and muscle regeneration by interacting with the Smarca5/p300 complex. Nucleic Acids Res 2022; 50:10733-10755. [PMID: 36200826 PMCID: PMC9561262 DOI: 10.1093/nar/gkac854] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/11/2022] [Accepted: 09/23/2022] [Indexed: 11/12/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) play important roles in the spatial and temporal regulation of muscle development and regeneration. Nevertheless, the determination of their biological functions and mechanisms underlying muscle regeneration remains challenging. Here, we identified a lncRNA named lncMREF (lncRNA muscle regeneration enhancement factor) as a conserved positive regulator of muscle regeneration among mice, pigs and humans. Functional studies demonstrated that lncMREF, which is mainly expressed in differentiated muscle satellite cells, promotes myogenic differentiation and muscle regeneration. Mechanistically, lncMREF interacts with Smarca5 to promote chromatin accessibility when muscle satellite cells are activated and start to differentiate, thereby facilitating genomic binding of p300/CBP/H3K27ac to upregulate the expression of myogenic regulators, such as MyoD and cell differentiation. Our results unravel a novel temporal-specific epigenetic regulation during muscle regeneration and reveal that lncMREF/Smarca5-mediated epigenetic programming is responsible for muscle cell differentiation, which provides new insights into the regulatory mechanism of muscle regeneration.
Collapse
Affiliation(s)
- Wei Lv
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
| | - Wei Jiang
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Hongmei Luo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Qian Tong
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Xiaoyu Niu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Xiao Liu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yang Miao
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Jingnan Wang
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Yiwen Guo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Jianan Li
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Xizhen Zhan
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Yunqing Hou
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Yaxin Peng
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
| | - Jian Wang
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shuhong Zhao
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| | - Zaiyan Xu
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Bo Zuo
- Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, China
| |
Collapse
|
24
|
Serina Secanechia YN, Bergiers I, Rogon M, Arnold C, Descostes N, Le S, López-Anguita N, Ganter K, Kapsali C, Bouilleau L, Gut A, Uzuotaite A, Aliyeva A, Zaugg JB, Lancrin C. Identifying a novel role for the master regulator Tal1 in the Endothelial to Hematopoietic Transition. Sci Rep 2022; 12:16974. [PMID: 36217016 PMCID: PMC9550822 DOI: 10.1038/s41598-022-20906-0] [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: 10/27/2021] [Accepted: 09/20/2022] [Indexed: 12/29/2022] Open
Abstract
Progress in the generation of Hematopoietic Stem and Progenitor Cells (HSPCs) in vitro and ex vivo has been built on the knowledge of developmental hematopoiesis, underscoring the importance of understanding this process. HSPCs emerge within the embryonic vasculature through an Endothelial-to-Hematopoietic Transition (EHT). The transcriptional regulator Tal1 exerts essential functions in the earliest stages of blood development, but is considered dispensable for the EHT. Nevertheless, Tal1 is expressed with its binding partner Lmo2 and it homologous Lyl1 in endothelial and transitioning cells at the time of EHT. Here, we investigated the function of these genes using a mouse embryonic-stem cell (mESC)-based differentiation system to model hematopoietic development. We showed for the first time that the expression of TAL1 in endothelial cells is crucial to ensure the efficiency of the EHT process and a sustained hematopoietic output. Our findings uncover an important function of Tal1 during the EHT, thus filling the current gap in the knowledge of the role of this master gene throughout the whole process of hematopoietic development.
Collapse
Affiliation(s)
- Yasmin Natalia Serina Secanechia
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy
| | - Isabelle Bergiers
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy ,grid.419619.20000 0004 0623 0341Present Address: Therapeutics Discovery, Pharmaceutical Companies of Johnson & Johnson, Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Matt Rogon
- grid.4709.a0000 0004 0495 846XEuropean Molecular Biology Laboratory, Centre for Biomolecular Network Analysis, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Christian Arnold
- grid.4709.a0000 0004 0495 846XEuropean Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Nicolas Descostes
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, Bioinformatics Services, via E. Ramarini 32, 00015 Monterotondo, Italy
| | - Stephanie Le
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy
| | - Natalia López-Anguita
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy ,grid.419538.20000 0000 9071 0620Present Address: Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, Germany
| | - Kerstin Ganter
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy
| | - Chrysi Kapsali
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy
| | - Lea Bouilleau
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy
| | - Aaron Gut
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy
| | - Auguste Uzuotaite
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy
| | - Ayshan Aliyeva
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy
| | - Judith B. Zaugg
- grid.4709.a0000 0004 0495 846XEuropean Molecular Biology Laboratory, Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Christophe Lancrin
- grid.418924.20000 0004 0627 3632European Molecular Biology Laboratory, EMBL Rome - Epigenetics and Neurobiology Unit, via E. Ramarini 32, 00015 Monterotondo, Italy
| |
Collapse
|
25
|
Barutcu AR, Elizalde G, Gonzalez AE, Soni K, Rinn JL, Wagers AJ, Almada AE. Prolonged FOS activity disrupts a global myogenic transcriptional program by altering 3D chromatin architecture in primary muscle progenitor cells. Skelet Muscle 2022; 12:20. [PMID: 35971133 PMCID: PMC9377060 DOI: 10.1186/s13395-022-00303-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/04/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The AP-1 transcription factor, FBJ osteosarcoma oncogene (FOS), is induced in adult muscle satellite cells (SCs) within hours following muscle damage and is required for effective stem cell activation and muscle repair. However, why FOS is rapidly downregulated before SCs enter cell cycle as progenitor cells (i.e., transiently expressed) remains unclear. Further, whether boosting FOS levels in the proliferating progeny of SCs can enhance their myogenic properties needs further evaluation. METHODS We established an inducible, FOS expression system to evaluate the impact of persistent FOS activity in muscle progenitor cells ex vivo. We performed various assays to measure cellular proliferation and differentiation, as well as uncover changes in RNA levels and three-dimensional (3D) chromatin interactions. RESULTS Persistent FOS activity in primary muscle progenitor cells severely antagonizes their ability to differentiate and form myotubes within the first 2 weeks in culture. RNA-seq analysis revealed that ectopic FOS activity in muscle progenitor cells suppressed a global pro-myogenic transcriptional program, while activating a stress-induced, mitogen-activated protein kinase (MAPK) transcriptional signature. Additionally, we observed various FOS-dependent, chromosomal re-organization events in A/B compartments, topologically associated domains (TADs), and genomic loops near FOS-regulated genes. CONCLUSIONS Our results suggest that elevated FOS activity in recently activated muscle progenitor cells perturbs cellular differentiation by altering the 3D chromosome organization near critical pro-myogenic genes. This work highlights the crucial importance of tightly controlling FOS expression in the muscle lineage and suggests that in states of chronic stress or disease, persistent FOS activity in muscle precursor cells may disrupt the muscle-forming process.
Collapse
Affiliation(s)
- A Rasim Barutcu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Present address: Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Gabriel Elizalde
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alfredo E Gonzalez
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Kartik Soni
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - John L Rinn
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Present address: BioFrontiers and Department of Biochemistry, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Albert E Almada
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, USA.
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
26
|
Butler E, Xu L, Rakheja D, Schwettmann B, Toubbeh S, Guo L, Kim J, Skapek SX, Zheng Y. Exon skipping in genes encoding lineage-defining myogenic transcription factors in rhabdomyosarcoma. Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006190. [PMID: 35933111 PMCID: PMC9528969 DOI: 10.1101/mcs.a006190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is a childhood sarcoma composed of myoblast-like cells, which suggests a defect in terminal skeletal muscle differentiation. To explore potential defects in the differentiation program, we searched for mRNA splicing variants in genes encoding transcription factors driving skeletal muscle lineage commitment and differentiation. We studied two RMS cases and identified altered splicing resulting in "skipping" the second of three exons in MYOD1. RNA-Seq data from 42 tumors and additional RMS cell lines revealed exon 2 skipping in both MYOD1 and MYF5 but not in MYF6 or MYOG. Complementary molecular analysis of MYOD1 mRNA found evidence for exon skipping in 5 additional RMS cases. Functional studies showed that so-called MYODΔEx2 protein failed to robustly induce muscle-specific genes, and its ectopic expression conferred a selective advantage in cultured fibroblasts and an RMS xenograft. In summary, we present previously unrecognized exon skipping within MYOD1 and MYF5 in RMS, and we propose that alternative splicing can represent a mechanism to alter the function of these two transcription factors in RMS.
Collapse
Affiliation(s)
- Erin Butler
- University of Texas Southwestern Medical Center;
| | - Lin Xu
- University of Texas Southwestern Medical Center
| | | | | | | | - Lei Guo
- University of Texas Southwestern Medical Center
| | - Jiwoon Kim
- University of Texas Southwestern Medical Center
| | | | | |
Collapse
|
27
|
Shi LL, Zhu KC, Wang HL. Characterization of myogenic regulatory factors, myod and myf5 from Megalobrama amblycephala and the effect of lipopolysaccharide on satellite cells in skeletal muscle. Gene 2022; 834:146608. [PMID: 35659893 DOI: 10.1016/j.gene.2022.146608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 04/27/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022]
Abstract
Myod and Myf5 are muscle-specific basic helix-loop-helix (bHLH) transcription factors that play essential roles in regulating skeletal muscle development and growth. In order to investigate potential function of myod and myf5 of Megalobrama amblycephala, an economically important freshwater fish species, in the present study, we characterized the sequences and expression profiles of M. amblycephala myod and myf5. The open reading frame (ORF) sequences of myod and myf5 encoded 275 and 240 amino acids, respectively, possessing analogous structure with the highly conserved domains, bHLH and C-terminal helix III domains. Spatio-temporal expression patterns revealed that myod and myf5 were predominant in skeletal muscle with the highest expression in white muscle, and the highest at 10 days post-hatching (dph) and the segmentation period, respectively. Furthermore, we evaluated the effects of lipopolysaccharide (LPS) on the expression of muscle-related genes in white and red muscle, and proliferation and differentiation of satellite cells. The myod, myf5 and pax-7 expression generally increased and then decreased with increase of LPS concentration and treatment time in red muscle, while these genes showed inconsistent expression patterns in white muscle. In addition, LPS administration caused the frequency increase of satellite cells in red and white muscle especially at 3 and 7 days after LPS-injection.
Collapse
Affiliation(s)
- Lin-Lin Shi
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China
| | - Ke-Cheng Zhu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300 Guangzhou, Guangdong Province, PR China
| | - Huan-Ling Wang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China.
| |
Collapse
|
28
|
Vicente-García C, Hernández-Camacho JD, Carvajal JJ. Regulation of myogenic gene expression. Exp Cell Res 2022; 419:113299. [DOI: 10.1016/j.yexcr.2022.113299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 12/22/2022]
|
29
|
Dogan SA, Giacchin G, Zito E, Viscomi C. Redox Signaling and Stress in Inherited Myopathies. Antioxid Redox Signal 2022; 37:301-323. [PMID: 35081731 DOI: 10.1089/ars.2021.0266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Reactive oxygen species (ROS) are highly reactive compounds that behave like a double-edged sword; they damage cellular structures and act as second messengers in signal transduction. Mitochondria and endoplasmic reticulum (ER) are interconnected organelles with a central role in ROS production, detoxification, and oxidative stress response. Skeletal muscle is the most abundant tissue in mammals and one of the most metabolically active ones and thus relies mainly on oxidative phosphorylation (OxPhos) to synthesize adenosine triphosphate. The impairment of OxPhos leads to myopathy and increased ROS production, thus affecting both redox poise and signaling. In addition, ROS enter the ER and trigger ER stress and its maladaptive response, which also lead to a myopathic phenotype with mitochondrial involvement. Here, we review the role of ROS signaling in myopathies due to either mitochondrial or ER dysfunction. Recent Advances: Relevant advances have been evolving over the last 10 years on the intricate ROS-dependent pathways that act as modifiers of the disease course in several myopathies. To this end, pathways related to mitochondrial biogenesis, satellite cell differentiation, and ER stress have been studied extensively in myopathies. Critical Issues: The analysis of the chemistry and the exact quantitation, as well as the localization of ROS, are still challenging due to the intrinsic labile nature of ROS and the technical limitations of their sensors. Future Directions: The mechanistic studies of the pathogenesis of mitochondrial and ER-related myopathies offer a unique possibility to discover novel ROS-dependent pathways. Antioxid. Redox Signal. 37, 301-323.
Collapse
Affiliation(s)
- Sukru Anil Dogan
- Department of Molecular Biology and Genetics, Center for Life Sciences and Technologies, Bogazici University, Istanbul, Turkey
| | - Giacomo Giacchin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ester Zito
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.,Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Carlo Viscomi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
30
|
Elia I, Realini G, Di Mauro V, Borghi S, Bottoni L, Tornambè S, Vitiello L, Weiss SJ, Chiariello M, Tamburrini A, Oliviero S, Neri F, Orlandini M, Galvagni F. SNAI1 is upregulated during muscle regeneration and represses FGF21 and ATF3 expression by directly binding their promoters. FASEB J 2022; 36:e22401. [PMID: 35726676 DOI: 10.1096/fj.202200215r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/19/2022] [Accepted: 05/26/2022] [Indexed: 11/11/2022]
Abstract
During skeletal myogenesis, the zinc-finger transcription factors SNAI1 and SNAI2, are expressed in proliferating myoblasts and regulate the transition to terminally differentiated myotubes while repressing pro-differentiation genes. Here, we demonstrate that SNAI1 is upregulated in vivo during the early phase of muscle regeneration induced by bupivacaine injury. Using shRNA-mediated gene silencing in C2C12 myoblasts and whole-transcriptome microarray analysis, we identified a collection of genes belonging to the endoplasmic reticulum (ER) stress pathway whose expression, induced by myogenic differentiation, was upregulated in absence of SNAI1. Among these, key ER stress genes, such as Atf3, Ddit3/Chop, Hspa5/Bip, and Fgf21, a myokine involved in muscle differentiation, were strongly upregulated. Furthermore, by promoter mutant analysis and Chromatin immune precipitation assay, we demonstrated that SNAI1 represses Fgf21 and Atf3 in proliferating myoblasts by directly binding to multiple E boxes in their respective promoter regions. Together, these data describe a new regulatory mechanism of myogenic differentiation involving the direct repressive action of SNAI1 on ER stress and Fgf21 expression, ultimately contributing to maintaining the proliferative and undifferentiated state of myoblasts.
Collapse
Affiliation(s)
- Ines Elia
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giulia Realini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Vittoria Di Mauro
- IRCCS-Humanitas Research Hospital, Rozzano, Italy.,Institute of Genetic and Biomedical Research (IRGB), Milan Unit, National Research Council, Via Fantoli 16/15, Milan, 20138, Italy
| | - Sara Borghi
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA.,Immune Monitoring Laboratory, NYU Langone Health, 550 First Avenue, New York, NY, 10016, USA
| | - Laura Bottoni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Salvatore Tornambè
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Stephen J Weiss
- Division of Genetic Medicine, Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Mario Chiariello
- Istituto di Fisiologia Clinica (IFC), Consiglio Nazionale delle Ricerche (CNR) and Core Research Laboratory (CRL), Istituto per lo Studio, la Prevenzione e la Rete Oncologica (ISPRO), Siena, Italy
| | - Annalaura Tamburrini
- Department of Life Science and Systems Biology, Università degli Studi di Torino, Turin, Italy.,IIGM - Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Italy
| | - Salvatore Oliviero
- Department of Life Science and Systems Biology, Università degli Studi di Torino, Turin, Italy.,IIGM - Italian Institute for Genomic Medicine, c/o IRCCS, Candiolo, Italy
| | - Francesco Neri
- Department of Life Science and Systems Biology, Università degli Studi di Torino, Turin, Italy
| | - Maurizio Orlandini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Federico Galvagni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| |
Collapse
|
31
|
Shirakawa T, Toyono T, Inoue A, Matsubara T, Kawamoto T, Kokabu S. Factors Regulating or Regulated by Myogenic Regulatory Factors in Skeletal Muscle Stem Cells. Cells 2022; 11:cells11091493. [PMID: 35563799 PMCID: PMC9104119 DOI: 10.3390/cells11091493] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/23/2022] Open
Abstract
MyoD, Myf5, myogenin, and MRF4 (also known as Myf6 or herculin) are myogenic regulatory factors (MRFs). MRFs are regarded as master transcription factors that are upregulated during myogenesis and influence stem cells to differentiate into myogenic lineage cells. In this review, we summarize MRFs, their regulatory factors, such as TLE3, NF-κB, and MRF target genes, including non-myogenic genes such as taste receptors. Understanding the function of MRFs and the physiology or pathology of satellite cells will contribute to the development of cell therapy and drug discovery for muscle-related diseases.
Collapse
Affiliation(s)
- Tomohiko Shirakawa
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan; (T.S.); (A.I.); (T.K.)
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan;
| | - Takashi Toyono
- Division of Anatomy, Department of Health Promotion, Kyushu Dental University, Kitakyushu 803-8580, Japan;
| | - Asako Inoue
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan; (T.S.); (A.I.); (T.K.)
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan;
| | - Takuma Matsubara
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan;
| | - Tatsuo Kawamoto
- Division of Orofacial Functions and Orthodontics, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan; (T.S.); (A.I.); (T.K.)
| | - Shoichiro Kokabu
- Division of Molecular Signaling and Biochemistry, Department of Health Improvement, Kyushu Dental University, Kitakyushu 803-8580, Japan;
- Correspondence: ; Tel.: +81-93-582-1131; Fax: +81-93-285-6000
| |
Collapse
|
32
|
Woods LM, Ali FR, Gomez R, Chernukhin I, Marcos D, Parkinson LM, Tayoun ANA, Carroll JS, Philpott A. Elevated ASCL1 activity creates de novo regulatory elements associated with neuronal differentiation. BMC Genomics 2022; 23:255. [PMID: 35366798 PMCID: PMC8977041 DOI: 10.1186/s12864-022-08495-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 03/23/2022] [Indexed: 12/14/2022] Open
Abstract
Background The pro-neural transcription factor ASCL1 is a master regulator of neurogenesis and a key factor necessary for the reprogramming of permissive cell types to neurons. Endogenously, ASCL1 expression is often associated with neuroblast stem-ness. Moreover, ASCL1-mediated reprogramming of fibroblasts to differentiated neurons is commonly achieved using artificially high levels of ASCL1 protein, where ASCL1 acts as an “on-target” pioneer factor. However, the genome-wide effects of enhancing ASCL1 activity in a permissive neurogenic environment has not been thoroughly investigated. Here, we overexpressed ASCL1 in the neuronally-permissive context of neuroblastoma (NB) cells where modest endogenous ASCL1 supports the neuroblast programme. Results Increasing ASCL1 in neuroblastoma cells both enhances binding at existing ASCL1 sites and also leads to creation of numerous additional, lower affinity binding sites. These extensive genome-wide changes in ASCL1 binding result in significant reprogramming of the NB transcriptome, redirecting it from a proliferative neuroblastic state towards one favouring neuronal differentiation. Mechanistically, ASCL1-mediated cell cycle exit and differentiation can be increased further by preventing its multi-site phosphorylation, which is associated with additional changes in genome-wide binding and gene activation profiles. Conclusions Our findings show that enhancing ASCL1 activity in a neurogenic environment both increases binding at endogenous ASCL1 sites and also results in additional binding to new low affinity sites that favours neuronal differentiation over the proliferating neuroblast programme supported by the endogenous protein. These findings have important implications for controlling processes of neurogenesis in cancer and cellular reprogramming. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08495-8.
Collapse
|
33
|
Hsu JY, Danis EP, Nance S, O'Brien JH, Gustafson AL, Wessells VM, Goodspeed AE, Talbot JC, Amacher SL, Jedlicka P, Black JC, Costello JC, Durbin AD, Artinger KB, Ford HL. SIX1 reprograms myogenic transcription factors to maintain the rhabdomyosarcoma undifferentiated state. Cell Rep 2022; 38:110323. [PMID: 35108532 PMCID: PMC8917510 DOI: 10.1016/j.celrep.2022.110323] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/21/2021] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is a pediatric muscle sarcoma characterized by expression of the myogenic lineage transcription factors (TFs) MYOD1 and MYOG. Despite high expression of these TFs, RMS cells fail to terminally differentiate, suggesting the presence of factors that alter their functions. Here, we demonstrate that the developmental TF SIX1 is highly expressed in RMS and critical for maintaining a muscle progenitor-like state. SIX1 loss induces differentiation of RMS cells into myotube-like cells and impedes tumor growth in vivo. We show that SIX1 maintains the RMS undifferentiated state by controlling enhancer activity and MYOD1 occupancy at loci more permissive to tumor growth over muscle differentiation. Finally, we demonstrate that a gene signature derived from SIX1 loss correlates with differentiation status and predicts RMS progression in human disease. Our findings demonstrate a master regulatory role of SIX1 in repression of RMS differentiation via genome-wide alterations in MYOD1 and MYOG-mediated transcription.
Collapse
Affiliation(s)
- Jessica Y Hsu
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, CO, USA; Pharmacology Graduate Program, UC-AMC, Aurora, CO, USA
| | - Etienne P Danis
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, CO, USA; University of Colorado Cancer Center, UC-AMC, Aurora, CO, USA
| | - Stephanie Nance
- Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jenean H O'Brien
- Department of Biology, College of St. Scholastica, Duluth, MN, USA
| | - Annika L Gustafson
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, CO, USA; Molecular Biology Graduate Program, UC-AMC, Aurora, CO, USA
| | | | - Andrew E Goodspeed
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, CO, USA; University of Colorado Cancer Center, UC-AMC, Aurora, CO, USA
| | - Jared C Talbot
- School of Biology and Ecology, University of Maine, Orono, ME, USA
| | - Sharon L Amacher
- Department of Molecular Genetics, Ohio State University, Columbus, OH, USA
| | | | - Joshua C Black
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, CO, USA; Pharmacology Graduate Program, UC-AMC, Aurora, CO, USA
| | - James C Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, CO, USA; Pharmacology Graduate Program, UC-AMC, Aurora, CO, USA; University of Colorado Cancer Center, UC-AMC, Aurora, CO, USA
| | - Adam D Durbin
- Division of Molecular Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kristin B Artinger
- Department of Craniofacial Biology, UC-AMC, Aurora, CO, USA; University of Colorado Cancer Center, UC-AMC, Aurora, CO, USA.
| | - Heide L Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, CO, USA; Pharmacology Graduate Program, UC-AMC, Aurora, CO, USA; University of Colorado Cancer Center, UC-AMC, Aurora, CO, USA.
| |
Collapse
|
34
|
Bianconi V, Mozzetta C. Epigenetic control of muscle stem cells: time for a new dimension. Trends Genet 2022; 38:501-513. [DOI: 10.1016/j.tig.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022]
|
35
|
Kostyuk SV, Proskurnina EV, Ershova ES, Kameneva LV, Malinovskaya EM, Savinova EA, Sergeeva VA, Umriukhin PE, Dolgikh OA, Khakina EA, Kraevaya OA, Troshin PA, Kutsev SI, Veiko NN. The Phosphonate Derivative of C 60 Fullerene Induces Differentiation towards the Myogenic Lineage in Human Adipose-Derived Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22179284. [PMID: 34502190 PMCID: PMC8431706 DOI: 10.3390/ijms22179284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 12/26/2022] Open
Abstract
Inductors of myogenic stem cell differentiation attract attention, as they can be used to treat myodystrophies and post-traumatic injuries. Functionalization of fullerenes makes it possible to obtain water-soluble derivatives with targeted biochemical activity. This study examined the effects of the phosphonate C60 fullerene derivatives on the expression of myogenic transcription factors and myogenic differentiation of human mesenchymal stem cells (MSCs). Uptake of the phosphonate C60 fullerene derivatives in human MSCs, intracellular ROS visualization, superoxide scavenging potential, and the expression of myogenic, adipogenic, and osteogenic differentiation genes were studied. The prolonged MSC incubation (within 7–14 days) with the C60 pentaphoshonate potassium salt promoted their differentiation towards the myogenic lineage. The transcription factors and gene expressions determining myogenic differentiation (MYOD1, MYOG, MYF5, and MRF4) increased, while the expression of osteogenic differentiation factors (BMP2, BMP4, RUNX2, SPP1, and OCN) and adipogenic differentiation factors (CEBPB, LPL, and AP2 (FABP4)) was reduced or did not change. The stimulation of autophagy may be one of the factors contributing to the increased expression of myogenic differentiation genes in MSCs. Autophagy may be caused by intracellular alkalosis and/or short-term intracellular oxidative stress.
Collapse
Affiliation(s)
- Svetlana V. Kostyuk
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Elena V. Proskurnina
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
- Correspondence:
| | - Elizaveta S. Ershova
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Larisa V. Kameneva
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Elena M. Malinovskaya
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Ekaterina A. Savinova
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Vasilina A. Sergeeva
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Pavel E. Umriukhin
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
- Department of Normal Physiology, I.M. Sechenov First Moscow State Medical University (Sechenov University) , Mohovaya Str. 11-4, 125009 Moscow, Russia
| | - Olga A. Dolgikh
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Ekaterina A. Khakina
- A.N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavylova St. 28, B-334, 119991 Moscow, Russia;
| | - Olga A. Kraevaya
- Institute of Problems of Chemical Physics of Russian Academy of Sciences, Semenov Prospect 1, 142432 Chernogolovka (Moscow Region), Russia; (O.A.K.); (P.A.T.)
| | - Pavel A. Troshin
- Institute of Problems of Chemical Physics of Russian Academy of Sciences, Semenov Prospect 1, 142432 Chernogolovka (Moscow Region), Russia; (O.A.K.); (P.A.T.)
| | - Sergey I. Kutsev
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| | - Natalia N. Veiko
- Research Centre for Medical Genetics, ul. Moskvorechye 1, 115522 Moscow, Russia; (S.V.K.); (E.S.E.); (L.V.K.); (E.M.M.); (E.A.S.); (V.A.S.); (P.E.U.); (O.A.D.); (S.I.K.); (N.N.V.)
| |
Collapse
|
36
|
Mechanisms of Binding Specificity among bHLH Transcription Factors. Int J Mol Sci 2021; 22:ijms22179150. [PMID: 34502060 PMCID: PMC8431614 DOI: 10.3390/ijms22179150] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 12/25/2022] Open
Abstract
The transcriptome of every cell is orchestrated by the complex network of interaction between transcription factors (TFs) and their binding sites on DNA. Disruption of this network can result in many forms of organism malfunction but also can be the substrate of positive natural selection. However, understanding the specific determinants of each of these individual TF-DNA interactions is a challenging task as it requires integrating the multiple possible mechanisms by which a given TF ends up interacting with a specific genomic region. These mechanisms include DNA motif preferences, which can be determined by nucleotide sequence but also by DNA’s shape; post-translational modifications of the TF, such as phosphorylation; and dimerization partners and co-factors, which can mediate multiple forms of direct or indirect cooperative binding. Binding can also be affected by epigenetic modifications of putative target regions, including DNA methylation and nucleosome occupancy. In this review, we describe how all these mechanisms have a role and crosstalk in one specific family of TFs, the basic helix-loop-helix (bHLH), with a very conserved DNA binding domain and a similar DNA preferred motif, the E-box. Here, we compile and discuss a rich catalog of strategies used by bHLH to acquire TF-specific genome-wide landscapes of binding sites.
Collapse
|
37
|
Feedback regulation of Notch signaling and myogenesis connected by MyoD-Dll1 axis. PLoS Genet 2021; 17:e1009729. [PMID: 34370738 PMCID: PMC8376015 DOI: 10.1371/journal.pgen.1009729] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/19/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022] Open
Abstract
Muscle precursor cells known as myoblasts are essential for muscle development and regeneration. Notch signaling is an ancient intercellular communication mechanism that plays prominent roles in controlling the myogenic program of myoblasts. Currently whether and how the myogenic cues feedback to refine Notch activities in these cells are largely unknown. Here, by mouse and human gene gain/loss-of-function studies, we report that MyoD directly turns on the expression of Notch-ligand gene Dll1 which activates Notch pathway to prevent precautious differentiation in neighboring myoblasts, while autonomously inhibits Notch to facilitate a myogenic program in Dll1 expressing cells. Mechanistically, we studied cis-regulatory DNA motifs underlying the MyoD-Dll1-Notch axis in vivo by characterizing myogenesis of a novel E-box deficient mouse model, as well as in human cells through CRISPR-mediated interference. These results uncovered the crucial transcriptional mechanism that mediates the reciprocal controls of Notch and myogenesis.
Collapse
|
38
|
Rauch A, Mandrup S. Transcriptional networks controlling stromal cell differentiation. Nat Rev Mol Cell Biol 2021; 22:465-482. [PMID: 33837369 DOI: 10.1038/s41580-021-00357-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 02/02/2023]
Abstract
Stromal progenitors are found in many different tissues, where they play an important role in the maintenance of tissue homeostasis owing to their ability to differentiate into parenchymal cells. These progenitor cells are differentially pre-programmed by their tissue microenvironment but, when cultured and stimulated in vitro, these cells - commonly referred to as mesenchymal stromal cells (MSCs) - exhibit a marked plasticity to differentiate into many different cell lineages. Loss-of-function studies in vitro and in vivo have uncovered the involvement of specific signalling pathways and key transcriptional regulators that work in a sequential and coordinated fashion to activate lineage-selective gene programmes. Recent advances in omics and single-cell technologies have made it possible to obtain system-wide insights into the gene regulatory networks that drive lineage determination and cell differentiation. These insights have important implications for the understanding of cell differentiation, the contribution of stromal cells to human disease and for the development of cell-based therapeutic applications.
Collapse
Affiliation(s)
- Alexander Rauch
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark.
| | - Susanne Mandrup
- Center for Functional Genomics and Tissue Plasticity, Functional Genomics & Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
39
|
Gentile A, Bensimon-Brito A, Priya R, Maischein HM, Piesker J, Guenther S, Gunawan F, Stainier DYR. The EMT transcription factor Snai1 maintains myocardial wall integrity by repressing intermediate filament gene expression. eLife 2021; 10:e66143. [PMID: 34152269 PMCID: PMC8216718 DOI: 10.7554/elife.66143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/07/2021] [Indexed: 12/29/2022] Open
Abstract
The transcription factor Snai1, a well-known regulator of epithelial-to-mesenchymal transition, has been implicated in early cardiac morphogenesis as well as in cardiac valve formation. However, a role for Snai1 in regulating other aspects of cardiac morphogenesis has not been reported. Using genetic, transcriptomic, and chimeric analyses in zebrafish, we find that Snai1b is required in cardiomyocytes for myocardial wall integrity. Loss of snai1b increases the frequency of cardiomyocyte extrusion away from the cardiac lumen. Extruding cardiomyocytes exhibit increased actomyosin contractility basally as revealed by enrichment of p-myosin and α-catenin epitope α-18, as well as disrupted intercellular junctions. Transcriptomic analysis of wild-type and snai1b mutant hearts revealed the dysregulation of intermediate filament genes, including desmin b (desmb) upregulation. Cardiomyocyte-specific desmb overexpression caused increased cardiomyocyte extrusion, recapitulating the snai1b mutant phenotype. Altogether, these results indicate that Snai1 maintains the integrity of the myocardial epithelium, at least in part by repressing desmb expression.
Collapse
Affiliation(s)
- Alessandra Gentile
- Max Planck Institute for Heart and Lung Research, Department of Developmental GeneticsBad NauheimGermany
| | - Anabela Bensimon-Brito
- Max Planck Institute for Heart and Lung Research, Department of Developmental GeneticsBad NauheimGermany
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-MainBad NauheimGermany
| | - Rashmi Priya
- Max Planck Institute for Heart and Lung Research, Department of Developmental GeneticsBad NauheimGermany
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-MainBad NauheimGermany
| | - Hans-Martin Maischein
- Max Planck Institute for Heart and Lung Research, Department of Developmental GeneticsBad NauheimGermany
| | - Janett Piesker
- Max Planck Institute for Heart and Lung Research, Microscopy Service GroupBad NauheimGermany
| | - Stefan Guenther
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-MainBad NauheimGermany
- Max Planck Institute for Heart and Lung Research, Bioinformatics and Deep Sequencing PlatformBad NauheimGermany
| | - Felix Gunawan
- Max Planck Institute for Heart and Lung Research, Department of Developmental GeneticsBad NauheimGermany
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-MainBad NauheimGermany
| | - Didier YR Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental GeneticsBad NauheimGermany
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-MainBad NauheimGermany
| |
Collapse
|
40
|
Blackburn DM, Lazure F, Soleimani VD. SMART approaches for genome-wide analyses of skeletal muscle stem and niche cells. Crit Rev Biochem Mol Biol 2021; 56:284-300. [PMID: 33823731 DOI: 10.1080/10409238.2021.1908950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Muscle stem cells (MuSCs) also called satellite cells are the building blocks of skeletal muscle, the largest tissue in the human body which is formed primarily of myofibers. While MuSCs are the principal cells that directly contribute to the formation of the muscle fibers, their ability to do so depends on critical interactions with a vast array of nonmyogenic cells within their niche environment. Therefore, understanding the nature of communication between MuSCs and their niche is of key importance to understand how the skeletal muscle is maintained and regenerated after injury. MuSCs are rare and therefore difficult to study in vivo within the context of their niche environment. The advent of single-cell technologies, such as switching mechanism at 5' end of the RNA template (SMART) and tagmentation based technologies using hyperactive transposase, afford the unprecedented opportunity to perform whole transcriptome and epigenome studies on rare cells within their niche environment. In this review, we will delve into how single-cell technologies can be applied to the study of MuSCs and muscle-resident niche cells and the impact this can have on our understanding of MuSC biology and skeletal muscle regeneration.
Collapse
Affiliation(s)
- Darren M Blackburn
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montreal, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Canada
| |
Collapse
|
41
|
Kimmel JC, Yi N, Roy M, Hendrickson DG, Kelley DR. Differentiation reveals latent features of aging and an energy barrier in murine myogenesis. Cell Rep 2021; 35:109046. [PMID: 33910007 DOI: 10.1016/j.celrep.2021.109046] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/23/2020] [Accepted: 04/07/2021] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle experiences a decline in lean mass and regenerative potential with age, in part due to intrinsic changes in progenitor cells. However, it remains unclear how age-related changes in progenitors manifest across a differentiation trajectory. Here, we perform single-cell RNA sequencing (RNA-seq) on muscle mononuclear cells from young and aged mice and profile muscle stem cells (MuSCs) and fibro-adipose progenitors (FAPs) after differentiation. Differentiation increases the magnitude of age-related change in MuSCs and FAPs, but it also masks a subset of age-related changes present in progenitors. Using a dynamical systems approach and RNA velocity, we find that aged MuSCs follow the same differentiation trajectory as young cells but stall in differentiation near a commitment decision. Our results suggest that differentiation reveals latent features of aging and that fate commitment decisions are delayed in aged myogenic cells in vitro.
Collapse
Affiliation(s)
- Jacob C Kimmel
- Calico Life Sciences, 1170 Veterans Blvd., South San Francisco, CA 94080, USA.
| | - Nelda Yi
- Calico Life Sciences, 1170 Veterans Blvd., South San Francisco, CA 94080, USA
| | - Margaret Roy
- Calico Life Sciences, 1170 Veterans Blvd., South San Francisco, CA 94080, USA
| | - David G Hendrickson
- Calico Life Sciences, 1170 Veterans Blvd., South San Francisco, CA 94080, USA
| | - David R Kelley
- Calico Life Sciences, 1170 Veterans Blvd., South San Francisco, CA 94080, USA.
| |
Collapse
|
42
|
Bao JM, Dang Q, Lin CJ, Lo UG, Feldkoren B, Dang A, Hernandez E, Li F, Panwar V, Lee CF, Cen JJ, Guan B, Margulis V, Kapur P, Brekken RA, Luo JH, Hsieh JT, Tan WL. SPARC is a key mediator of TGF-β-induced renal cancer metastasis. J Cell Physiol 2021; 236:1926-1938. [PMID: 32780451 DOI: 10.1002/jcp.29975] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/16/2020] [Indexed: 12/15/2022]
Abstract
Aberrant expression of transforming growth factor-β1 (TGF-β1) is associated with renal cell carcinoma (RCC) progression by inducing cancer metastasis. However, the downstream effector(s) in TGF-β signaling pathway is not fully characterized. In the present study, the elevation of secreted protein acidic and rich in cysteine (SPARC) as a TGF-β regulated gene in RCC was identified by applying differentially expressed gene analysis and microarray analysis, we further confirmed this result in several RCC cell lines. Clinically, the expression of these two genes is positively correlated in RCC patient specimens. Furthermore, elevated SPARC expression is found in all the subtypes of RCC and positively correlated with the RCC stage and grade. In contrast, SPARC expression is inversely correlated with overall and disease-free survival of patients with RCC, suggesting SPARC as a potent prognostic marker of RCC patient survival. Knocking down SPARC significantly inhibits RCC cell invasion and metastasis both in vitro and in vivo. Similarly, in vitro cell invasion can be diminished by using a specific monoclonal antibody. Mechanistically, SPARC activates protein kinase B (AKT) pathway leading to elevated expression of matrix metalloproteinase-2 that can facilitate RCC invasion. Altogether, our data support that SPARC is a critical role of TGF-β signaling network underlying RCC progression and a potential therapeutic target as well as a prognostic marker.
Collapse
Affiliation(s)
- Ji-Ming Bao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Qiang Dang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chun-Jung Lin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Boris Feldkoren
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Andrew Dang
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Elizabeth Hernandez
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Fei Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Vandana Panwar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Cheng-Fan Lee
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jun-Jie Cen
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bing Guan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rolf A Brekken
- Division of Surgical Oncology, Department of Surgery and Pharmacology, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jun-Hang Luo
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Wan-Long Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
Park J, Lee J, Song KD, Kim SJ, Kim DC, Lee SC, Son YJ, Choi HW, Shim K. Growth factors improve the proliferation of Jeju black pig muscle cells by regulating myogenic differentiation 1 and growth-related genes. Anim Biosci 2021; 34:1392-1402. [PMID: 33561926 PMCID: PMC8255883 DOI: 10.5713/ab.20.0585] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/12/2020] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE The growth rate of pigs is related to differentiation and proliferation of muscle cells, which are regulated by growth factors and expression of growth-related genes. Thus, the objective of this study was to establish optimal culture conditions for Jeju black pig (JBP) muscle cells and determine the relationship of various factors involved in muscle growth with the proliferation of JBP muscle cells. METHODS Muscles were taken from the femur skeletal muscle of JBP embryos. After isolation of the muscle cells, cells were cultured in a 6-well plate under four different culture conditions to optimize culture conditions for JBP muscle cells. To analyze proliferation rate of JBP muscle cells, these muscle cells were seeded into 6-well plates at a density of 1.5×105 cells per well and cultured for 3 days. Western blot and quantitative real-time polymerase chain reaction were applied to verify the myogenic differentiation 1 (MyoD) expression and growth-related gene expression in JBP muscle cells, respectively. RESULTS We established a muscle cell line from JBP embryos and optimized its culture conditions. These muscle cells were positive for MyoD, but not for paired box 7. The proliferation rate of these muscle cells was significantly higher in a culture medium containing bFGF and epidermal growth factor + basic fibroblast growth factor (EGF+bFGF) than that without a growth factor or containing EGF alone. Treatment with EGF and bFGF significantly induced the expression of MyoD protein, an important transcription factor in muscle cells. Moreover, we checked the changes of expression of growth-related genes in JBP muscle cells by presence or absence of growth factors. Expression level of collagen type XXI alpha 1 gene was changed only when EGF and bFGF were added together to culture media for JBP muscle cells. CONCLUSION Concurrent use of EGF and bFGF increased the expression of MyoD protein, thus regulating the proliferation of JBP muscle cells and the expression of growth-related genes.
Collapse
Affiliation(s)
- Jinryong Park
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju 54896, Korea
| | - Jeongeun Lee
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju 54896, Korea
| | - Ki-Duk Song
- The Animal Molecular Genetics and Breeding Center, Jeonbuk National University, Jeonju 54896, Korea.,Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Korea
| | - Sung-Jo Kim
- Division of Cosmetics and Biotechnology, Hoseo University, Asan 31499, Korea
| | - Dae Cheol Kim
- Livestock Promotion Institute, Jeju Special Self-Governing Province, Jeju 63122, Korea
| | | | | | - Hyun Woo Choi
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Korea.,Department of Animal Science, Jeonbuk National University, Jeonju 54896, Korea
| | - Kwanseob Shim
- Department of Animal Biotechnology, Jeonbuk National University, Jeonju 54896, Korea.,Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
44
|
Dill TL, Carroll A, Pinheiro A, Gao J, Naya FJ. The long noncoding RNA Meg3 regulates myoblast plasticity and muscle regeneration through epithelial-mesenchymal transition. Development 2021; 148:dev.194027. [PMID: 33298462 DOI: 10.1242/dev.194027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
Formation of skeletal muscle is among the most striking examples of cellular plasticity in animal tissue development, and while muscle progenitor cells are reprogrammed by epithelial-mesenchymal transition (EMT) to migrate during embryonic development, the regulation of EMT in post-natal myogenesis remains poorly understood. Here, we demonstrate that the long noncoding RNA (lncRNA) Meg3 regulates EMT in myoblast differentiation and skeletal muscle regeneration. Chronic inhibition of Meg3 in C2C12 myoblasts induced EMT, and suppressed cell state transitions required for differentiation. Furthermore, adenoviral Meg3 knockdown compromised muscle regeneration, which was accompanied by abnormal mesenchymal gene expression and interstitial cell proliferation. Transcriptomic and pathway analyses of Meg3-depleted C2C12 myoblasts and injured skeletal muscle revealed a significant dysregulation of EMT-related genes, and identified TGFβ as a key upstream regulator. Importantly, inhibition of TGFβR1 and its downstream effectors, and the EMT transcription factor Snai2, restored many aspects of myogenic differentiation in Meg3-depleted myoblasts in vitro We further demonstrate that reduction of Meg3-dependent Ezh2 activity results in epigenetic alterations associated with TGFβ activation. Thus, Meg3 regulates myoblast identity to facilitate progression into differentiation.
Collapse
Affiliation(s)
- Tiffany L Dill
- Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, MA 02215, USA
| | - Alina Carroll
- Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, MA 02215, USA
| | - Amanda Pinheiro
- Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, MA 02215, USA
| | - Jiachen Gao
- Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, MA 02215, USA
| | - Francisco J Naya
- Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, MA 02215, USA
| |
Collapse
|
45
|
Massenet J, Gardner E, Chazaud B, Dilworth FJ. Epigenetic regulation of satellite cell fate during skeletal muscle regeneration. Skelet Muscle 2021; 11:4. [PMID: 33431060 PMCID: PMC7798257 DOI: 10.1186/s13395-020-00259-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/20/2020] [Indexed: 12/13/2022] Open
Abstract
In response to muscle injury, muscle stem cells integrate environmental cues in the damaged tissue to mediate regeneration. These environmental cues are tightly regulated to ensure expansion of muscle stem cell population to repair the damaged myofibers while allowing repopulation of the stem cell niche. These changes in muscle stem cell fate result from changes in gene expression that occur in response to cell signaling from the muscle environment. Integration of signals from the muscle environment leads to changes in gene expression through epigenetic mechanisms. Such mechanisms, including post-translational modification of chromatin and nucleosome repositioning, act to make specific gene loci more, or less, accessible to the transcriptional machinery. In youth, the muscle environment is ideally structured to allow for coordinated signaling that mediates efficient regeneration. Both age and disease alter the muscle environment such that the signaling pathways that shape the healthy muscle stem cell epigenome are altered. Altered epigenome reduces the efficiency of cell fate transitions required for muscle repair and contributes to muscle pathology. However, the reversible nature of epigenetic changes holds out potential for restoring cell fate potential to improve muscle repair in myopathies. In this review, we will describe the current knowledge of the mechanisms allowing muscle stem cell fate transitions during regeneration and how it is altered in muscle disease. In addition, we provide some examples of how epigenetics could be harnessed therapeutically to improve regeneration in various muscle pathologies.
Collapse
Affiliation(s)
- Jimmy Massenet
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Rd, Mailbox 511, Ottawa, ON, K1H 8L6, Canada.,Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS 5310, INSERM U1217, 8 Rockefeller Ave, 69008, Lyon, France
| | - Edward Gardner
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Rd, Mailbox 511, Ottawa, ON, K1H 8L6, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L6, Canada
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS 5310, INSERM U1217, 8 Rockefeller Ave, 69008, Lyon, France
| | - F Jeffrey Dilworth
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Rd, Mailbox 511, Ottawa, ON, K1H 8L6, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8L6, Canada. .,LIFE Research Institute, University of Ottawa, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
46
|
Pomella S, Sreenivas P, Gryder BE, Wang L, Milewski D, Cassandri M, Baxi K, Hensch NR, Carcarino E, Song Y, Chou HC, Yohe ME, Stanton BZ, Amadio B, Caruana I, De Stefanis C, De Vito R, Locatelli F, Chen Y, Chen EY, Houghton P, Khan J, Rota R, Ignatius MS. Interaction between SNAI2 and MYOD enhances oncogenesis and suppresses differentiation in Fusion Negative Rhabdomyosarcoma. Nat Commun 2021; 12:192. [PMID: 33420019 PMCID: PMC7794422 DOI: 10.1038/s41467-020-20386-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 11/26/2020] [Indexed: 01/29/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is an aggressive pediatric malignancy of the muscle, that includes Fusion Positive (FP)-RMS harboring PAX3/7-FOXO1 and Fusion Negative (FN)-RMS commonly with RAS pathway mutations. RMS express myogenic master transcription factors MYOD and MYOG yet are unable to terminally differentiate. Here, we report that SNAI2 is highly expressed in FN-RMS, is oncogenic, blocks myogenic differentiation, and promotes growth. MYOD activates SNAI2 transcription via super enhancers with striped 3D contact architecture. Genome wide chromatin binding analysis demonstrates that SNAI2 preferentially binds enhancer elements and competes with MYOD at a subset of myogenic enhancers required for terminal differentiation. SNAI2 also suppresses expression of a muscle differentiation program modulated by MYOG, MEF2, and CDKN1A. Further, RAS/MEK-signaling modulates SNAI2 levels and binding to chromatin, suggesting that the differentiation blockade by oncogenic RAS is mediated in part by SNAI2. Thus, an interplay between SNAI2, MYOD, and RAS prevents myogenic differentiation and promotes tumorigenesis.
Collapse
Affiliation(s)
- Silvia Pomella
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Genetics Branch, NCI, NIH, Bethesda, MD, USA
| | - Prethish Sreenivas
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | | | - Long Wang
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | | | - Matteo Cassandri
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Kunal Baxi
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Nicole R Hensch
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Elena Carcarino
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Young Song
- Genetics Branch, NCI, NIH, Bethesda, MD, USA
| | | | - Marielle E Yohe
- Genetics Branch, NCI, NIH, Bethesda, MD, USA
- Pediatric Oncology Branch, NCI, NIH, Bethesda, MD, USA
| | - Benjamin Z Stanton
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, 43205, USA
| | - Bruno Amadio
- SAFU Laboratory, Translational Research Area, Regina Elena National Cancer Institute, Rome, Italy
| | - Ignazio Caruana
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Rita De Vito
- Department of Pathology Unit, Department of Laboratories, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Departmentof Pediatrics, Sapienza University of Rome, Rome, Italy
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Eleanor Y Chen
- Department of Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Peter Houghton
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA
| | - Javed Khan
- Genetics Branch, NCI, NIH, Bethesda, MD, USA.
| | - Rossella Rota
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Myron S Ignatius
- Greehey Children's Cancer Research Institute, Department of Molecular Medicine, University of Texas Health Sciences Center, San Antonio, Texas, USA.
| |
Collapse
|
47
|
Zhang H, Wen J, Bigot A, Chen J, Shang R, Mouly V, Bi P. Human myotube formation is determined by MyoD-Myomixer/Myomaker axis. SCIENCE ADVANCES 2020; 6:eabc4062. [PMID: 33355126 PMCID: PMC11206528 DOI: 10.1126/sciadv.abc4062] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/27/2020] [Indexed: 06/12/2023]
Abstract
Myoblast fusion is essential for formations of myofibers, the basic cellular and functional units of skeletal muscles. Recent genetic studies in mice identified two long-sought membrane proteins, Myomaker and Myomixer, which cooperatively drive myoblast fusion. It is unknown whether and how human muscles, with myofibers of tremendously larger size, use this mechanism to achieve multinucleations. Here, we report an interesting fusion model of human myoblasts where Myomaker is sufficient to induce low-grade fusion, while Myomixer boosts its efficiency to generate giant myotubes. By CRISPR mutagenesis and biochemical assays, we identified MyoD as the key molecular switch of fusion that is required and sufficient to initiate Myomixer and Myomaker expression. Mechanistically, we defined the E-box motifs on promoters of Myomixer and Myomaker by which MyoD induces their expression for multinucleations of human muscle cells. Together, our study uncovered the key molecular apparatus and the transcriptional control mechanism underlying human myoblast fusion.
Collapse
Affiliation(s)
- Haifeng Zhang
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA
| | - Junfei Wen
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA
| | - Anne Bigot
- Center for Research in Myology UMRS974, Sorbonne Université, INSERM, Myology Institute AIM, Paris, France
| | - Jiacheng Chen
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA
| | - Renjie Shang
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Vincent Mouly
- Center for Research in Myology UMRS974, Sorbonne Université, INSERM, Myology Institute AIM, Paris, France
| | - Pengpeng Bi
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA.
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
48
|
Marceca GP, Nigita G, Calore F, Croce CM. MicroRNAs in Skeletal Muscle and Hints on Their Potential Role in Muscle Wasting During Cancer Cachexia. Front Oncol 2020; 10:607196. [PMID: 33330108 PMCID: PMC7732629 DOI: 10.3389/fonc.2020.607196] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Cancer-associated cachexia is a heterogeneous, multifactorial syndrome characterized by systemic inflammation, unintentional weight loss, and profound alteration in body composition. The main feature of cancer cachexia is represented by the loss of skeletal muscle tissue, which may or may not be accompanied by significant adipose tissue wasting. Such phenotypic alteration occurs as the result of concomitant increased myofibril breakdown and reduced muscle protein synthesis, actively contributing to fatigue, worsening of quality of life, and refractoriness to chemotherapy. According to the classical view, this condition is primarily triggered by interactions between specific tumor-induced pro-inflammatory cytokines and their cognate receptors expressed on the myocyte membrane. This causes a shift in gene expression of muscle cells, eventually leading to a pronounced catabolic condition and cell death. More recent studies, however, have shown the involvement of regulatory non-coding RNAs in the outbreak of cancer cachexia. In particular, the role exerted by microRNAs is being widely addressed, and several mechanistic studies are in progress. In this review, we discuss the most recent findings concerning the role of microRNAs in triggering or exacerbating muscle wasting in cancer cachexia, while mentioning about possible roles played by long non-coding RNAs and ADAR-mediated miRNA modifications.
Collapse
Affiliation(s)
- Gioacchino P Marceca
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giovanni Nigita
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Federica Calore
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
49
|
Lazure F, Blackburn DM, Corchado AH, Sahinyan K, Karam N, Sharanek A, Nguyen D, Lepper C, Najafabadi HS, Perkins TJ, Jahani-Asl A, Soleimani VD. Myf6/MRF4 is a myogenic niche regulator required for the maintenance of the muscle stem cell pool. EMBO Rep 2020; 21:e49499. [PMID: 33047485 PMCID: PMC7726801 DOI: 10.15252/embr.201949499] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 12/16/2022] Open
Abstract
The function and maintenance of muscle stem cells (MuSCs) are tightly regulated by signals originating from their niche environment. Skeletal myofibers are a principle component of the MuSC niche and are in direct contact with the muscle stem cells. Here, we show that Myf6 establishes a ligand/receptor interaction between muscle stem cells and their associated muscle fibers. Our data show that Myf6 transcriptionally regulates a broad spectrum of myokines and muscle‐secreted proteins in skeletal myofibers, including EGF. EGFR signaling blocks p38 MAP kinase‐induced differentiation of muscle stem cells. Homozygous deletion of Myf6 causes a significant reduction in the ability of muscle to produce EGF, leading to a deregulation in EGFR signaling. Consequently, although Myf6‐knockout mice are born with a normal muscle stem cell compartment, they undergo a progressive reduction in their stem cell pool during postnatal life due to spontaneous exit from quiescence. Taken together, our data uncover a novel role for Myf6 in promoting the expression of key myokines, such as EGF, in the muscle fiber which prevents muscle stem cell exhaustion by blocking their premature differentiation.
Collapse
Affiliation(s)
- Felicia Lazure
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Darren M Blackburn
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Aldo H Corchado
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Korin Sahinyan
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Nabila Karam
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Ahmad Sharanek
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Duy Nguyen
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Christoph Lepper
- Department of Physiology & Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Theodore J Perkins
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Arezu Jahani-Asl
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada.,Faculty of Medicine, Gerald Bronfman Department of Oncology, McGill University, Montréal, QC, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| |
Collapse
|
50
|
Buchanan SM, Price FD, Castiglioni A, Gee AW, Schneider J, Matyas MN, Hayhurst M, Tabebordbar M, Wagers AJ, Rubin LL. Pro-myogenic small molecules revealed by a chemical screen on primary muscle stem cells. Skelet Muscle 2020; 10:28. [PMID: 33036659 PMCID: PMC7547525 DOI: 10.1186/s13395-020-00248-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/14/2020] [Indexed: 11/10/2022] Open
Abstract
Satellite cells are the canonical muscle stem cells that regenerate damaged skeletal muscle. Loss of function of these cells has been linked to reduced muscle repair capacity and compromised muscle health in acute muscle injury and congenital neuromuscular diseases. To identify new pathways that can prevent loss of skeletal muscle function or enhance regenerative potential, we established an imaging-based screen capable of identifying small molecules that promote the expansion of freshly isolated satellite cells. We found several classes of receptor tyrosine kinase (RTK) inhibitors that increased freshly isolated satellite cell numbers in vitro. Further exploration of one of these compounds, the RTK inhibitor CEP-701 (also known as lestaurtinib), revealed potent activity on mouse satellite cells both in vitro and in vivo. This expansion potential was not seen upon exposure of proliferating committed myoblasts or non-myogenic fibroblasts to CEP-701. When delivered subcutaneously to acutely injured animals, CEP-701 increased both the total number of satellite cells and the rate of muscle repair, as revealed by an increased cross-sectional area of regenerating fibers. Moreover, freshly isolated satellite cells expanded ex vivo in the presence of CEP-701 displayed enhanced muscle engraftment potential upon in vivo transplantation. We provide compelling evidence that certain RTKs, and in particular RET, regulate satellite cell expansion during muscle regeneration. This study demonstrates the power of small molecule screens of even rare adult stem cell populations for identifying stem cell-targeting compounds with therapeutic potential.
Collapse
Affiliation(s)
- Sean M Buchanan
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Feodor D Price
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Alessandra Castiglioni
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA.,Cancer Immunology Department, Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Amanda Wagner Gee
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Joel Schneider
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Mark N Matyas
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Monica Hayhurst
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Mohammadsharif Tabebordbar
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Amy J Wagers
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA
| | - Lee L Rubin
- Harvard University Department of Stem Cell and Regenerative Biology, 7 Divinity Ave, Cambridge, MA, 02138, USA.
| |
Collapse
|