1
|
Catapano A, Cimmino F, Petrella L, Pizzella A, D'Angelo M, Ambrosio K, Marino F, Sabbatini A, Petrelli M, Paolini B, Lucchin L, Cavaliere G, Cristino L, Crispino M, Trinchese G, Mollica MP. Iron metabolism and ferroptosis in health and diseases: The crucial role of mitochondria in metabolically active tissues. J Nutr Biochem 2025; 140:109888. [PMID: 40057002 DOI: 10.1016/j.jnutbio.2025.109888] [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: 05/17/2024] [Revised: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/30/2025]
Abstract
Iron is essential in various physiological processes, but its accumulation leads to oxidative stress and cell damage, thus iron homeostasis has to be tightly regulated. Ferroptosis is an iron-dependent non-apoptotic regulated cell death characterized by iron overload and reactive oxygen species accumulation. Mitochondria are organelles playing a crucial role in iron metabolism and involved in ferroptosis. MitoNEET, a protein of mitochondrial outer membrane, is a key element in this process. Ferroptosis, altering iron levels in several metabolically active organs, is linked to several non-communicable diseases. For example, iron overload in the liver leads to hepatic fibrosis and cirrhosis, accelerating non-alcholic fatty liver diseases progression, in the muscle cells contributes to oxidative damage leading to sarcopenia, and in the brain is associated to neurodegeneration. The aim of this review is to investigate the intricate balance of iron regulation focusing on the role of mitochondria and oxidative stress, and analyzing the ferroptosis implications in health and disease.
Collapse
Affiliation(s)
- Angela Catapano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy; Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Lidia Petrella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Katia Ambrosio
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Francesca Marino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Annarita Sabbatini
- Dietetic and Clinical Nutrition Unit, IEO European Institute of Oncology IRCSS, Milan, Italy
| | - Massimiliano Petrelli
- Department of Clinical and Molecular Sciences, Clinic of Endocrinology and Metabolic Diseases, Università Politecnica delle Marche, Ancona, Italy
| | - Barbara Paolini
- Department of Innovation, experimentation and clinical research, Unit of dietetics and clinical nutrition, S. Maria Alle Scotte Hospital, University of Siena, Siena, Italy
| | - Lucio Lucchin
- Dietetics and Clinical Nutrition, Bolzano Health District, Bolzano, Italy
| | - Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | | | | |
Collapse
|
2
|
Li Y, Li C, Sun Q, Liu X, Chen F, Cheung Y, Zhao Y, Xie T, Chazaud B, Sun H, Wang H. Skeletal muscle stem cells modulate niche function in Duchenne muscular dystrophy mouse through YY1-CCL5 axis. Nat Commun 2025; 16:1324. [PMID: 39900599 PMCID: PMC11790879 DOI: 10.1038/s41467-025-56474-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 01/15/2025] [Indexed: 02/05/2025] Open
Abstract
Adult skeletal muscle stem cells (MuSCs) are indispensable for muscle regeneration and tightly regulated by macrophages (MPs) and fibro-adipogenic progenitors (FAPs) in their niche. Deregulated MuSC/MP/FAP interactions and the ensuing inflammation and fibrosis are hallmarks of dystrophic muscle. Here we demonstrate intrinsic deletion of transcription factor Yin Yang 1 (YY1) in MuSCs exacerbates dystrophic pathologies by altering composition and heterogeneity of MPs and FAPs. Further analysis reveals YY1 loss induces expression of immune genes in MuSCs, including C-C motif chemokine ligand 5 (Ccl5). Augmented CCL5 secretion promotes MP recruitment via CCL5/C-C chemokine receptor 5 (CCR5) crosstalk, which subsequently hinders FAP clearance through elevated Transforming growth factor-β1 (TGFβ1). Maraviroc-mediated pharmacological blockade of the CCL5/CCR5 axis effectively mitigates muscle dystrophy and improves muscle performance. Lastly, we demonstrate YY1 represses Ccl5 transcription by binding to its enhancer thus facilitating promoter-enhancer looping. Altogether, our study demonstrates the critical role of MuSCs in actively shaping their niche and provides novel insight into the therapeutic intervention of muscle dystrophy.
Collapse
MESH Headings
- YY1 Transcription Factor/metabolism
- YY1 Transcription Factor/genetics
- Animals
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/cytology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Chemokine CCL5/metabolism
- Chemokine CCL5/genetics
- Mice
- Stem Cell Niche
- Receptors, CCR5/metabolism
- Receptors, CCR5/genetics
- Macrophages/metabolism
- Stem Cells/metabolism
- Mice, Inbred C57BL
- Transforming Growth Factor beta1/metabolism
- Male
- Mice, Inbred mdx
- Mice, Knockout
- Signal Transduction
Collapse
Grants
- 82172436 National Natural Science Foundation of China (National Science Foundation of China)
- 14115319, 14100620, 14106521, 14105823, 14120420, 14103522, 14105123 Research Grants Council, University Grants Committee (RGC, UGC)
- T13-602/21-N Research Grants Council, University Grants Committee (RGC, UGC)
- C6018-19GF Research Grants Council, University Grants Committee (RGC, UGC)
- 10210906, 08190626 Research Grants Council, University Grants Committee (RGC, UGC)
- AoE/M-402/20 Research Grants Council, University Grants Committee (RGC, UGC)
- STG1/E-403/24-N Research Grants Council, University Grants Committee (RGC, UGC)
- National Key R&D Program of China to H.W. (2022YFA0806003) Health and Medical Research Fund (HMRF) from Health Bureau of the Hong Kong Special Administrative Region, China to H.W. (10210906 and 08190626)
Collapse
Affiliation(s)
- Yang Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine Limited, Hong Kong Science Park, Hong Kong SAR, China
| | - Chuhan Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qiang Sun
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine Limited, Hong Kong Science Park, Hong Kong SAR, China
| | - Xingyuan Liu
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fengyuan Chen
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yeelo Cheung
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Zhao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ting Xie
- Center for Tissue Regeneration and Engineering, Division of Life Science, Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Bénédicte Chazaud
- Unité Physiopathologie et Génétique du Neurone et du Muscle, UMR CNRS 5261, Inserm U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Hao Sun
- Warshel Institute for Computational Biology, Faculty of Medicine, Chinese University of Hong Kong (Shenzhen), Guangdong, China.
| | - Huating Wang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China.
- Center for Neuromusculoskeletal Restorative Medicine Limited, Hong Kong Science Park, Hong Kong SAR, China.
| |
Collapse
|
3
|
Shi M, Yang S, Zhao X, Sun D, Li Y, Yang J, Li M, Cai C, Guo X, Li B, Lu C, Cao G. Effect of LncRNA LOC106505926 on myogenesis and Lipogenesis of porcine primary cells. BMC Genomics 2024; 25:530. [PMID: 38816813 PMCID: PMC11137989 DOI: 10.1186/s12864-024-10422-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Skeletal muscle development and fat deposition have important effects on meat quality. The study of regulating skeletal muscle development and fat deposition is of great significance in improving the quality of carcass and meat. In the present study, whole transcriptome sequencing (including RNA-Seq and miRNA-Seq) was performed on the longissimus dorsi muscle (LDM) of Jinfen White pigs at 1, 90, and 180 days of age. RESULTS The results showed that a total of 245 differentially expressed miRNAs were screened in any two comparisons, which may be involved in the regulation of myogenesis. Among them, compared with 1-day-old group, miR-22-5p was significantly up-regulated in 90-day-old group and 180-day-old group. Functional studies demonstrated that miR-22-5p inhibited the proliferation and differentiation of porcine skeletal muscle satellite cells (PSCs). Pearson correlation coefficient analysis showed that long non-coding RNA (lncRNA) LOC106505926 and CXXC5 gene had strong negative correlations with miR-22-5p. The LOC106505926 and CXXC5 were proven to promote the proliferation and differentiation of PSCs, as opposed to miR-22-5p. In terms of mechanism, LOC106505926 functions as a molecular sponge of miR-22-5p to modulate the expression of CXXC5, thereby inhibits the differentiation of PSCs. In addition, LOC106505926 regulates the differentiation of porcine preadipocytes through direct binding with FASN. CONCLUSIONS Collectively, our results highlight the multifaceted regulatory role of LOC106505926 in controlling skeletal muscle and adipose tissue development in pigs and provide new targets for improving the quality of livestock products by regulating skeletal muscle development and fat deposition.
Collapse
Affiliation(s)
- Mingyue Shi
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Shuai Yang
- Shanxi Animal Husbandry Technology Extension Service Center, Taiyuan, 030001, China
| | - Xiaolei Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Di Sun
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Yifei Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Jingxian Yang
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Meng Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Chunbo Cai
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Xiaohong Guo
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Bugao Li
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Chang Lu
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
| | - Guoqing Cao
- College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
| |
Collapse
|
4
|
Liu F, Cao Y, Wang X, Zhang K, Li N, Su Y, Zhang Y, Meng Q. Islr regulates satellite cells asymmetric division through the SPARC/p-ERK1/2 signaling pathway. FASEB J 2024; 38:e23534. [PMID: 38597911 DOI: 10.1096/fj.202302614r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/03/2024] [Accepted: 02/20/2024] [Indexed: 04/11/2024]
Abstract
Satellite cells (SCs) are adult muscle stem cells responsible for muscle regeneration after acute and chronic muscle injuries. The balance between stem cell self-renewal and differentiation determines the kinetics and efficiency of skeletal muscle regeneration. This study assessed the function of Islr in SC asymmetric division. The deletion of Islr reduced muscle regeneration in adult mice by decreasing the SC pool. Islr is pivotal for SC proliferation, and its deletion promoted the asymmetric division of SCs. A mechanistic search revealed that Islr bound to and degraded secreted protein acidic and rich in cysteine (SPARC), which activated p-ERK1/2 signaling required for asymmetric division. These findings demonstrate that Islr is a key regulator of SC division through the SPARC/p-ERK1/2 signaling pathway. These data provide a basis for treating myopathy.
Collapse
Affiliation(s)
- Fan Liu
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yuxin Cao
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Xiong Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Kuo Zhang
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Na Li
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yang Su
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| | - Yali Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qingyong Meng
- State Key Laboratories for Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing, China
| |
Collapse
|
5
|
Pietrangelo T, Cagnin S, Bondi D, Santangelo C, Marramiero L, Purcaro C, Bonadio RS, Di Filippo ES, Mancinelli R, Fulle S, Verratti V, Cheng X. Myalgic encephalomyelitis/chronic fatigue syndrome from current evidence to new diagnostic perspectives through skeletal muscle and metabolic disturbances. Acta Physiol (Oxf) 2024; 240:e14122. [PMID: 38483046 DOI: 10.1111/apha.14122] [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: 11/19/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 04/17/2024]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a demanding medical condition for patients and society. It has raised much more public awareness after the COVID-19 pandemic since ME/CFS and long-COVID patients share many clinical symptoms such as debilitating chronic fatigue. However, unlike long COVID, the etiopathology of ME/CFS remains a mystery despite several decades' research. This review moves from pathophysiology of ME/CFS through the compelling evidence and most interesting hypotheses. It focuses on the pathophysiology of skeletal muscle by proposing the hypothesis that skeletal muscle tissue offers novel opportunities for diagnosis and treatment of this syndrome and that new evidence can help resolve the long-standing debate on terminology.
Collapse
Affiliation(s)
- Tiziana Pietrangelo
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- IIM-Interuniversity Institute of Myology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Stefano Cagnin
- Department of Biology, University of Padua, Padova, Italy
- CIR-Myo Myology Center, University of Padua, Padova, Italy
| | - Danilo Bondi
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- IIM-Interuniversity Institute of Myology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Carmen Santangelo
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- IIM-Interuniversity Institute of Myology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Lorenzo Marramiero
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- IIM-Interuniversity Institute of Myology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Cristina Purcaro
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- IIM-Interuniversity Institute of Myology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | | | - Ester Sara Di Filippo
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- IIM-Interuniversity Institute of Myology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Rosa Mancinelli
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- IIM-Interuniversity Institute of Myology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Stefania Fulle
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- IIM-Interuniversity Institute of Myology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Vittore Verratti
- Department of Psychological, Health and Territorial Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Xuanhong Cheng
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| |
Collapse
|
6
|
Tzimorotas D, Solberg NT, Andreassen RC, Moutsatsou P, Bodiou V, Pedersen ME, Rønning SB. Expansion of bovine skeletal muscle stem cells from spinner flasks to benchtop stirred-tank bioreactors for up to 38 days. Front Nutr 2023; 10:1192365. [PMID: 37609488 PMCID: PMC10442166 DOI: 10.3389/fnut.2023.1192365] [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: 03/23/2023] [Accepted: 07/10/2023] [Indexed: 08/24/2023] Open
Abstract
Introduction Successful long-term expansion of skeletal muscle satellite cells (MuSCs) on a large scale is fundamental for cultivating animal cells for protein production. Prerequisites for efficient cell expansion include maintaining essential native cell activities such as cell adhesion, migration, proliferation, and differentiation while ensuring consistent reproducibility. Method This study investigated the growth of bovine MuSC culture using low-volume spinner flasks and a benchtop stirred-tank bioreactor (STR). Results and discussion Our results showed for the first time the expansion of primary MuSCs for 38 days in a bench-top STR run with low initial seeding density and FBS reduction, supported by increased expression of the satellite cell marker PAX7 and reduced expression of differentiation-inducing genes like MYOG, even without adding p38-MAPK inhibitors. Moreover, the cells retained their ability to proliferate, migrate, and differentiate after enzymatic dissociation from the microcarriers. We also showed reproducible results in a separate biological benchtop STR run.
Collapse
|
7
|
Shams AS, Kyba M. The Satellite Cell Colony Forming Cell Assay as a Tool to Measure Self-Renewal and Differentiation Potential. Methods Mol Biol 2023; 2640:45-55. [PMID: 36995586 DOI: 10.1007/978-1-0716-3036-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
The muscle satellite cell population is responsible for homeostatic maintenance of muscle fibers in response to muscle injury and normal wear and tear. This population is heterogeneous, and its capacity for self-renewal and differentiation can be altered either by mutation of genes that regulate these processes or with natural processes such as aging. The satellite cell colony assay is a facile way to extract information about the proliferation and differentiation potential of individual cells. Here, we provide a detailed protocol for the isolation, single cell plating, culture, and evaluation of colonies derived from single satellite cells. The variables of cell survival (cloning efficiency), proliferative potential (nuclei per colony), and differentiation propensity (ratio of nuclei within myosin heavy chain-positive cytoplasm to total nuclei) can thus be obtained.
Collapse
Affiliation(s)
- Ahmed S Shams
- Department of Pediatrics, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA
| | - Michael Kyba
- Department of Pediatrics, Lillehei Heart Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
8
|
Wang Y, Zhang Z, Jiao W, Wang Y, Wang X, Zhao Y, Fan X, Tian L, Li X, Mi J. Ferroptosis and its role in skeletal muscle diseases. Front Mol Biosci 2022; 9:1051866. [PMID: 36406272 PMCID: PMC9669482 DOI: 10.3389/fmolb.2022.1051866] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Ferroptosis is characterized by the accumulation of iron and lipid peroxidation products, which regulates physiological and pathological processes in numerous organs and tissues. A growing body of research suggests that ferroptosis is a key causative factor in a variety of skeletal muscle diseases, including sarcopenia, rhabdomyolysis, rhabdomyosarcoma, and exhaustive exercise-induced fatigue. However, the relationship between ferroptosis and various skeletal muscle diseases has not been investigated systematically. This review’s objective is to provide a comprehensive summary of the mechanisms and signaling factors that regulate ferroptosis, including lipid peroxidation, iron/heme, amino acid metabolism, and autophagy. In addition, we tease out the role of ferroptosis in the progression of different skeletal muscle diseases and ferroptosis as a potential target for the treatment of multiple skeletal muscle diseases. This review can provide valuable reference for the research on the pathogenesis of skeletal muscle diseases, as well as for clinical prevention and treatment.
Collapse
Affiliation(s)
- Ying Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zepeng Zhang
- Research Center of Traditional Chinese Medicine, The First Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Weikai Jiao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yanyan Wang
- Department of Endocrinology, The First Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xiuge Wang
- Department of Endocrinology, The First Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yunyun Zhao
- Department of Endocrinology, The First Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Xuechun Fan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Lulu Tian
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Xiangyan Li, ; Jia Mi,
| | - Jia Mi
- Department of Endocrinology, The First Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Xiangyan Li, ; Jia Mi,
| |
Collapse
|
9
|
Campbell TM, Dilworth FJ, Allan DS, Trudel G. The Hunt Is On! In Pursuit of the Ideal Stem Cell Population for Cartilage Regeneration. Front Bioeng Biotechnol 2022; 10:866148. [PMID: 35711627 PMCID: PMC9196866 DOI: 10.3389/fbioe.2022.866148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/27/2022] [Indexed: 01/15/2023] Open
Abstract
Cartilage injury and degeneration are hallmarks of osteoarthritis (OA), the most common joint disease. OA is a major contributor to pain, loss of function, and reduced quality of life. Over the last decade, considerable research efforts have focused on cell-based therapies, including several stem cell-derived approaches to reverse the cartilage alterations associated with OA. Although several tissue sources for deriving cell-based therapies have been identified, none of the resident stem cell populations have adequately fulfilled the promise of curing OA. Indeed, many cell products do not contain true stem cells. As well, issues with aggressive marketing efforts, combined with a lack of evidence regarding efficacy, lead the several national regulatory bodies to discontinue the use of stem cell therapy for OA until more robust evidence becomes available. A review of the evidence is timely to address the status of cell-based cartilage regeneration. The promise of stem cell therapy is not new and has been used successfully to treat non-arthritic diseases, such as hematopoietic and muscle disorders. These fields of regenerative therapy have the advantage of a considerable foundation of knowledge in the area of stem cell repair mechanisms, the role of the stem cell niche, and niche-supporting cells. This foundation is lacking in the field of cartilage repair. So, where should we look for the ideal stem cell to regenerate cartilage? It has recently been discovered that cartilage itself may contain a population of SC-like progenitors. Other potential tissues include stem cell-rich dental pulp and the adolescent growth plate, the latter of which contains chondrocyte progenitors essential for producing the cartilage scaffold needed for bone growth. In this article, we review the progress on stem cell therapies for arthritic disorders, focusing on the various stem cell populations previously used for cartilage regeneration, successful cases of stem cell therapies in muscle and hemopoietic disorders, some of the reasons why these other fields have been successful (i.e., "lessons learned" to be applied to OA stem cell therapy), and finally, novel potential sources of stem cells for regenerating damaged cartilage in vivo.
Collapse
Affiliation(s)
- T Mark Campbell
- Elisabeth Bruyère Hospital, Ottawa, ON, Canada
- Bone and Joint Research Laboratory, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - F Jeffrey Dilworth
- Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - David S Allan
- Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - Guy Trudel
- Bone and Joint Research Laboratory, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
- Department of Biochemistry, Immunology and Microbiology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
10
|
Profiling and Functional Analysis of mRNAs during Skeletal Muscle Differentiation in Goats. Animals (Basel) 2022; 12:ani12081048. [PMID: 35454294 PMCID: PMC9024908 DOI: 10.3390/ani12081048] [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: 03/01/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
Skeletal myogenesis is a complicated biological event that involves a succession of tightly controlled gene expressions. In order to identify novel regulators of this process, we performed mRNA-Seq studies of goat skeletal muscle satellite cells (MuSCs) cultured under proliferation (GM) and differentiation (DM1/DM5) conditions. A total of 19,871 goat genes were expressed during these stages, 198 of which represented novel transcripts. Notably, in pairwise comparisons at the different stages, 2551 differentially expressed genes (DEGs) were identified (p < 0.05), including 1560 in GM vs. DM1, 1597 in GM vs. DM5, and 959 in DM1 vs. DM5 DEGs. The time-series expression profile analysis clustered the DEGs into eight gene groups, three of which had significantly upregulated and downregulated patterns (p < 0.05). Functional enrichment analysis showed that DEGs were enriched for essential biological processes such as muscle structure development, muscle contraction, muscle cell development, striated muscle cell differentiation, and myofibril assembly, and were involved in pathways such as the MAPK, Wnt and PPAR signaling pathways. Moreover, the expression of eight DEGs (MYL2, DES, MYOG, FAP, PLK2, ADAM, WWC1, and PRDX1) was validated. These findings offer novel insights into the transcriptional regulation of skeletal myogenesis in goats.
Collapse
|
11
|
Martinez P, Ballarin L, Ereskovsky AV, Gazave E, Hobmayer B, Manni L, Rottinger E, Sprecher SG, Tiozzo S, Varela-Coelho A, Rinkevich B. Articulating the "stem cell niche" paradigm through the lens of non-model aquatic invertebrates. BMC Biol 2022; 20:23. [PMID: 35057814 PMCID: PMC8781081 DOI: 10.1186/s12915-022-01230-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Stem cells (SCs) in vertebrates typically reside in "stem cell niches" (SCNs), morphologically restricted tissue microenvironments that are important for SC survival and proliferation. SCNs are broadly defined by properties including physical location, but in contrast to vertebrates and other "model" organisms, aquatic invertebrate SCs do not have clearly documented niche outlines or properties. Life strategies such as regeneration or asexual reproduction may have conditioned the niche architectural variability in aquatic or marine animal groups. By both establishing the invertebrates SCNs as independent types, yet allowing inclusiveness among them, the comparative analysis will allow the future functional characterization of SCNs.
Collapse
Affiliation(s)
- P Martinez
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
- Institut Català de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| | - L Ballarin
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35100, Padova, Italy
| | - A V Ereskovsky
- Aix Marseille University, Avignon Université, CNRS, IRD, IMBE, Marseille, France
- St. Petersburg State University, Biological Faculty, Universitetskaya emb. 7/9, St. Petersburg, 199034, Russia
- N. K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova Street 26, Moscow, 119334, Russia
| | - E Gazave
- Université de Paris, CNRS, Institut Jacques Monod, F-75006, Paris, France
| | - B Hobmayer
- Department of Zoology and Center of Molecular Biosciences, University of Innsbruck, Technikerstr. 25, 6020, Innsbruck, Austria
| | - L Manni
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35100, Padova, Italy
| | - E Rottinger
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
- Université Côte d'Azur, Federative Research Institute - Marine Resources (IFR MARRES), Nice, France
| | - S G Sprecher
- Department of Biology, University of Fribourg, Chemin du Musee 10, 1700, Fribourg, Switzerland
| | - S Tiozzo
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), Paris, France
| | - A Varela-Coelho
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Av. da República, 2780-157, Oeiras, Portugal
| | - B Rinkevich
- Israel Oceanography and Limnological Research, National Institute of Oceanography, Tel Shikmona, P.O. Box 8030, 31080, Haifa, Israel.
| |
Collapse
|
12
|
Olson LC, Redden JT, Schwartz Z, Cohen DJ, McClure MJ. Advanced Glycation End-Products in Skeletal Muscle Aging. Bioengineering (Basel) 2021; 8:bioengineering8110168. [PMID: 34821734 PMCID: PMC8614898 DOI: 10.3390/bioengineering8110168] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/17/2022] Open
Abstract
Advanced age causes skeletal muscle to undergo deleterious changes including muscle atrophy, fast-to-slow muscle fiber transition, and an increase in collagenous material that culminates in the age-dependent muscle wasting disease known as sarcopenia. Advanced glycation end-products (AGEs) non-enzymatically accumulate on the muscular collagens in old age via the Maillard reaction, potentiating the accumulation of intramuscular collagen and stiffening the microenvironment through collagen cross-linking. This review contextualizes known aspects of skeletal muscle extracellular matrix (ECM) aging, especially the role of collagens and AGE cross-linking, and underpins the motor nerve’s role in this aging process. Specific directions for future research are also discussed, with the understudied role of AGEs in skeletal muscle aging highlighted. Despite more than a half century of research, the role that intramuscular collagen aggregation and cross-linking plays in sarcopenia is well accepted yet not well integrated with current knowledge of AGE’s effects on muscle physiology. Furthermore, the possible impact that motor nerve aging has on intramuscular cross-linking and muscular AGE levels is posited.
Collapse
Affiliation(s)
- Lucas C. Olson
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
- Department of Gerontology, College of Health Professions, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - James T. Redden
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - David J. Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
| | - Michael J. McClure
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
- Correspondence:
| |
Collapse
|
13
|
Translational control by DHX36 binding to 5'UTR G-quadruplex is essential for muscle stem-cell regenerative functions. Nat Commun 2021; 12:5043. [PMID: 34413292 PMCID: PMC8377060 DOI: 10.1038/s41467-021-25170-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 06/06/2021] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle has a remarkable ability to regenerate owing to its resident stem cells (also called satellite cells, SCs). SCs are normally quiescent; when stimulated by damage, they activate and expand to form new fibers. The mechanisms underlying SC proliferative progression remain poorly understood. Here we show that DHX36, a helicase that unwinds RNA G-quadruplex (rG4) structures, is essential for muscle regeneration by regulating SC expansion. DHX36 (initially named RHAU) is barely expressed at quiescence but is highly induced during SC activation and proliferation. Inducible deletion of Dhx36 in adult SCs causes defective proliferation and muscle regeneration after damage. System-wide mapping in proliferating SCs reveals DHX36 binding predominantly to rG4 structures at various regions of mRNAs, while integrated polysome profiling shows that DHX36 promotes mRNA translation via 5′-untranslated region (UTR) rG4 binding. Furthermore, we demonstrate that DHX36 specifically regulates the translation of Gnai2 mRNA by unwinding its 5′ UTR rG4 structures and identify GNAI2 as a downstream effector of DHX36 for SC expansion. Altogether, our findings uncover DHX36 as an indispensable post-transcriptional regulator of SC function and muscle regeneration acting through binding and unwinding rG4 structures at 5′ UTR of target mRNAs. Skeletal muscle stem cells (or satellite cells, SCs) are normally quiescent but activate and expand in response to injury. Here the authors show that induction of DHX36 helicase during SC activation promotes mRNA translation by binding to 5′UTR mRNA G-quadruplexes (rG4) in targets including Gnai2 and unwinding them.
Collapse
|
14
|
Zhu Q, Liang F, Cai S, Luo X, Duo T, Liang Z, He Z, Chen Y, Mo D. KDM4A regulates myogenesis by demethylating H3K9me3 of myogenic regulatory factors. Cell Death Dis 2021; 12:514. [PMID: 34011940 PMCID: PMC8134519 DOI: 10.1038/s41419-021-03799-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/20/2022]
Abstract
Histone lysine demethylase 4A (KDM4A) plays a crucial role in regulating cell proliferation, cell differentiation, development and tumorigenesis. However, little is known about the function of KDM4A in muscle development and regeneration. Here, we found that the conditional ablation of KDM4A in skeletal muscle caused impairment of embryonic and postnatal muscle formation. The loss of KDM4A in satellite cells led to defective muscle regeneration and blocked the proliferation and differentiation of satellite cells. Myogenic differentiation and myotube formation in KDM4A-deficient myoblasts were inhibited. Chromatin immunoprecipitation assay revealed that KDM4A promoted myogenesis by removing the histone methylation mark H3K9me3 at MyoD, MyoG and Myf5 locus. Furthermore, inactivation of KDM4A in myoblasts suppressed myoblast differentiation and accelerated H3K9me3 level. Knockdown of KDM4A in vitro reduced myoblast proliferation through enhancing the expression of the cyclin-dependent kinase inhibitor P21 and decreasing the expression of cell cycle regulator Cyclin D1. Together, our findings identify KDM4A as an important regulator for skeletal muscle development and regeneration, orchestrating myogenic cell proliferation and differentiation.
Collapse
Affiliation(s)
- Qi Zhu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, North Third Road, Higher Education Mega Center, 510006, Guangzhou, Guangdong, China
| | - Feng Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, North Third Road, Higher Education Mega Center, 510006, Guangzhou, Guangdong, China
| | - Shufang Cai
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, North Third Road, Higher Education Mega Center, 510006, Guangzhou, Guangdong, China
| | - Xiaorong Luo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, North Third Road, Higher Education Mega Center, 510006, Guangzhou, Guangdong, China
| | - Tianqi Duo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, North Third Road, Higher Education Mega Center, 510006, Guangzhou, Guangdong, China
| | - Ziyun Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, North Third Road, Higher Education Mega Center, 510006, Guangzhou, Guangdong, China
| | - Zuyong He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, North Third Road, Higher Education Mega Center, 510006, Guangzhou, Guangdong, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, North Third Road, Higher Education Mega Center, 510006, Guangzhou, Guangdong, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, North Third Road, Higher Education Mega Center, 510006, Guangzhou, Guangdong, China.
| |
Collapse
|
15
|
Negative elongation factor regulates muscle progenitor expansion for efficient myofiber repair and stem cell pool repopulation. Dev Cell 2021; 56:1014-1029.e7. [PMID: 33735618 DOI: 10.1016/j.devcel.2021.02.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 01/08/2021] [Accepted: 02/19/2021] [Indexed: 11/24/2022]
Abstract
Negative elongation factor (NELF) is a critical transcriptional regulator that stabilizes paused RNA polymerase to permit rapid gene expression changes in response to environmental cues. Although NELF is essential for embryonic development, its role in adult stem cells remains unclear. In this study, through a muscle-stem-cell-specific deletion, we showed that NELF is required for efficient muscle regeneration and stem cell pool replenishment. In mechanistic studies using PRO-seq, single-cell trajectory analyses and myofiber cultures revealed that NELF works at a specific stage of regeneration whereby it modulates p53 signaling to permit massive expansion of muscle progenitors. Strikingly, transplantation experiments indicated that these progenitors are also necessary for stem cell pool repopulation, implying that they are able to return to quiescence. Thus, we identified a critical role for NELF in the expansion of muscle progenitors in response to injury and revealed that progenitors returning to quiescence are major contributors to the stem cell pool repopulation.
Collapse
|
16
|
Dystrophin Dp71ab is monoclonally expressed in human satellite cells and enhances proliferation of myoblast cells. Sci Rep 2020; 10:17123. [PMID: 33051488 PMCID: PMC7553993 DOI: 10.1038/s41598-020-74157-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Dystrophin Dp71 is the smallest isoform of the DMD gene, mutations in which cause Duchenne muscular dystrophy (DMD). Dp71 has also been shown to have roles in various cellular processes. Stem cell-based therapy may be effective in treating DMD, but the inability to generate a sufficient number of stem cells remains a significant obstacle. Although Dp71 is comprised of many variants, Dp71 in satellite cells has not yet been studied. Here, the full-length Dp71 consisting of 18 exons from exons G1 to 79 was amplified by reverse transcription-PCR from total RNA of human satellite cells. The amplified product showed deletion of both exons 71 and 78 in all sequenced clones, indicating monoclonal expression of Dp71ab. Western blotting of the satellite cell lysate showed a band corresponding to over-expressed Dp71ab. Transfection of a plasmid expressing Dp71ab into human myoblasts significantly enhanced cell proliferation when compared to the cells transfected with the mock plasmid. However, transfection of the Dp71 expression plasmid encoding all 18 exons did not enhance myoblast proliferation. These findings indicated that Dp71ab, but not Dp71, is a molecular enhancer of myoblast proliferation and that transfection with Dp71ab may generate a high yield of stem cells for DMD treatment.
Collapse
|
17
|
Zhang W, Yu L, Han X, Pan J, Deng J, Zhu L, Lu Y, Huang W, Liu S, Li Q, Liu Y. The secretome of human dental pulp stem cells protects myoblasts from hypoxia‑induced injury via the Wnt/β‑catenin pathway. Int J Mol Med 2020; 45:1501-1513. [PMID: 32323739 PMCID: PMC7138287 DOI: 10.3892/ijmm.2020.4525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022] Open
Abstract
Human dental pulp stem cells (hDPSCs) present several advantages, including their ability to be non-invasively harvested without ethical concern. The secretome of hDPSCs can promote the functional recovery of various tissue injuries. However, the protective effects on hypoxia-induced skeletal muscle injury remain to be explored. The present study demonstrated that C2C12 myoblast coculture with hDPSCs attenuated CoCl2-induced hypoxic injury compared with C2C12 alone. The hDPSC secretome increased cell viability and differentiation and decreased G2/M cell cycle arrest under hypoxic conditions. These results were further verified using hDPSC-conditioned medium (hDPSC-CM). The present data revealed that the protective effects of hDPSC-CM depend on the concentration ratio of the CM. In terms of the underlying molecular mechanism, hDPSC-CM activated the Wnt/β-catenin pathway, which increased the protein levels of Wnt1, phosphorylated-glycogen synthase kinase-3β and β-catenin and the mRNA levels of Wnt target genes. By contrast, an inhibitor (XAV939) of Wnt/β-catenin diminished the protective effects of hDPSC-CM. Taken together, the findings of the present study demonstrated that the hDPSC secretome alleviated the hypoxia-induced myoblast injury potentially through regulating the Wnt/β-catenin pathway. These findings may provide new insight into a therapeutic alternative using the hDPSC secretome in skeletal muscle hypoxia-related diseases.
Collapse
Affiliation(s)
- Weihua Zhang
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Liming Yu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Xinxin Han
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Jie Pan
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Jiajia Deng
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Luying Zhu
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Yun Lu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Wei Huang
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Shangfeng Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Qiang Li
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| |
Collapse
|
18
|
Rosero Salazar DH, Carvajal Monroy PL, Wagener FADTG, Von den Hoff JW. Orofacial Muscles: Embryonic Development and Regeneration after Injury. J Dent Res 2019; 99:125-132. [PMID: 31675262 PMCID: PMC6977159 DOI: 10.1177/0022034519883673] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Orofacial congenital defects such as cleft lip and/or palate are associated with impaired muscle regeneration and fibrosis after surgery. Also, other orofacial reconstructions or trauma may end up in defective muscle regeneration and fibrosis. The aim of this review is to discuss current knowledge on the development and regeneration of orofacial muscles in comparison to trunk and limb muscles. The orofacial muscles include the tongue muscles and the branchiomeric muscles in the lower face. Their main functions are chewing, swallowing, and speech. All orofacial muscles originate from the mesoderm of the pharyngeal arches under the control of cranial neural crest cells. Research in vertebrate models indicates that the molecular regulation of orofacial muscle development is different from that of trunk and limb muscles. In addition, the regenerative ability of orofacial muscles is lower, and they develop more fibrosis than other skeletal muscles. Therefore, specific approaches need to be developed to stimulate orofacial muscle regeneration. Regeneration may be stimulated by growth factors such fibroblast growth factors and hepatocyte growth factor, while fibrosis may be reduced by targeting the transforming growth factor β1 (TGFβ1)/myofibroblast axis. New approaches that combine these 2 aspects will improve the surgical treatment of orofacial muscle defects.
Collapse
Affiliation(s)
- D H Rosero Salazar
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - P L Carvajal Monroy
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands.,Department of Oral and Maxillofacial Surgery, Special Dental Care and Orthodontics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - F A D T G Wagener
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - J W Von den Hoff
- Department of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| |
Collapse
|
19
|
Chen F, Zhou J, Li Y, Zhao Y, Yuan J, Cao Y, Wang L, Zhang Z, Zhang B, Wang CC, Cheung TH, Wu Z, Wong CCL, Sun H, Wang H. YY1 regulates skeletal muscle regeneration through controlling metabolic reprogramming of satellite cells. EMBO J 2019; 38:embj.201899727. [PMID: 30979776 DOI: 10.15252/embj.201899727] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 02/04/2023] Open
Abstract
Skeletal muscle satellite cells (SCs) are adult muscle stem cells responsible for muscle regeneration after acute or chronic injuries. The lineage progression of quiescent SC toward activation, proliferation, and differentiation during the regeneration is orchestrated by cascades of transcription factors (TFs). Here, we elucidate the function of TF Yin Yang1 (YY1) in muscle regeneration. Muscle-specific deletion of YY1 in embryonic muscle progenitors leads to severe deformity of diaphragm muscle formation, thus neonatal death. Inducible deletion of YY1 in SC almost completely blocks the acute damage-induced muscle repair and exacerbates the chronic injury-induced dystrophic phenotype. Examination of SC revealed that YY1 loss results in cell-autonomous defect in activation and proliferation. Mechanistic search revealed that YY1 binds and represses mitochondrial gene expression. Simultaneously, it also stabilizes Hif1α protein and activates Hif1α-mediated glycolytic genes to facilitate a metabolic reprogramming toward glycolysis which is needed for SC proliferation. Altogether, our findings have identified YY1 as a key regulator of SC metabolic reprogramming through its dual roles in modulating both mitochondrial and glycolytic pathways.
Collapse
Affiliation(s)
- Fengyuan Chen
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Jiajian Zhou
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yuying Li
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yu Zhao
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Jie Yuan
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yang Cao
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Lijun Wang
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Baoting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynecology, Li Ka Shing Institute of Health Sciences, The Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Tom H Cheung
- The State Key Lab in Molecular Neuroscience, Division of Life Science, Center for Stem Cell Research and Center for Systems Biology and Human Diseases, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhenguo Wu
- The State Key Lab in Molecular Neuroscience, Division of Life Science, Center for Stem Cell Research and Center for Systems Biology and Human Diseases, The Hong Kong University of Science and Technology, Hong Kong, China
| | | | - Hao Sun
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Huating Wang
- Department of Orthopedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
20
|
Monge C, DiStasio N, Rossi T, Sébastien M, Sakai H, Kalman B, Boudou T, Tajbakhsh S, Marty I, Bigot A, Mouly V, Picart C. Quiescence of human muscle stem cells is favored by culture on natural biopolymeric films. Stem Cell Res Ther 2017; 8:104. [PMID: 28464938 PMCID: PMC5414338 DOI: 10.1186/s13287-017-0556-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 03/01/2017] [Accepted: 04/06/2017] [Indexed: 12/18/2022] Open
Abstract
Background Satellite cells are quiescent resident muscle stem cells that present an important potential to regenerate damaged tissue. However, this potential is diminished once they are removed from their niche environment in vivo, prohibiting the long-term study and genetic investigation of these cells. This study therefore aimed to provide a novel biomaterial platform for the in-vitro culture of human satellite cells that maintains their stem-like quiescent state, an important step for cell therapeutic studies. Methods Human muscle satellite cells were isolated from two donors and cultured on soft biopolymeric films of controlled stiffness. Cell adhesive phenotype, maintenance of satellite cell quiescence and capacity for gene manipulation were investigated using FACS, western blotting, fluorescence microscopy and electron microscopy. Results About 85% of satellite cells cultured in vitro on soft biopolymer films for 3 days maintained expression of the quiescence marker Pax7, as compared with 60% on stiffer films and 50% on tissue culture plastic. The soft biopolymeric films allowed satellite cell culture for up to 6 days without renewing the media. These cells retained their stem-like properties, as evidenced by the expression of stem cell markers and reduced expression of differentiated markers. In addition, 95% of cells grown on these soft biopolymeric films were in the G0/G1 stage of the cell cycle, as opposed to those grown on plastic that became activated and began to proliferate and differentiate. Conclusions Our study identifies a new biomaterial made of a biopolymer thin film for the maintenance of the quiescence state of muscle satellite cells. These cells could be activated at any point simply by replating them onto a plastic culture dish. Furthermore, these cells could be genetically manipulated by viral transduction, showing that this biomaterial may be further used for therapeutic strategies. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0556-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claire Monge
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France. .,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France.
| | - Nicholas DiStasio
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France.,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France
| | - Thomas Rossi
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France.,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France
| | - Muriel Sébastien
- Université de Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Chemin Fortuné Ferrini, F-3800, Grenoble, France.,INSERM, U1216, F-38000, Grenoble, France
| | - Hiroshi Sakai
- Stem Cells & Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France.,CNRS, UMR 3738; Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
| | - Benoit Kalman
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France.,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France
| | - Thomas Boudou
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France.,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France
| | - Shahragim Tajbakhsh
- Stem Cells & Development, Department of Developmental & Stem Cell Biology, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France.,CNRS, UMR 3738; Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
| | - Isabelle Marty
- Université de Grenoble Alpes, Grenoble Institut des Neurosciences, GIN, Chemin Fortuné Ferrini, F-3800, Grenoble, France.,INSERM, U1216, F-38000, Grenoble, France
| | - Anne Bigot
- Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, 47 Boulevard de l'hôpital, 75013, Paris, France
| | - Vincent Mouly
- Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology, 47 Boulevard de l'hôpital, 75013, Paris, France
| | - Catherine Picart
- CNRS, UMR 5628, LMGP, 3 parvis Louis Néel, F-38016, Grenoble, France. .,Université de Grenoble Alpes, Grenoble Institute of Technology, 3 parvis Louis Néel, F-38016, Grenoble, France.
| |
Collapse
|
21
|
Leon AS. Attenuation of Adverse Effects of Aging on Skeletal Muscle by Regular Exercise and Nutritional Support. Am J Lifestyle Med 2017; 11:4-16. [PMID: 30202306 PMCID: PMC6124840 DOI: 10.1177/1559827615589319] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 05/06/2015] [Indexed: 12/19/2022] Open
Abstract
Beginning early in midlife, natural/primary aging is inevitably associated with a progressive reduction in muscle mass and function. This process can progress with aging to a substantial loss of strength, particularly in the lower extremities, reducing mobility. This condition, commonly referred to as sarcopenia, can result in frailty, reducing one's ability to live independently. This article reviews the underlying biological process contributing to the development of sarcopenia and the roles of regular exercise and nutritional support for attenuating aging-associated muscle loss as well as risk and management of sarcopenia and associated frailty.
Collapse
|
22
|
Beyer S, Pontis J, Schirwis E, Battisti V, Rudolf A, Le Grand F, Ait-Si-Ali S. Canonical Wnt signalling regulates nuclear export of Setdb1 during skeletal muscle terminal differentiation. Cell Discov 2016; 2:16037. [PMID: 27790377 PMCID: PMC5067623 DOI: 10.1038/celldisc.2016.37] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 09/19/2016] [Indexed: 02/06/2023] Open
Abstract
The histone 3 lysine 9 methyltransferase Setdb1 is essential for both stem cell pluripotency and terminal differentiation of different cell types. To shed light on the roles of Setdb1 in these mutually exclusive processes, we used mouse skeletal myoblasts as a model of terminal differentiation. Ex vivo studies on isolated single myofibres showed that Setdb1 is required for adult muscle stem cells expansion following activation. In vitro studies in skeletal myoblasts confirmed that Setdb1 suppresses terminal differentiation. Genomic binding analyses showed a release of Setdb1 from selected target genes upon myoblast terminal differentiation, concomitant to a nuclear export of Setdb1 to the cytoplasm. Both genomic release and cytoplasmic Setdb1 relocalisation during differentiation were dependent on canonical Wnt signalling. Transcriptomic assays in myoblasts unravelled a significant overlap between Setdb1 and Wnt3a regulated genetic programmes. Together, our findings revealed Wnt-dependent subcellular relocalisation of Setdb1 as a novel mechanism regulating Setdb1 functions and myogenesis.
Collapse
Affiliation(s)
- Sophie Beyer
- Centre National de la Recherche Scientifique CNRS-Université Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate UMR7216 , Paris, France
| | - Julien Pontis
- Centre National de la Recherche Scientifique CNRS-Université Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate UMR7216 , Paris, France
| | - Elija Schirwis
- Institut Cochin, Université Paris-Descartes, Centre National de la Recherche Scientifique (CNRS) UMR8104, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Valentine Battisti
- Centre National de la Recherche Scientifique CNRS-Université Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate UMR7216 , Paris, France
| | - Anja Rudolf
- Institut Cochin, Université Paris-Descartes, Centre National de la Recherche Scientifique (CNRS) UMR8104, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Fabien Le Grand
- Institut Cochin, Université Paris-Descartes, Centre National de la Recherche Scientifique (CNRS) UMR8104, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Paris, France
| | - Slimane Ait-Si-Ali
- Centre National de la Recherche Scientifique CNRS-Université Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate UMR7216 , Paris, France
| |
Collapse
|
23
|
Naldaiz-Gastesi N, Goicoechea M, Alonso-Martín S, Aiastui A, López-Mayorga M, García-Belda P, Lacalle J, San José C, Araúzo-Bravo MJ, Trouilh L, Anton-Leberre V, Herrero D, Matheu A, Bernad A, García-Verdugo JM, Carvajal JJ, Relaix F, Lopez de Munain A, García-Parra P, Izeta A. Identification and Characterization of the Dermal Panniculus Carnosus Muscle Stem Cells. Stem Cell Reports 2016; 7:411-424. [PMID: 27594590 PMCID: PMC5032673 DOI: 10.1016/j.stemcr.2016.08.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 01/05/2023] Open
Abstract
The dermal Panniculus carnosus (PC) muscle is important for wound contraction in lower mammals and represents an interesting model of muscle regeneration due to its high cell turnover. The resident satellite cells (the bona fide muscle stem cells) remain poorly characterized. Here we analyzed PC satellite cells with regard to developmental origin and purported function. Lineage tracing shows that they originate in Myf5+, Pax3/Pax7+ cell populations. Skin and muscle wounding increased PC myofiber turnover, with the satellite cell progeny being involved in muscle regeneration but with no detectable contribution to the wound-bed myofibroblasts. Since hematopoietic stem cells fuse to PC myofibers in the absence of injury, we also studied the contribution of bone marrow-derived cells to the PC satellite cell compartment, demonstrating that cells of donor origin are capable of repopulating the PC muscle stem cell niche after irradiation and bone marrow transplantation but may not fully acquire the relevant myogenic commitment. PC satellite cells originate from Myf5+, Pax3/Pax7+ cell lineages Skin and muscle wounding increase PC myofiber turnover Donor bone marrow cells repopulate the PC satellite niche after BMT Dermis-derived myogenesis originates from the PC satellite cell population
Collapse
Affiliation(s)
- Neia Naldaiz-Gastesi
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - María Goicoechea
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Sonia Alonso-Martín
- INSERM U955-E10, Université Paris Est, Faculté de Médicine, IMRB U955-E10, Creteil 94000, France
| | - Ana Aiastui
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Macarena López-Mayorga
- Molecular Embryology Team, Centro Andaluz de Biología del Desarrollo, Sevilla 41013, Spain
| | - Paula García-Belda
- CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, Spain
| | - Jaione Lacalle
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; Faculty of Medicine and Nursing, UPV-EHU, San Sebastián 20014, Spain
| | - Carlos San José
- Animal Facility and Experimental Surgery, Instituto Biodonostia, San Sebastián 20014, Spain
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Instituto Biodonostia, San Sebastián 20014, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
| | - Lidwine Trouilh
- INSA, UPS, INP, LISBP, Université de Toulouse, 31077 Toulouse, France; INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Véronique Anton-Leberre
- INSA, UPS, INP, LISBP, Université de Toulouse, 31077 Toulouse, France; INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Diego Herrero
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid 28049, Spain
| | - Ander Matheu
- IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain; Cellular Oncology Group, Oncology Area, Instituto Biodonostia, San Sebastián 20014, Spain
| | - Antonio Bernad
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid 28049, Spain
| | - José Manuel García-Verdugo
- CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, Spain
| | - Jaime J Carvajal
- Molecular Embryology Team, Centro Andaluz de Biología del Desarrollo, Sevilla 41013, Spain
| | - Frédéric Relaix
- INSERM U955-E10, Université Paris Est, Faculté de Médicine, IMRB U955-E10, Creteil 94000, France
| | - Adolfo Lopez de Munain
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Faculty of Medicine and Nursing, Department of Neurosciences, UPV-EHU, San Sebastián 20014, Spain; Department of Neurology, Hospital Universitario Donostia, San Sebastián 20014, Spain
| | - Patricia García-Parra
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Ander Izeta
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra, San Sebastián 20009, Spain.
| |
Collapse
|
24
|
Abstract
Satellite cells comprise a pool of quiescent stem cells that repair muscle damage, but the mechanisms enforcing their quiescence are poorly defined. In this issue of Cell Stem Cell, Boonsanay et al. (2016) show that the histone methyltransferase Suv4-20H1 maintains satellite cell quiescence by promoting a heterochromatic state through transcriptional repression of the myogenic master regulator MyoD.
Collapse
Affiliation(s)
- Yuefeng Li
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - F Jeffrey Dilworth
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
25
|
Role of integrin α7β1 signaling in myoblast differentiation on aligned polydioxanone scaffolds. Acta Biomater 2016; 39:44-54. [PMID: 27142254 DOI: 10.1016/j.actbio.2016.04.046] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/25/2016] [Accepted: 04/29/2016] [Indexed: 12/30/2022]
Abstract
UNLABELLED The aligned structural environment in skeletal muscle is believed to be a crucial component in functional muscle regeneration. Myotube formation is increased on aligned biomaterials, but we do not fully understand the mechanisms that direct this enhanced fusion. Previous studies indicate that the α7 integrin subunit is upregulated during myoblast differentiation, suggesting that signaling via α7β1 mediates the effect of alignment. To test this hypothesis, we took advantage of an in vitro model using random and aligned polydioxanone (PDO) matrices and C2C12 myoblasts. We measured expression and production of myoblast markers: paired box-7 (Pax7), myogenic differentiation factor-1 (MyoD), myogenin (MyoG), myogenic factor-6 (Myf6), and myosin heavy chain (MyHC). To examine the role of α7β1 signaling, we measured expression and production of α7, α5, and β1 and myoblast markers in wild type cells and in cells silenced for α7 and assessed effects of silencing on myogenic differentiation. Downstream signaling via ERK1/2 mitogen activated protein kinase (MAPK) was examined using a specific MEK1/2 inhibitor. Alignment increased mRNAs and protein for early (MyoD) and late (MyoG, MyHC) myoblast markers in comparison to non-aligned matrices, and these levels corresponded with increased α7 protein. α7-silencing reduced MyoG and MyHC protein in cells cultured on tissue culture polystyrene and aligned PDO matrices compared to wild type cells. Inhibition of ERK1/2 blocked effects of alignment. These data suggest that alignment regulates myogenic differentiation via α7β1 integrin signaling and ERK1/2 mediated gene expression. STATEMENT OF SIGNIFICANCE Muscle regeneration in severe muscle injuries is complex, requiring a sequence of events to promote healing and not fibrosis. Aligned biomaterials that recapitulate muscle environments hold potential to facilitate regeneration, but it is important to understand cell-substrate signaling to form functional muscle. A critical component of muscle signaling is integrin α7β1, where mice lacking α7 exhibit a dystrophic phenotype and impaired regeneration. Here, we report the role of α7β1 signaling in myoblast differentiation on aligned biomaterials. α7-silenced myoblasts were found to regulate myogenic differentiation and demonstrate defective fusion. Our data shows reduced levels of myogenin and myosin heavy chain protein, while MyoD remains unchanged. These results support the hypothesis that α7β1 signaling plays a role in substrate-dependent tissue engineering strategies.
Collapse
|
26
|
Faralli H, Wang C, Nakka K, Benyoucef A, Sebastian S, Zhuang L, Chu A, Palii CG, Liu C, Camellato B, Brand M, Ge K, Dilworth FJ. UTX demethylase activity is required for satellite cell-mediated muscle regeneration. J Clin Invest 2016; 126:1555-65. [PMID: 26999603 DOI: 10.1172/jci83239] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 01/07/2016] [Indexed: 12/31/2022] Open
Abstract
The X chromosome-encoded histone demethylase UTX (also known as KDM6A) mediates removal of repressive trimethylation of histone H3 lysine 27 (H3K27me3) to establish transcriptionally permissive chromatin. Loss of UTX in female mice is embryonic lethal. Unexpectedly, male UTX-null mice escape embryonic lethality due to expression of UTY, a paralog that lacks H3K27 demethylase activity, suggesting an enzyme-independent role for UTX in development and thereby challenging the need for active H3K27 demethylation in vivo. However, the requirement for active H3K27 demethylation in stem cell-mediated tissue regeneration remains untested. Here, we employed an inducible mouse KO that specifically ablates Utx in satellite cells (SCs) and demonstrated that active H3K27 demethylation is necessary for muscle regeneration. Loss of UTX in SCs blocked myofiber regeneration in both male and female mice. Furthermore, we demonstrated that UTX mediates muscle regeneration through its H3K27 demethylase activity, as loss of demethylase activity either by chemical inhibition or knock-in of demethylase-dead UTX resulted in defective muscle repair. Mechanistically, dissection of the muscle regenerative process revealed that the demethylase activity of UTX is required for expression of the transcription factor myogenin, which in turn drives differentiation of muscle progenitors. Thus, we have identified a critical role for the enzymatic activity of UTX in activating muscle-specific gene expression during myofiber regeneration and have revealed a physiological role for active H3K27 demethylation in vivo.
Collapse
|
27
|
Ding K, Yang Z, Xu JZ, Liu WY, Zeng Q, Hou F, Lin S. Elastic hydrogel substrate supports robust expansion of murine myoblasts and enhances their engraftment. Exp Cell Res 2015. [PMID: 26210646 DOI: 10.1016/j.yexcr.2015.07.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The application of satellite cell-derived myoblasts in regenerative medicine has been restricted by the rapid loss of stemness during in vitro cell expansion using traditional culture systems. However, studies published in the past decade have highlighted the influence of substrate elasticity on stem cell fate and revealed that culture on a soft hydrogel substrate can promote self-renewal and prolong the regenerative potential of muscle stem cells. Whether hydrogel substrates have similar effects after long-term robust expansion remains to be determined. Herein we prepared an elastic chitosan/beta-glycerophosphate/collagen hydrogel mimicking the soft microenvironment of muscle tissues for use as the substrate for satellite cell culture and investigated its influence on long-term cell expansion. After 20 passages in culture, satellite cell-derived myoblasts cultured on our hydrogel substrate exhibited significant improvements in proliferation capability, cell viability, colony forming frequency, and potential for myogenic differentiation compared to those cultured on a routine rigid culture surface. Immunochemical staining and western blot analysis both confirmed that myoblasts cultured on the hydrogel substrate expressed higher levels of several differentiation-related markers, including Pax7, Pax3, and SSEA-1, and a lower level of MyoD compared to myoblasts cultured on rigid culture plates (all p<0.05). After transplantation into the tibialis anterior of nude mice, myoblasts that had been cultured on the hydrogel substrate demonstrated a significantly greater engraftment efficacy than those cultured on the traditional surface. Collectively, these results indicate that the elastic hydrogel substrate supported robust expansion of murine myoblasts and enhanced their engraftment in vivo.
Collapse
Affiliation(s)
- Ke Ding
- Department of Pediatric Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China.
| | - Zhong Yang
- Department of Clinical Hematology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jian-Zhong Xu
- Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| | - Wen-Ying Liu
- Department of Pediatric Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Qiang Zeng
- Department of Pediatric Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Fang Hou
- Department of Pediatric Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Sen Lin
- Department of Anatomy and Histology & Embryology, Chengdu Medical College, Chengdu 610500, China
| |
Collapse
|
28
|
Jank BJ, Xiong L, Moser PT, Guyette JP, Ren X, Cetrulo CL, Leonard DA, Fernandez L, Fagan SP, Ott HC. Engineered composite tissue as a bioartificial limb graft. Biomaterials 2015; 61:246-56. [PMID: 26004237 DOI: 10.1016/j.biomaterials.2015.04.051] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/22/2015] [Accepted: 04/30/2015] [Indexed: 12/18/2022]
Abstract
The loss of an extremity is a disastrous injury with tremendous impact on a patient's life. Current mechanical prostheses are technically highly sophisticated, but only partially replace physiologic function and aesthetic appearance. As a biologic alternative, approximately 70 patients have undergone allogeneic hand transplantation to date worldwide. While outcomes are favorable, risks and side effects of transplantation and long-term immunosuppression pose a significant ethical dilemma. An autologous, bio-artificial graft based on native extracellular matrix and patient derived cells could be produced on demand and would not require immunosuppression after transplantation. To create such a graft, we decellularized rat and primate forearms by detergent perfusion and yielded acellular scaffolds with preserved composite architecture. We then repopulated muscle and vasculature with cells of appropriate phenotypes, and matured the composite tissue in a perfusion bioreactor under electrical stimulation in vitro. After confirmation of composite tissue formation, we transplanted the resulting bio-composite grafts to confirm perfusion in vivo.
Collapse
Affiliation(s)
- Bernhard J Jank
- Center for Regenerative Medicine, Massachusetts General Hospital, USA; Harvard Medical School, Boston, MA, USA
| | - Linjie Xiong
- Center for Regenerative Medicine, Massachusetts General Hospital, USA
| | - Philipp T Moser
- Center for Regenerative Medicine, Massachusetts General Hospital, USA; Harvard Medical School, Boston, MA, USA
| | - Jacques P Guyette
- Center for Regenerative Medicine, Massachusetts General Hospital, USA; Harvard Medical School, Boston, MA, USA
| | - Xi Ren
- Center for Regenerative Medicine, Massachusetts General Hospital, USA; Harvard Medical School, Boston, MA, USA
| | - Curtis L Cetrulo
- Harvard Medical School, Boston, MA, USA; Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, USA
| | - David A Leonard
- Harvard Medical School, Boston, MA, USA; Transplantation Biology Research Center, Department of Surgery, Massachusetts General Hospital, USA
| | | | - Shawn P Fagan
- Massachusetts General Hospital, Division of Burn Surgery, Harvard Medical School, USA
| | - Harald C Ott
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Harada A, Mallappa C, Okada S, Butler JT, Baker SP, Lawrence JB, Ohkawa Y, Imbalzano AN. Spatial re-organization of myogenic regulatory sequences temporally controls gene expression. Nucleic Acids Res 2015; 43:2008-21. [PMID: 25653159 PMCID: PMC4344497 DOI: 10.1093/nar/gkv046] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 11/05/2014] [Accepted: 01/13/2015] [Indexed: 12/14/2022] Open
Abstract
During skeletal muscle differentiation, the activation of some tissue-specific genes occurs immediately while others are delayed. The molecular basis controlling temporal gene regulation is poorly understood. We show that the regulatory sequences, but not other regions of genes expressed at late times of myogenesis, are in close physical proximity in differentiating embryonic tissue and in differentiating culture cells, despite these genes being located on different chromosomes. Formation of these inter-chromosomal interactions requires the lineage-determinant MyoD and functional Brg1, the ATPase subunit of SWI/SNF chromatin remodeling enzymes. Ectopic expression of myogenin and a specific Mef2 isoform induced myogenic differentiation without activating endogenous MyoD expression. Under these conditions, the regulatory sequences of late gene loci were not in close proximity, and these genes were prematurely activated. The data indicate that the spatial organization of late genes contributes to temporal regulation of myogenic transcription by restricting late gene expression during the early stages of myogenesis.
Collapse
Affiliation(s)
- Akihito Harada
- Department of Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | - Chandrashekara Mallappa
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Seiji Okada
- Department of Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | - John T Butler
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Stephen P Baker
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jeanne B Lawrence
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Yasuyuki Ohkawa
- Department of Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Anthony N Imbalzano
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
30
|
Sohi G, Dilworth FJ. Noncoding RNAs as epigenetic mediators of skeletal muscle regeneration. FEBS J 2015; 282:1630-46. [PMID: 25483175 DOI: 10.1111/febs.13170] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 12/16/2022]
Abstract
Skeletal muscle regeneration is a well-characterized biological process in which resident adult stem cells must undertake a series of cell-fate decisions to ensure efficient repair of the damaged muscle fibers while also maintaining the stem cell niche. Satellite cells, the main stem cell contributing to the repaired muscle fiber, are maintained in a quiescent state in healthy muscle. Upon injury, the satellite cells become activated, and proliferate to expand the muscle progenitor cell population before returning to the quiescent state or differentiating to become myofibers. Importantly, the determination of cell fate is controlled at the epigenetic level in response to environmental cues. In this review, we discuss our current understanding of the role played by noncoding RNAs (both miRNAs and long-noncoding RNAs) in the epigenetic control of muscle regeneration.
Collapse
Affiliation(s)
- Gurjeev Sohi
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Canada
| | | |
Collapse
|
31
|
Fu X, Wang H, Hu P. Stem cell activation in skeletal muscle regeneration. Cell Mol Life Sci 2015; 72:1663-77. [PMID: 25572293 PMCID: PMC4412728 DOI: 10.1007/s00018-014-1819-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/21/2014] [Accepted: 12/22/2014] [Indexed: 12/31/2022]
Abstract
Muscle stem cell (satellite cell) activation post muscle injury is a transient and critical step in muscle regeneration. It is regulated by physiological cues, signaling molecules, and epigenetic regulatory factors. The mechanisms that coherently turn on the complex activation process shortly after trauma are just beginning to be illuminated. In this review, we will discuss the current knowledge of satellite cell activation regulation.
Collapse
Affiliation(s)
- Xin Fu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | | | | |
Collapse
|
32
|
Genetic Dissection of the Physiological Role of Skeletal Muscle in Metabolic Syndrome. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/635146] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The primary deficiency underlying metabolic syndrome is insulin resistance, in which insulin-responsive peripheral tissues fail to maintain glucose homeostasis. Because skeletal muscle is the major site for insulin-induced glucose uptake, impairments in skeletal muscle’s insulin responsiveness play a major role in the development of insulin resistance and type 2 diabetes. For example, skeletal muscle of type 2 diabetes patients and their offspring exhibit reduced ratios of slow oxidative muscle. These observations suggest the possibility of applying muscle remodeling to recover insulin sensitivity in metabolic syndrome. Skeletal muscle is highly adaptive to external stimulations such as exercise; however, in practice it is often not practical or possible to enforce the necessary intensity to obtain measurable benefits to the metabolic syndrome patient population. Therefore, identifying molecular targets for inducing muscle remodeling would provide new approaches to treat metabolic syndrome. In this review, the physiological properties of skeletal muscle, genetic analysis of metabolic syndrome in human populations and model organisms, and genetically engineered mouse models will be discussed in regard to the prospect of applying skeletal muscle remodeling as possible therapy for metabolic syndrome.
Collapse
|
33
|
Kozakowska M, Szade K, Dulak J, Jozkowicz A. Role of heme oxygenase-1 in postnatal differentiation of stem cells: a possible cross-talk with microRNAs. Antioxid Redox Signal 2014; 20:1827-50. [PMID: 24053682 PMCID: PMC3961774 DOI: 10.1089/ars.2013.5341] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. HO-1 via heme removal and degradation products acts as a cytoprotective, anti-inflammatory, immunomodulatory, and proangiogenic protein, regulating also a cell cycle. Additionally, HO-1 can translocate to nucleus and regulate transcription factors, so it can also act independently of enzymatic function. RECENT ADVANCES Recently, a body of evidence has emerged indicating a role for HO-1 in postnatal differentiation of stem and progenitor cells. Maturation of satellite cells, skeletal myoblasts, adipocytes, and osteoclasts is inhibited by HO-1, whereas neurogenic differentiation and formation of cardiomyocytes perhaps can be enhanced. Moreover, HO-1 influences a lineage commitment in pluripotent stem cells and maturation of hematopoietic cells. It may play a role in development of osteoblasts, but descriptions of its exact effects are inconsistent. CRITICAL ISSUES In this review we discuss a role of HO-1 in cell differentiation, and possible HO-1-dependent signal transduction pathways. Among the potential mediators, we focused on microRNA (miRNA). These small, noncoding RNAs are critical for cell differentiation. Recently we have found that HO-1 not only influences expression of specific miRNAs but also regulates miRNA processing enzymes. FUTURE DIRECTIONS It seems that interplay between HO-1 and miRNAs may be important in regulating fates of stem and progenitor cells and needs further intensive studies.
Collapse
Affiliation(s)
- Magdalena Kozakowska
- 1 Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University , Krakow, Poland
| | | | | | | |
Collapse
|
34
|
Abstract
Skeletal muscle satellite cells (myoblasts) are the primary stem cells of skeletal muscle which contribute to growth, maintenance, and repair of the muscles. Satellite cells are the first stem cells used for cellular cardiomyoplasty more than 20 years ago. The isolation, culture, labeling, and identification of satellite cells are described in detail here. The implantation and outcomes of cellular cardiomyoplasty using satellite cells have been summarized in the previous chapter (Chapter 1).
Collapse
|
35
|
Wang M, Xie H, Shrestha S, Sredni S, Morgan GA, Pachman LM. Methylation alterations of WT1 and homeobox genes in inflamed muscle biopsy samples from patients with untreated juvenile dermatomyositis suggest self-renewal capacity. ACTA ACUST UNITED AC 2013; 64:3478-85. [PMID: 22674142 DOI: 10.1002/art.34573] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To determine the effect of methylation alteration in inflamed muscles from children with juvenile dermatomyositis (DM) and other idiopathic inflammatory myopathies (IIMs). METHODS Magnetic resonance imaging-directed diagnostic muscle biopsies yielded samples from 20 children with juvenile DM, which were used for genome-wide DNA methylation profiling, as were muscle biopsy samples from 4 healthy controls. Bisulfite treatment followed by pyrosequencing confirmed methylation status in juvenile DM and other IIMs. Immunohistochemistry defined localization and expression levels of WT1. RESULTS Comparison of genome-wide DNA methylation profiling between juvenile DM muscle and normal control muscle revealed 27 genes with a significant methylation difference between the groups. These genes were enriched with transcription factors and/or cell cycle regulators and were unrelated to duration of untreated disease. Six homeobox genes were among them; ALX4, HOXC11, HOXD3, and HOXD4 were hypomethylated, while EMX2 and HOXB1 were hypermethylated. WT1 was significantly hypomethylated in juvenile DM (Δβ = -0.41, P < 0.001). Bisulfite pyrosequencing verification in samples from 56 patients with juvenile DM confirmed the methylation alterations of these genes. Similar methylation alterations were observed in juvenile polymyositis (n = 5) and other IIMs (n = 9). Concordant with the other findings, WT1 protein was increased in juvenile DM muscle, with average positive staining of 11.6%, but was undetectable in normal muscle (P < 0.001). CONCLUSION These results suggest that affected muscles of children with juvenile DM and IIMs have the capacity to be repaired, and that homeobox and WT1 genes are epigenetically marked to facilitate this repair process, potentially suggesting new avenues of therapeutic intervention.
Collapse
Affiliation(s)
- Min Wang
- Children's Hospital of Chicago Research Center and Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Cellular cardiomyoplasty is a cell therapy using stem cells or progenitor cells for myocardial regeneration to improve cardiac function and mitigate heart failure. Since we first published cellular cardiomyoplasty in 1989, this procedure became the innovative method to treat damaged myocardium other than heart transplantation. A significant improvement in cardiac function, metabolism, and perfusion is generally observed in experimental and clinical studies, but the improvement is mild and incomplete. Although safety, feasibility, and efficacy have been well documented for the procedure, the beneficial mechanisms remain unclear and optimization of the procedure requires further study. This chapter briefly reviews the stem cells used for cellular cardiomyoplasty and their clinical outcomes with possible improvements in future studies.
Collapse
Affiliation(s)
- Elizabeth K Lamb
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | | | | |
Collapse
|
37
|
Liu QC, Zha XH, Faralli H, Yin H, Louis-Jeune C, Perdiguero E, Pranckeviciene E, Muñoz-Cànoves P, Rudnicki MA, Brand M, Perez-Iratxeta C, Dilworth FJ. Comparative expression profiling identifies differential roles for Myogenin and p38α MAPK signaling in myogenesis. J Mol Cell Biol 2012; 4:386-97. [PMID: 22847234 DOI: 10.1093/jmcb/mjs045] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle differentiation is mediated by a complex gene expression program requiring both the muscle-specific transcription factor Myogenin (Myog) and p38α MAPK (p38α) signaling. However, the relative contribution of Myog and p38α to the formation of mature myotubes remains unknown. Here, we have uncoupled the activity of Myog from that of p38α to gain insight into the individual roles of these proteins in myogenesis. Comparative expression profiling confirmed that Myog activates the expression of genes involved in muscle function. Furthermore, we found that in the absence of p38α signaling, Myog expression leads to the down-regulation of genes involved in cell cycle progression. Consistent with this, the expression of Myog is sufficient to induce cell cycle exit. Interestingly, p38α-defective, Myog-expressing myoblasts fail to form multinucleated myotubes, suggesting an important role for p38α in cell fusion. Through the analysis of p38α up-regulated genes, the tetraspanin CD53 was identified as a candidate fusion protein, a role confirmed both ex vivo in primary myoblasts, and in vivo during myofiber regeneration in mice. Thus, our study has revealed an unexpected role for Myog in mediating cell cycle exit and has identified an essential role for p38α in cell fusion through the up-regulation of CD53.
Collapse
Affiliation(s)
- Qi-Cai Liu
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada K1H 8L6
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fiorotto M. The making of a muscle. THE BIOCHEMIST 2012; 34:4-11. [PMID: 28190934 PMCID: PMC5298863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The skeletal musculature is usually thought of as the primary organ of locomotion, and, like the tyres of a high-performance racing car, their composition, design, preparation and plasticity can make the difference between winner and 'wannabe'. The similarities do not end there, however. Their primary components (cells of the mesodermal layer in the embryo and latex from the rubber tree) begin their existence in locations that can be quite distant from their final point of use and in forms that bear no resemblance to the final product. Their differentiation from primary material to final product entails extensive processing, and the integration of other materials and structures are essential to ensure their function. A fundamental difference, however, is that, in the case of muscle, once the embryo is formed, the progression from relatively undifferentiated mesodermal cells to the final structures is on autopilot, provided there are no contextual aberrations either from genetic or environmental causes. Our current understanding of how muscles develop is a synthesis of observations made on a wide array of organisms, including nematode worms, fruitflies, fish, frogs, birds and various mammals, as well as from the in vitro study of cells isolated from these species. The study of myogenesis in mammals, although less amenable to experimental manipulation, has been facilitated by the recent advances in mouse genetic engineering which has enabled the function of individual genes and cell types to be investigated, as well as the lineage of cells to be traced back to their origin. In this rapid trek through the life of a muscle, how the production of a mature functional muscle from its early inception is orchestrated will be outlined in exceedingly broad strokes so as to convey the wide range of processes that must be engaged in order to generate a functional muscle. Hopefully, enough information will be provided to encourage those interested to explore further.
Collapse
Affiliation(s)
- Marta Fiorotto
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine
| |
Collapse
|
39
|
|