1
|
Wang X, Xing T, Zhu X, Wang X, Zhao L, Gao F, Zhang L. Dysfunctional terminal differentiation of satellite cells in the wooden breast muscle of Ross 308 broiler chickens. Poult Sci 2025; 104:105041. [PMID: 40121757 PMCID: PMC11979995 DOI: 10.1016/j.psj.2025.105041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025] Open
Abstract
Muscle satellite cells (MuSCs), defined as skeletal muscle-resident stem cells, serve as the principal cellular mediators of the repair and regeneration processes in impaired skeletal muscle tissue. However, myofibers with degeneration and necrosis in wooden breast (WB) myopathy are still not completely repaired, and this study aims to elucidate the functional changes of MuSCs between normal and WB-affected pectoralis major (PM) muscle of broilers. A total of 100 Ross 308 broilers (42 days old) fed the same diet were randomly selected. Initial identification of normal and WB phenotypes was conducted through behavioral assessments and manual palpation of the PM muscle. Ultimately, PM samples were harvested from 8 chickens per experimental group, classified into normal (NOR), moderate (MOD), and severe (SEV) groups based on necropsy grading. The superficial cranial region of each PM sample was isolated for quantitative analysis of MuSCs functional markers. One-way ANOVA was used to compare differences among groups, with p < 0.05 indicating significant differences. Results showed that breast muscle weight and morphological indices (length, top/middle/bottom heights) of MOD and SEV groups were higher than those of NOR group (P < 0.05). The SEV group exhibited larger myofiber diameter and cross-sectional area but lower fiber density compared to NOR group (P < 0.05). There was higher mitotic activity of MuSCs, populations of MuSCs and myoblasts in severe WB muscle, characterized by elevated BrdU+, Pax7+, and MyoD+ cells (P < 0.05). The mRNA or protein expressions of Myf5, MyoD, MyoG, Myomaker, Myomerger, MSTN, HGF, IGF-1 and IGF-1R were upregulated in SEV group, along with downregulation of MRF4 protein (P < 0.05). Taken together, the proliferation of MuSCs is promoted in WB muscle, while terminal differentiation and fusion were failure, inhibiting the formation of multinucleated myotubes and mature myofibers. These findings elucidate novel mechanistic insights into the pathogenesis of WB myopathy and potential strategies for stem cell-mediated WB prophylaxis in poultry industry.
Collapse
Affiliation(s)
- Xin Wang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Tong Xing
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Xudong Zhu
- College of Science, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Xiaofei Wang
- College of Science, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Liang Zhao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Feng Gao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China
| | - Lin Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Provincial Key Laboratory of Gastrointestinal Nutrition and Animal Health, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
2
|
Yue F, Gu L, Qiu J, Oprescu SN, Beckett LM, Ellis JM, Donkin SS, Kuang S. Mitochondrial fatty acid oxidation regulates adult muscle stem cell function through modulating metabolic flux and protein acetylation. EMBO J 2025; 44:2566-2595. [PMID: 40065099 PMCID: PMC12048568 DOI: 10.1038/s44318-025-00397-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/31/2025] [Accepted: 02/10/2025] [Indexed: 05/04/2025] Open
Abstract
During homeostasis and regeneration, satellite cells, the resident stem cells of skeletal muscle, have distinct metabolic requirements for fate transitions between quiescence, proliferation and differentiation. However, the contribution of distinct energy sources to satellite cell metabolism and function remains largely unexplored. Here, we uncover a role of mitochondrial fatty acid oxidation (FAO) in satellite cell integrity and function. Single-cell RNA sequencing revealed progressive enrichment of mitochondrial FAO and downstream pathways during activation, proliferation and myogenic commitment of satellite cells. Deletion of Carnitine palmitoyltransferase 2 (Cpt2), the rate-limiting enzyme in FAO, hampered muscle stem cell expansion and differentiation upon acute muscle injury, markedly delaying regeneration. Cpt2 deficiency reduces acetyl-CoA levels in satellite cells, impeding the metabolic flux and acetylation of selective proteins including Pax7, the central transcriptional regulator of satellite cells. Notably, acetate supplementation restored cellular metabolic flux and partially rescued the regenerative defects of Cpt2-null satellite cells. These findings highlight an essential role of fatty acid oxidation in controlling satellite cell function and suggest an integration of lipid metabolism and protein acetylation in adult stem cells.
Collapse
Affiliation(s)
- Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Animal Sciences, University of Florida, Gainesville, FL, 32611, USA.
| | - Lijie Gu
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Stephanie N Oprescu
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Linda M Beckett
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Jessica M Ellis
- East Carolina Diabetes and Obesity Institute and Department of Physiology, East Carolina University, Greenville, NC, 27858, USA
| | - Shawn S Donkin
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
3
|
Wang C, Wu W, Chen J, Wang H, Zhao P. LncRNA SNHG1 regulates muscle stem cells fate through Wnt/β-catenin pathway. Dev Dyn 2025. [PMID: 40116401 DOI: 10.1002/dvdy.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/22/2025] [Accepted: 03/01/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Skeletal muscle stem cells (MuSCs) played an important role in maintaining the proper function of muscle tissues. In adults, they normally remained in a quiescent state and activated upon stimulation to undergo self-renewal or myogenic differentiation. This process was complexly regulated by cytokines, and the molecular mechanisms that promoted MuSCs activation remained largely unknown. RESULTS Here, we analyzed transcriptome data from MuSCs activated by different stimuli using weighted gene co-expression network analysis (WGCNA) and identified the key long non-coding RNA SNHG1 (lncSNHG1), which promotes the transition from the quiescent to the activated state of MuSCs. Overexpression of lncSNHG1 was able to promote the proliferation and differentiation of MuSCs, whereas knockdown resulted in the opposite results. Mechanistically, the disruption of the Wnt/β-catenin pathway blocked the quiescence exit induced by lncSNHG1. CONCLUSIONS We conclude that lncSNHG1 is a key factor that promotes the transition from the quiescent to the activated state of MuSCs and promotes cell proliferation and differentiation through the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Changying Wang
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, Taian, People's Republic of China
| | - Wenwen Wu
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, Taian, People's Republic of China
| | - Junyi Chen
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, Taian, People's Republic of China
| | - Heng Wang
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, Taian, People's Republic of China
| | - Pengxiang Zhao
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, Taian, People's Republic of China
| |
Collapse
|
4
|
Yoshigai E, Hara T, Hashimoto M, Tsuzuki H, Abe T, Inoue K, Noguchi A, Ohashi T, Fukada T. ZIP13 marks muscle satellite cells and contributes to their quiescent and active phase balance. Sci Rep 2025; 15:9206. [PMID: 40097560 PMCID: PMC11914201 DOI: 10.1038/s41598-025-92501-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Loss of ZIP13 causes Ehlers-Danlos syndrome spondylodysplastic type 3 involving connective tissue dysplasias associated with a reduction in muscular strength. However, ZIP13 role in skeletal muscle homeostasis, particularly for the regulation of muscle satellite cells (MuSCs), remains poorly understood. In this study, we investigated Zip13-knockout (KO) mice and found a reduction in MuSCs of Zip13-KO mice, in which the quiescent and activated phase balances were disrupted. To clarify the physiological role and dynamics of ZIP13 expression in MuSCs, we generated Zip13-GFP knock-in (KI) mice encoding GFP at the Zip13 locus, which showed that ZIP13 contributes to the phase balance regulation of quiescent and activated MuSCs and their functions. Indeed, Zip13-KO mice exhibited delayed recovery from skeletal muscle injury, indicating ZIP13 requirement for proper skeletal muscle regeneration. Moreover, GFP expression was reduced in the MuSCs of homozygous Zip13-GFP KI mice whose intact ZIP13 expression was perturbed, suggesting that positive feedback mechanisms exist to maintain ZIP13 expression. Altogether, our results illustrate that ZIP13 might be positively involved in skeletal muscle regeneration by controlling the quiescent/activated phase balance of MuSCs through autoregulatory ZIP13 expression, and that newly generated Zip13-GFP KI mice would be useful for investigating the roles and dynamics of ZIP13-expressing cells.
Collapse
Affiliation(s)
- Emi Yoshigai
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihama-Boji, Yamashiro, Tokushima, 770-8514, Japan.
- JSPS Research Fellowship for Young Scientists, Tokyo, Japan.
| | - Takafumi Hara
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihama-Boji, Yamashiro, Tokushima, 770-8514, Japan
| | - Masaki Hashimoto
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihama-Boji, Yamashiro, Tokushima, 770-8514, Japan
| | - Hidenao Tsuzuki
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihama-Boji, Yamashiro, Tokushima, 770-8514, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 6500047, Japan
| | - Kenichi Inoue
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 6500047, Japan
| | - Ayaka Noguchi
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihama-Boji, Yamashiro, Tokushima, 770-8514, Japan
| | - Takuto Ohashi
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihama-Boji, Yamashiro, Tokushima, 770-8514, Japan
| | - Toshiyuki Fukada
- Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihama-Boji, Yamashiro, Tokushima, 770-8514, Japan.
| |
Collapse
|
5
|
Georgiou K, Sarigol F, Nimpf T, Knapp C, Filipczak D, Foisner R, Naetar N. MyoD1 localization at the nuclear periphery is mediated by association of WFS1 with active enhancers. Nat Commun 2025; 16:2614. [PMID: 40097443 PMCID: PMC11914251 DOI: 10.1038/s41467-025-57758-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
Spatial organization of the mammalian genome influences gene expression and cell identity. While association of genes with the nuclear periphery is commonly linked to transcriptional repression, also active, expressed genes can localize at the nuclear periphery. The transcriptionally active MyoD1 gene, a master regulator of myogenesis, exhibits peripheral localization in proliferating myoblasts, yet the underlying mechanisms remain elusive. Here, we generate a reporter cell line to demonstrate that peripheral association of the MyoD1 locus is independent of mechanisms involved in heterochromatin anchoring. Instead, we identify the nuclear envelope transmembrane protein WFS1 that tethers MyoD1 to the nuclear periphery. WFS1 primarily associates with active distal enhancer elements upstream of MyoD1, and with a subset of enhancers genome-wide, which are enriched in active histone marks and linked to expressed myogenic genes. Overall, our data identify a mechanism involved in tethering regulatory elements of active genes to the nuclear periphery.
Collapse
Affiliation(s)
- Konstantina Georgiou
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Max Perutz Labs, Medical University of Vienna, Vienna, Austria
- Vienna BioCenter PhD Program, a Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Fatih Sarigol
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Max Perutz Labs, Medical University of Vienna, Vienna, Austria
| | - Tobias Nimpf
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Max Perutz Labs, Medical University of Vienna, Vienna, Austria
| | - Christian Knapp
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Max Perutz Labs, Medical University of Vienna, Vienna, Austria
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Castelldefels, Barcelona, Spain
| | - Daria Filipczak
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Max Perutz Labs, Medical University of Vienna, Vienna, Austria
- Vienna BioCenter PhD Program, a Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Roland Foisner
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria.
- Max Perutz Labs, Medical University of Vienna, Vienna, Austria.
| | - Nana Naetar
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria.
- Max Perutz Labs, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
6
|
Huang CJ, Choo KB. Circular RNAs and host genes act synergistically in regulating cellular processes and functions in skeletal myogenesis. Gene 2025; 940:149189. [PMID: 39724991 DOI: 10.1016/j.gene.2024.149189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/14/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Circular RNAs (circRNAs) are post-transcriptional regulators generated from backsplicing of pre-mRNAs of host genes. A major circRNA regulatory mechanism involves microRNA (miRNA) sequestering, relieving miRNA-blocked mRNAs for translation and functions. To investigate possible circRNA-host gene relationship, skeletal myogenesis is chosen as a study model for its developmental importance and for readily available muscle tissues from farm animals for studies at different myogenic stages. This review aims to provide an integrated interpretations on methodologies, regulatory mechanisms and possible host gene-circRNA synergistic functional relationships in skeletal myogenesis, focusing on myoblast differentiation and proliferation, core drivers of muscle formation in myogenesis, while other myogenic processes that play supportive roles in the structure, maintenance and function of muscle tissues are also briefly discussed. On literature review,thirty-two circRNAs derived from thirty-one host genes involved in various myogenic stages are identified; twenty-two (68.6 %) of these circRNAs regulate myogenesis by sequestering miRNAs to engage PI3K/AKT and other signaling pathways while four (12.5 %) are translated into proteins for functions. In circRNA-host gene relationship,ten (32.3 %) host genes are shown to regulate myogenesis,nine (29.0 %) are specific to skeletal muscle functions,and twelve (38.8 %) are linked to skeletal muscle disorders.Our analysis of skeletal myogenesis suggests that circRNAs and host genes act synergistically to regulate cellular functions. Such circRNA-host gene functional synergism may also be found in other major cellular processes. CircRNAs may have evolved later than miRNAs to counteract the suppressive effects of miRNAs and to augment host gene functions to further fine-tune gene regulation.
Collapse
Affiliation(s)
- Chiu-Jung Huang
- Department of Animal Science & Graduate Institute of Biotechnology, College of Environmental Planning & Bioresources (former School of Agriculture), Chinese Culture University, Taipei, Taiwan.
| | - Kong Bung Choo
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
7
|
Bai H, Ding J, Wang S, Zhang S, Jiang N, Wu X, Chen G, Dang Q, Liu M, Tang B, Wang X. Murine skeletal muscle satellite cells isolation and preliminary study on regulation in immune microenvironment during nurse cells formation of Trichinella spiralis infection. Vet Parasitol 2025; 333:110175. [PMID: 38614824 DOI: 10.1016/j.vetpar.2024.110175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/19/2024] [Accepted: 03/25/2024] [Indexed: 04/15/2024]
Abstract
As an intracellular parasitic nematode, Trichinella spiralis (T. spiralis) can induce the formation of nurse cells (NC) in host muscles and keep it to survive within the NC for an extended period. The formation of NC is similar to muscle cell injury and repair which lead to the arrest of satellite cells in the G2/M phase and build a suitable parasitic environment for the muscle larvae of T. spiralis. However, the molecular mechanisms involved in skeletal muscle repair through skeletal muscle satellite cells (SMSC) and the host immune response during T. spiralis infection have not been fully elucidated. In this study, histopathological examination revealed that the severity of damage increased as the infection progressed in the soleus muscle. SMSCs were isolated from BALB/c mice infected with T. spiralis at 4, 21 and 35 days post-infection (dpi). The immunological characteristics of these cells were analyzed by real-time PCR and flow cytometry (FCM). FCM analysis revealed a notable increase in the expression of B7 homolog 1 (B7-H1) in SMSCs following T. spiralis infection, while conversely, the expression of inducible costimulatory ligand (ICOSL) significantly decreased. Furthermore, real-time PCR results showed that toll like receptor 3 (TLR3) expression in SMSCs of the infected mice was upregulated at 21 dpi. The expression levels of three subtypes (PPARα, PPARβ and PPARγ) of peroxisome proliferator-activated receptors (PPARs) also increased in the cells. This study highlights the immunological regulation significance of SMSCs host during T. spiralis infection and suggests that SMSCs actively participant in the local immune response to T. spiralis by regulating the interaction between the parasite and the host.
Collapse
Affiliation(s)
- Huifang Bai
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jing Ding
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Saining Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Shuyan Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Ning Jiang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaoxia Wu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guoliang Chen
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Qianqian Dang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Mingyuan Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Bin Tang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xuelin Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
8
|
Barai P, Chen J. Cytokine expression and cytokine-mediated cell-cell communication during skeletal muscle regeneration revealed by integrative analysis of single-cell RNA sequencing data. J Cell Commun Signal 2024; 18:e12055. [PMID: 39691872 PMCID: PMC11647049 DOI: 10.1002/ccs3.12055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/20/2024] [Accepted: 10/15/2024] [Indexed: 12/19/2024] Open
Abstract
Skeletal muscles undergo self-repair upon injury, owing to the resident muscle stem cells and their extensive communication with the microenvironment of injured muscles. Cytokines play a critical role in orchestrating intercell communication to ensure successful regeneration. Immune cells as well as other types of cells in the injury site, including muscle stem cells, are known to secret cytokines. However, the extent to which various cell types express distinct cytokines and how the secreted cytokines are involved in intercell communication during regeneration are largely unknown. Here we integrated 15 publicly available single-cell RNA-sequencing (scRNA-seq) datasets of mouse skeletal muscles at early regeneration timepoints (0, 2, 5, and 7 days after injury). The resulting dataset was analyzed for the expression of 393 annotated mouse cytokines. We found widespread and dynamic cytokine expression by all cell types in the regenerating muscle. Interrogating the integrated dataset using CellChat revealed extensive, bidirectional cell-cell communications during regeneration. Our findings provide a comprehensive view of cytokine signaling in the regenerating muscle, which can guide future studies of ligand-receptor signaling and cell-cell interaction to achieve new mechanistic insights into the regulation of muscle regeneration.
Collapse
Affiliation(s)
- Pallob Barai
- Department of Cell and Developmental BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Jie Chen
- Department of Cell and Developmental BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Biomedical and Translational SciencesCarle Illinois College of MedicineUrbanaIllinoisUSA
| |
Collapse
|
9
|
Lee SH, Kim JM. Genome to phenome Association for Pork Belly Parameters Elucidates Three Regulation Distinctions: Adipogenesis, muscle formation, and their transcription factors. Meat Sci 2024; 217:109617. [PMID: 39116533 DOI: 10.1016/j.meatsci.2024.109617] [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: 02/15/2024] [Revised: 05/21/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
Genome to phenome analysis is necessary in livestock areas because of its various and complex phenotypes. Pork belly is a favorable part of meat worldwide, including East Asia. A previous study has suggested that the three key transcription factors (ZNF444, NFYA and PPARG) affecting pork belly traits include total volume, the volume of total fat and muscle, and component muscles of the corresponding slice. However, other transcription factor genes affecting each slice other than pork belly component traits still needed to be identified. Thus, we aimed to analyze pork belly components at the genome to phenome level for identifying key transcription factor genes and their co-associated networks. The range of node numbers against each component trait via the association weight matrix was from 598 to 3020. Premised on the result, an in silico functional approach was performed. Each co-association network enriched three key transcription factors in adipogenesis and skeletal muscle proliferation, mesoderm development, metabolism, and gene transcription. The three key transcription factors and their related genes may be useful in comprehending their effect of pork belly construction.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea.
| |
Collapse
|
10
|
Jiwan NC, Appell CR, Sterling R, Shen CL, Luk HY. The Effect of Geranylgeraniol and Ginger on Satellite Cells Myogenic State in Type 2 Diabetic Rats. Curr Issues Mol Biol 2024; 46:12299-12310. [PMID: 39590324 PMCID: PMC11592527 DOI: 10.3390/cimb46110730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Type 2 diabetes (T2D) is associated with increased inflammation and reactive oxygen species (ROS) in muscles, leading to basal satellite cell (SC) myogenic impairment (i.e., reduction in SC pool), which is critical for maintaining skeletal muscle mass. T2D may contribute to muscle atrophy, possibly due to reductions in the SC pool. Geranylgeraniol (GGOH) and ginger can reduce inflammation and enhance SC myogenesis in damaged muscles, thereby alleviating muscle atrophy; however, their effect on basal SC myogenic state and muscle mass in T2D rats is limited. Rats consumed a control diet (CON), high-fat diet with 35 mg/kg of streptozotocin (HFD), a HFD with 800 mg/kg body weight of GGOH (GG), or a HFD with 0.75% ginger root extract (GRE). In the eighth week, their soleus muscles were analyzed for Pax7, MyoD, and MSTN gene and protein expression, SC myogenic state, and muscle cross-sectional area (CSA). The HFD group had a significantly lower number of Pax7+/MyoD- and Pax7+/MSTN+ cells, less Pax7 and MyoD gene expression, and less MyoD and MSTN protein expression, with a smaller CSA than the CON group. Compared to the GG and GRE groups, the HFD group had a significantly lower number of Pax7+/MSTN+ cells, less MyoD protein expression, and smaller CSA. The GRE group also had a significantly lower number of Pax7-/MyoD+ and greater MSTN protein expression than the HFD group. Nevertheless, the CON group had a significantly greater number of Pax7+/MyoD- than the GG and GRE groups, and a greater number of Pax7-/MyoD+ cells than the GRE group with a larger CSA than the GG group. GGOH and ginger persevered muscle CSA, possibly through increased MyoD and the ability to maintain the SC pool in T2D rats.
Collapse
Affiliation(s)
- Nigel C. Jiwan
- Department of Kinesiology, Hope College, Holland, MI 49423, USA;
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79406, USA; (C.R.A.); (R.S.)
| | - Casey R. Appell
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79406, USA; (C.R.A.); (R.S.)
| | - Raoul Sterling
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79406, USA; (C.R.A.); (R.S.)
| | - Chwan-Li Shen
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Hui-Ying Luk
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79406, USA; (C.R.A.); (R.S.)
| |
Collapse
|
11
|
Wang X, Zong X, Ye M, Jin C, Xu T, Yang J, Gao C, Wang X, Yan H. Lysine Distinctively Manipulates Myogenic Regulatory Factors and Wnt/Ca 2+ Pathway in Slow and Fast Muscles, and Their Satellite Cells of Postnatal Piglets. Cells 2024; 13:650. [PMID: 38607088 PMCID: PMC11011516 DOI: 10.3390/cells13070650] [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: 12/05/2023] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024] Open
Abstract
Muscle regeneration, representing an essential homeostatic process, relies mainly on the myogenic progress of resident satellite cells, and it is modulated by multiple physical and nutritional factors. Here, we investigated how myogenic differentiation-related factors and pathways respond to the first limiting amino acid lysine (Lys) in the fast and slow muscles, and their satellite cells (SCs), of swine. Thirty 28-day-old weaned piglets with similar body weights were subjected to three diet regimens: control group (d 0-28: 1.31% Lys, n = 12), Lys-deficient group (d 0-28: 0.83% Lys, n = 12), and Lys rescue group (d 0-14: 0.83% Lys; d 15-28: 1.31% Lys, n = 6). Pigs on d 15 and 29 were selectively slaughtered for muscular parameters evaluation. Satellite cells isolated from fast (semimembranosus) and slow (semitendinosus) muscles were also selected to investigate differentiation ability variations. We found Lys deficiency significantly hindered muscle development in both fast and slow muscles via the distinct manipulation of myogenic regulatory factors and the Wnt/Ca2+ pathway. In the SC model, Lys deficiency suppressed the Wnt/Ca2+ pathways and myosin heavy chain, myogenin, and myogenic regulatory factor 4 in slow muscle SCs but stimulated them in fast muscle SCs. When sufficient Lys was attained, the fast muscle-derived SCs Wnt/Ca2+ pathway (protein kinase C, calcineurin, calcium/calmodulin-dependent protein kinase II, and nuclear factor of activated T cells 1) was repressed, while the Wnt/Ca2+ pathway of its counterpart was stimulated to further the myogenic differentiation. Lys potentially manipulates the differentiation of porcine slow and fast muscle myofibers via the Wnt/Ca2+ pathway in opposite trends.
Collapse
Affiliation(s)
- Xiaofan Wang
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Xiaoyin Zong
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Mao Ye
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Chenglong Jin
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Tao Xu
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Jinzeng Yang
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI 96822, USA;
| | - Chunqi Gao
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Xiuqi Wang
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| | - Huichao Yan
- College of Animal Science, South China Agricultural University, State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, Guangzhou 510642, China; (X.W.); (X.Z.); (M.Y.); (C.J.); (T.X.); (C.G.); (X.W.)
| |
Collapse
|
12
|
Olson LC, Nguyen T, Sabalewski EL, Puetzer JL, Schwartz Z, McClure MJ. S100b treatment overcomes RAGE signaling deficits in myoblasts on advanced glycation end-product cross-linked collagen and promotes myogenic differentiation. Am J Physiol Cell Physiol 2024; 326:C1080-C1093. [PMID: 38314727 DOI: 10.1152/ajpcell.00502.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Advanced glycation end-products (AGEs) stochastically accrue in skeletal muscle and on collagen over an individual's lifespan, stiffening the muscle and modifying the stem cell (MuSC) microenvironment while promoting proinflammatory, antiregenerative signaling via the receptor for advanced glycation end-products (RAGEs). In the present study, a novel in vitro model was developed of this phenomenon by cross linking a 3-D collagen scaffold with AGEs and investigating how myoblasts responded to such an environment. Briefly, collagen scaffolds were incubated with d-ribose (0, 25, 40, 100, or 250 mM) for 5 days at 37°C. C2C12 immortalized mouse myoblasts were grown on the scaffolds for 6 days in growth conditions for proliferation, and 12 days for differentiation and fusion. Human primary myoblasts were also used to confirm the C2C12 data. AGEs aberrantly extended the DNA production stage of C2C12s (but not in human primary myoblasts) which is known to delay differentiation in myogenesis, and this effect was prevented by RAGE inhibition. Furthermore, the differentiation and fusion of myoblasts were disrupted by AGEs, which were associated with reductions in integrins and suppression of RAGE. The addition of S100b (RAGE agonist) recovered the differentiation and fusion of myoblasts, and the addition of RAGE inhibitors (FPS-ZM1 and Azeliragon) inhibited the differentiation and fusion of myoblasts. Our results provide novel insights into the role of the AGE-RAGE axis in skeletal muscle aging, and future work is warranted on the potential application of S100b as a proregenerative factor in aged skeletal muscle.NEW & NOTEWORTHY Collagen cross-linked by advanced glycation end-products (AGEs) induced myoblast proliferation but prevented differentiation, myotube formation, and RAGE upregulation. RAGE inhibition occluded AGE-induced myoblast proliferation, while the delivery of S100b, a RAGE ligand, recovered fusion deficits.
Collapse
Affiliation(s)
- Lucas C Olson
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Gerontology, College of Health Professionals, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Tri Nguyen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Eleanor L Sabalewski
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Jennifer L Puetzer
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Orthopaedic Surgery, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Periodontics, School of Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Michael J McClure
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, Virginia, United States
- Department of Orthopaedic Surgery, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States
| |
Collapse
|
13
|
Sun C, Serra C, Kalicharan BH, Harding J, Rao M. Challenges and Considerations of Preclinical Development for iPSC-Based Myogenic Cell Therapy. Cells 2024; 13:596. [PMID: 38607035 PMCID: PMC11011706 DOI: 10.3390/cells13070596] [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: 02/06/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Cell therapies derived from induced pluripotent stem cells (iPSCs) offer a promising avenue in the field of regenerative medicine due to iPSCs' expandability, immune compatibility, and pluripotent potential. An increasing number of preclinical and clinical trials have been carried out, exploring the application of iPSC-based therapies for challenging diseases, such as muscular dystrophies. The unique syncytial nature of skeletal muscle allows stem/progenitor cells to integrate, forming new myonuclei and restoring the expression of genes affected by myopathies. This characteristic makes genome-editing techniques especially attractive in these therapies. With genetic modification and iPSC lineage specification methodologies, immune-compatible healthy iPSC-derived muscle cells can be manufactured to reverse the progression of muscle diseases or facilitate tissue regeneration. Despite this exciting advancement, much of the development of iPSC-based therapies for muscle diseases and tissue regeneration is limited to academic settings, with no successful clinical translation reported. The unknown differentiation process in vivo, potential tumorigenicity, and epigenetic abnormality of transplanted cells are preventing their clinical application. In this review, we give an overview on preclinical development of iPSC-derived myogenic cell transplantation therapies including processes related to iPSC-derived myogenic cells such as differentiation, scaling-up, delivery, and cGMP compliance. And we discuss the potential challenges of each step of clinical translation. Additionally, preclinical model systems for testing myogenic cells intended for clinical applications are described.
Collapse
Affiliation(s)
- Congshan Sun
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| | - Carlo Serra
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Mahendra Rao
- Vita Therapeutics, Baltimore, MD 21043, USA (M.R.)
| |
Collapse
|
14
|
Wang L, Valencak TG, Shan T. Fat infiltration in skeletal muscle: Influential triggers and regulatory mechanism. iScience 2024; 27:109221. [PMID: 38433917 PMCID: PMC10907799 DOI: 10.1016/j.isci.2024.109221] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Fat infiltration in skeletal muscle (also known as myosteatosis) is now recognized as a distinct disease from sarcopenia and is directly related to declining muscle capacity. Hence, understanding the origins and regulatory mechanisms of fat infiltration is vital for maintaining skeletal muscle development and improving human health. In this article, we summarized the triggering factors such as aging, metabolic diseases and metabolic syndromes, nonmetabolic diseases, and muscle injury that all induce fat infiltration in skeletal muscle. We discussed recent advances on the cellular origins of fat infiltration and found several cell types including myogenic cells and non-myogenic cells that contribute to myosteatosis. Furthermore, we reviewed the molecular regulatory mechanism, detection methods, and intervention strategies of fat infiltration in skeletal muscle. Based on the current findings, our review will provide new insight into regulating function and lipid metabolism of skeletal muscle and treating muscle-related diseases.
Collapse
Affiliation(s)
- Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| | | | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, China
| |
Collapse
|
15
|
Ahmad K, Lee EJ, Ali S, Han KS, Hur SJ, Lim JH, Choi I. Licochalcone A and B enhance muscle proliferation and differentiation by regulating Myostatin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155350. [PMID: 38237512 DOI: 10.1016/j.phymed.2024.155350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Myostatin (MSTN) inhibition has demonstrated promise for the treatment of diseases associated with muscle loss. In a previous study, we discovered that Glycyrrhiza uralensis (G. uralensis) crude water extract (CWE) inhibits MSTN expression while promoting myogenesis. Furthermore, three specific compounds of G. uralensis, namely liquiritigenin, tetrahydroxymethoxychalcone, and Licochalcone B (Lic B), were found to promote myoblast proliferation and differentiation, as well as accelerate the regeneration of injured muscle tissue. PURPOSE The purpose of this study was to build on our previous findings on G. uralensis and demonstrate the potential of its two components, Licochalcone A (Lic A) and Lic B, in muscle mass regulation (by inhibiting MSTN), aging and muscle formation. METHODS G. uralensis, Lic A, and Lic B were evaluated thoroughly using in silico, in vitro and in vivo approaches. In silico analyses included molecular docking, and dynamics simulations of these compounds with MSTN. Protein-protein docking was carried out for MSTN, as well as for the docked complex of MSTN-Lic with its receptor, activin type IIB receptor (ACVRIIB). Subsequent in vitro studies used C2C12 cell lines and primary mouse muscle stem cells to acess the cell proliferation and differentiation of normal and aged cells, levels of MSTN, Atrogin 1, and MuRF1, and plasma MSTN concentrations, employing techniques such as western blotting, immunohistochemistry, immunocytochemistry, cell proliferation and differentiation assays, and real-time RT-PCR. Furthermore, in vivo experiments using mouse models focused on measuring muscle fiber diameters. RESULTS CWE of G. uralensis and two of its components, namely Lic A and B, promote myoblast proliferation and differentiation by inhibiting MSTN and reducing Atrogin1 and MuRF1 expressions and MSTN protein concentration in serum. In silico interaction analysis revealed that Lic A (binding energy -6.9 Kcal/mol) and B (binding energy -5.9 Kcal/mol) bind to MSTN and reduce binding between it and ACVRIIB, thereby inhibiting downstream signaling. The experimental analysis, which involved both in vitro and in vivo studies, demonstrated that the levels of MSTN, Atrogin 1, and MuRF1 were decreased when G. uralensis CWE, Lic A, or Lic B were administered into mice or treated in the mouse primary muscle satellite cells (MSCs) and C2C12 myoblasts. The diameters of muscle fibers increased in orally treated mice, and the differentiation and proliferation of C2C12 cells were enhanced. G. uralensis CWE, Lic A, and Lic B also promoted cell proliferation in aged cells, suggesting that they may have anti-muslce aging properties. They also reduced the expression and phosphorylation of SMAD2 and SMAD3 (MSTN downstream effectors), adding to the evidence that MSTN is inhibited. CONCLUSION These findings suggest that CWE and its active constituents Lic A and Lic B have anti-mauscle aging potential. They also have the potential to be used as natural inhibitors of MSTN and as therapeutic options for disorders associated with muscle atrophy.
Collapse
Affiliation(s)
- Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Shahid Ali
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Ki Soo Han
- Neo Cremar Co., Ltd., Seoul 05702, South Korea
| | - Sun Jin Hur
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, South Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, South Korea.
| |
Collapse
|
16
|
Metti S, Da Ros F, Toniato G, Cescon M, Bonaldo P. Native collagen VI delays early muscle stem cell differentiation. J Cell Sci 2024; 137:jcs261419. [PMID: 38224152 PMCID: PMC10911284 DOI: 10.1242/jcs.261419] [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: 06/17/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024] Open
Abstract
Adult muscle stem cells (MuSCs) are critical for muscle homeostasis and regeneration, and their behavior relies on a finely regulated niche made of specific extracellular matrix (ECM) components and soluble factors. Among ECM proteins, collagen VI (Col6) influences the mechanical properties of the niche and, in turn, MuSC self-renewal capabilities. Here, we investigated whether Col6 can exert a direct function as a biochemical signal for regulating the stemness and differentiation of murine MuSCs and myoblasts. Native Col6, but not its pepsin-resistant fragment, counteracts the early differentiation of myogenic cells by reducing the expression of differentiation marker genes and preserving stemness features, with inhibition of the canonical Wnt pathway. Our data indicate that extracellular Col6 acts as a soluble ligand in delaying early myogenic differentiation by regulating intracellular signals involved in adult myogenesis.
Collapse
Affiliation(s)
- Samuele Metti
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Francesco Da Ros
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Giorgia Toniato
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
| |
Collapse
|
17
|
Johnson DC, Bryan EE, Burris ES, Dilger RN, Harsh BN, Dilger AC. Effects of maternal inflammation on growth performance, carcass characteristics, and meat quality of offspring pigs. J Anim Sci 2024; 102:skae215. [PMID: 39066604 PMCID: PMC11336995 DOI: 10.1093/jas/skae215] [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: 05/13/2024] [Accepted: 07/25/2024] [Indexed: 07/28/2024] Open
Abstract
The objective of this research was to determine the effects of mid-gestational maternal inflammation on performance, carcass characteristics, and meat quality of offspring. Pregnant gilts were administered either lipopolysaccharide (LPS; n = 7) or saline (CON, n = 7) from days 70 to 84 of gestation. Gilts assigned to the LPS treatment were administered an intravenous injection of reconstituted LPS every other day with a beginning dose of 10 μg LPS/kg body weight and subsequent doses increasing by 12%, while CON gilts received intravenous injections of comparable volumes of saline. Gilts farrowed naturally, and at day 66 of age, a total of 59 pigs, both barrows and gilts began a 3-phase feeding regimen designed to meet or exceed nutrient requirements for growing-finishing pigs. Pigs were weighed on days 0, 35, 70, and 105 of the finishing trial to determine average daily gain, average daily feed intake, and gain-to-feed ratio (G:F). On day 106, pigs were slaughtered under the supervision of the U.S. Department of Agriculture Food Safety Inspection Service. Ending live weight, hot carcass weight, and dressing percentage were determined. The left side of carcasses was weighed and fabricated to determine carcass cutting yields. The semitendinosus was collected for histological samples. Fresh belly characteristics and loin quality were measured. Two chops were collected for Warner-Bratzler shear force and proximate analysis. No differences (P ≥ 0.13) between LPS and CON pigs were observed for growth performance in phases 1, 2, 3, or overall (days 0 to 105) performance with the exception of overall G:F reduced in CON pigs compared with LPS pigs (P = 0.03). There was a tendency for carcass yield to be reduced (P = 0.06; 0.82% units) in LPS pigs compared with CON pigs. Additionally, longissimus muscle area tended to be reduced (P = 0.10) 2.27 cm2 in LPS compared with CON pigs. Loin chop quality traits including instrumental color, subjective color, marbling, firmness, pH, and drip loss were not different (P ≥ 0.09) between LPS and CON pigs. Fresh belly characteristics were not different (P ≥ 0.22) between LPS and CON pigs. There were no differences in primal and subprimal weights, except that LPS pigs tended to have a reduction (P ≥ 0.07) in tenderloin and Canadian back weights compared with CON pigs. Furthermore, LPS pigs had no differences (P ≥ 0.25) in muscle fiber composition or size; however, LPS pigs tended (P = 0.10) to have a 13% reduction in estimated muscle fibers number compared with CON pigs. In summary, mid-gestational inflammation did not result in reduced meat quality, growth performance, or carcass yields of offspring.
Collapse
Affiliation(s)
- Danielle C Johnson
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Erin E Bryan
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Elli S Burris
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Bailey N Harsh
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Anna C Dilger
- Department of Animal Sciences, University of Illinois, Urbana-Champaign, IL 61801, USA
| |
Collapse
|
18
|
Careccia G, Mangiavini L, Cirillo F. Regulation of Satellite Cells Functions during Skeletal Muscle Regeneration: A Critical Step in Physiological and Pathological Conditions. Int J Mol Sci 2023; 25:512. [PMID: 38203683 PMCID: PMC10778731 DOI: 10.3390/ijms25010512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Skeletal muscle regeneration is a complex process involving the generation of new myofibers after trauma, competitive physical activity, or disease. In this context, adult skeletal muscle stem cells, also known as satellite cells (SCs), play a crucial role in regulating muscle tissue homeostasis and activating regeneration. Alterations in their number or function have been associated with various pathological conditions. The main factors involved in the dysregulation of SCs' activity are inflammation, oxidative stress, and fibrosis. This review critically summarizes the current knowledge on the role of SCs in skeletal muscle regeneration. It examines the changes in the activity of SCs in three of the most common and severe muscle disorders: sarcopenia, muscular dystrophy, and cancer cachexia. Understanding the molecular mechanisms involved in their dysregulations is essential for improving current treatments, such as exercise, and developing personalized approaches to reactivate SCs.
Collapse
Affiliation(s)
- Giorgia Careccia
- Department of Biosciences, University of Milan, 20133 Milan, Italy;
| | - Laura Mangiavini
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Federica Cirillo
- IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
- Institute for Molecular and Translational Cardiology (IMTC), 20097 San Donato Milanese, Italy
| |
Collapse
|
19
|
Sharma G, Banerjee R, Srivastava S. Molecular Mechanisms and the Interplay of Important Chronic Obstructive Pulmonary Disease Biomarkers Reveals Novel Therapeutic Targets. ACS OMEGA 2023; 8:46376-46389. [PMID: 38107961 PMCID: PMC10719921 DOI: 10.1021/acsomega.3c07480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/02/2023] [Indexed: 12/19/2023]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a progressive, age-dependent, and unmet chronic inflammatory disease of the peripheral airways, leading to difficulty in exhalation. Several biomarkers have been tested in general towards the resolution for a long time, but no apparent success was achieved. Ongoing therapies of COPD have only symptomatic relief but no cure. Reactive oxygen species (ROS) are highly reactive species which include oxygen radicals and nonradical derivatives, and are the prominent players in COPD. They are produced as natural byproducts of cellular metabolism, but their levels can vary due to exposure to indoor air pollution, occupational pollution, and environmental pollutants such as cigarette smoke. In COPD, the lungs are continuously exposed to high levels of ROS thus leading to oxidative stress. ROS can cause damage to cells, proteins, lipids, and DNA which further contributes to the chronic inflammation in COPD and exacerbates the disease condition. Excessive ROS production can overwhelm cellular antioxidant systems and act as signaling molecules that regulate cellular processes, including antioxidant defense mechanisms involving glutathione and sirtuins which further leads to cellular apoptosis, cellular senescence, inflammation, and sarcopenia. In this review paper, we focused on COPD from different perspectives including potential markers and different cellular processes such as apoptosis, cellular senescence, inflammation, sirtuins, and sarcopenia, and tried to connect the dots between them so that novel therapeutic strategies to evaluate and target the possible underlying mechanisms in COPD could be explored.
Collapse
Affiliation(s)
- Gautam Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | | | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| |
Collapse
|
20
|
Ahmad K, Shaikh S, Chun HJ, Ali S, Lim JH, Ahmad SS, Lee EJ, Choi I. Extracellular matrix: the critical contributor to skeletal muscle regeneration-a comprehensive review. Inflamm Regen 2023; 43:58. [PMID: 38008778 PMCID: PMC10680355 DOI: 10.1186/s41232-023-00308-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/01/2023] [Indexed: 11/28/2023] Open
Abstract
The regenerative ability of skeletal muscle (SM) in response to damage, injury, or disease is a highly intricate process that involves the coordinated activities of multiple cell types and biomolecular factors. Of these, extracellular matrix (ECM) is considered a fundamental component of SM regenerative ability. This review briefly discusses SM myogenesis and regeneration, the roles played by muscle satellite cells (MSCs), other cells, and ECM components, and the effects of their dysregulations on these processes. In addition, we review the various types of ECM scaffolds and biomaterials used for SM regeneration, their applications, recent advances in ECM scaffold research, and their impacts on tissue engineering and SM regeneration, especially in the context of severe muscle injury, which frequently results in substantial muscle loss and impaired regenerative capacity. This review was undertaken to provide a comprehensive overview of SM myogenesis and regeneration, the stem cells used for muscle regeneration, the significance of ECM in SM regeneration, and to enhance understanding of the essential role of the ECM scaffold during SM regeneration.
Collapse
Affiliation(s)
- Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Hee Jin Chun
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Shahid Ali
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea.
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea.
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea.
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea.
| |
Collapse
|
21
|
Peng J, Han L, Liu B, Song J, Wang Y, Wang K, Guo Q, Liu X, Li Y, Zhang J, Wu W, Li S, Fu X, Zhuang CL, Zhang W, Suo S, Hu P, Zhao Y. Gli1 marks a sentinel muscle stem cell population for muscle regeneration. Nat Commun 2023; 14:6993. [PMID: 37914731 PMCID: PMC10620419 DOI: 10.1038/s41467-023-42837-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023] Open
Abstract
Adult skeletal muscle regeneration is mainly driven by muscle stem cells (MuSCs), which are highly heterogeneous. Although recent studies have started to characterize the heterogeneity of MuSCs, whether a subset of cells with distinct exists within MuSCs remains unanswered. Here, we find that a population of MuSCs, marked by Gli1 expression, is required for muscle regeneration. The Gli1+ MuSC population displays advantages in proliferation and differentiation both in vitro and in vivo. Depletion of this population leads to delayed muscle regeneration, while transplanted Gli1+ MuSCs support muscle regeneration more effectively than Gli1- MuSCs. Further analysis reveals that even in the uninjured muscle, Gli1+ MuSCs have elevated mTOR signaling activity, increased cell size and mitochondrial numbers compared to Gli1- MuSCs, indicating Gli1+ MuSCs are displaying the features of primed MuSCs. Moreover, Gli1+ MuSCs greatly contribute to the formation of GAlert cells after muscle injury. Collectively, our findings demonstrate that Gli1+ MuSCs represents a distinct MuSC population which is more active in the homeostatic muscle and enters the cell cycle shortly after injury. This population functions as the tissue-resident sentinel that rapidly responds to injury and initiates muscle regeneration.
Collapse
Affiliation(s)
- Jiayin Peng
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Lili Han
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Biao Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Jiawen Song
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Yuang Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Kunpeng Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai, 201210, PR China
| | - Qian Guo
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - XinYan Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Yu Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Jujin Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China
| | - Wenqing Wu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China
| | - Sheng Li
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200023, PR China
| | - Xin Fu
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200023, PR China
| | - Cheng-le Zhuang
- The 10th People's Hospital affiliated to Tongji University, Shanghai, 200072, PR China
| | - Weikang Zhang
- Guangzhou Laboratory-Guangzhou Medical University, Guangzhou, 510005, PR China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Shengbao Suo
- Guangzhou Laboratory-Guangzhou Medical University, Guangzhou, 510005, PR China
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, Guangdong, 510005, PR China
| | - Ping Hu
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200023, PR China.
- The 10th People's Hospital affiliated to Tongji University, Shanghai, 200072, PR China.
- Guangzhou Laboratory-Guangzhou Medical University, Guangzhou, 510005, PR China.
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510005, PR China.
| | - Yun Zhao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, PR China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, PR China.
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai, 201210, PR China.
| |
Collapse
|
22
|
Corvelyn M, Meirlevede J, Deschrevel J, Huyghe E, De Wachter E, Gayan-Ramirez G, Sampaolesi M, Van Campenhout A, Desloovere K, Costamagna D. Ex vivo adult stem cell characterization from multiple muscles in ambulatory children with cerebral palsy during early development of contractures. Differentiation 2023; 133:25-39. [PMID: 37451110 DOI: 10.1016/j.diff.2023.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 05/25/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
Cerebral palsy (CP) is one of the most common conditions leading to lifelong childhood physical disability. Literature reported previously altered muscle properties such as lower number of satellite cells (SCs), with altered fusion capacity. However, these observations highly vary among studies, possibly due to heterogeneity in patient population, lack of appropriate control data, methodology and different assessed muscle. In this study we aimed to strengthen previous observations and to understand the heterogeneity of CP muscle pathology. Myogenic differentiation of SCs from the Medial Gastrocnemius (MG) muscle of patients with CP (n = 16, 3-9 years old) showed higher fusion capacity compared to age-matched typically developing children (TD, n = 13). Furthermore, we uniquely assessed cells of two different lower limb muscles and showed a decreased myogenic potency in cells from the Semitendinosus (ST) compared to the MG (TD: n = 3, CP: n = 6). Longitudinal assessments, one year after the first botulinum toxin treatment, showed slightly reduced SC representations and lower fusion capacity (n = 4). Finally, we proved the robustness of our data, by assessing in parallel the myogenic capacity of two samples from the same TD muscle. In conclusion, these data confirmed previous findings of increased SC fusion capacity from MG muscle of young patients with CP compared to age-matched TD. Further elaboration is reported on potential factors contributing to heterogeneity, such as assessed muscle, CP progression and reliability of primary outcome parameters.
Collapse
Affiliation(s)
- M Corvelyn
- Stem Cell and Developmental Biology, Dept. of Development and Regeneration, KU Leuven, Belgium
| | - J Meirlevede
- Stem Cell and Developmental Biology, Dept. of Development and Regeneration, KU Leuven, Belgium
| | - J Deschrevel
- Laboratory of Respiratory Diseases and Thoracic Surgery, Dept. of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - E Huyghe
- Research Group for Neurorehabilitation, Dept. of Rehabilitation Sciences, KU Leuven, Belgium
| | - E De Wachter
- Dept. of Orthopaedic Surgery, University Hospitals Leuven, Belgium
| | - G Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery, Dept. of Chronic Diseases and Metabolism, KU Leuven, Belgium
| | - M Sampaolesi
- Stem Cell and Developmental Biology, Dept. of Development and Regeneration, KU Leuven, Belgium
| | - A Van Campenhout
- Dept. of Orthopaedic Surgery, University Hospitals Leuven, Belgium; Dept. of Development and Regeneration, KU Leuven, Belgium
| | - K Desloovere
- Research Group for Neurorehabilitation, Dept. of Rehabilitation Sciences, KU Leuven, Belgium.
| | - D Costamagna
- Stem Cell and Developmental Biology, Dept. of Development and Regeneration, KU Leuven, Belgium; Research Group for Neurorehabilitation, Dept. of Rehabilitation Sciences, KU Leuven, Belgium.
| |
Collapse
|
23
|
Maeno T, Arimatsu R, Ojima K, Yamaya Y, Imakyure H, Watanabe N, Komiya Y, Kobayashi K, Nakamura M, Nishimura T, Tatsumi R, Suzuki T. Netrin-4 synthesized in satellite cell-derived myoblasts stimulates autonomous fusion. Exp Cell Res 2023; 430:113698. [PMID: 37437770 DOI: 10.1016/j.yexcr.2023.113698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
Satellite cells are indispensable for skeletal muscle regeneration and hypertrophy by forming nascent myofibers (myotubes). They synthesize multi-potent modulator netrins (secreted subtypes: netrin-1, -3, and -4), originally found as classical neural axon guidance molecules. While netrin-1 and -3 have key roles in myogenic differentiation, the physiological significance of netrin-4 is still unclear. This study examined whether netrin-4 regulates myofiber type commitment and myotube formation. Initially, the expression profiles indicated that satellite cells isolated from the extensor digitorum longus muscle (EDL muscle: fast-twitch myofiber-abundant) expressed slightly more netrin-4 than the soleus muscle (slow-type abundant) cells. As netrin-4 knockdown inhibited both slow- and fast-type myotube formation, netrin-4 may not directly regulate myofiber type commitment. However, netrin-4 knockdown in satellite cell-derived myoblasts reduced the myotube fusion index, while exogenous netrin-4 promoted myotube formation, even though netrin-4 expression level was maximum during the initiation stage of myogenic differentiation. Furthermore, netrin-4 knockdown also inhibited MyoD (a master transcriptional factor of myogenesis) and Myomixer (a myoblast fusogenic molecule) expression. These data suggest that satellite cells synthesize netrin-4 during myogenic differentiation initiation to promote their own fusion, stimulating the MyoD-Myomixer signaling axis.
Collapse
Affiliation(s)
- Takahiro Maeno
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Rio Arimatsu
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Koichi Ojima
- Muscle Biology Research Unit, Division of Animal Products Research, Institute of Livestock and Grassland Science, NARO, Tsukuba, Japan
| | - Yuki Yamaya
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Hikaru Imakyure
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Naruha Watanabe
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Yusuke Komiya
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Ken Kobayashi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Mako Nakamura
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takanori Nishimura
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Ryuichi Tatsumi
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takahiro Suzuki
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
24
|
Pang KT, Loo LSW, Chia S, Ong FYT, Yu H, Walsh I. Insight into muscle stem cell regeneration and mechanobiology. Stem Cell Res Ther 2023; 14:129. [PMID: 37173707 PMCID: PMC10176686 DOI: 10.1186/s13287-023-03363-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Stem cells possess the unique ability to differentiate into specialized cell types. These specialized cell types can be used for regenerative medicine purposes such as cell therapy. Myosatellite cells, also known as skeletal muscle stem cells (MuSCs), play important roles in the growth, repair, and regeneration of skeletal muscle tissues. However, despite its therapeutic potential, the successful differentiation, proliferation, and expansion processes of MuSCs remain a significant challenge due to a variety of factors. For example, the growth and differentiation of MuSCs can be greatly influenced by actively replicating the MuSCs microenvironment (known as the niche) using mechanical forces. However, the molecular role of mechanobiology in MuSC growth, proliferation, and differentiation for regenerative medicine is still poorly understood. In this present review, we comprehensively summarize, compare, and critically analyze how different mechanical cues shape stem cell growth, proliferation, differentiation, and their potential role in disease development (Fig. 1). The insights developed from the mechanobiology of stem cells will also contribute to how these applications can be used for regenerative purposes using MuSCs.
Collapse
Affiliation(s)
- Kuin Tian Pang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.
- School of Chemistry, Chemical Engineering, and Biotechnology, Nanyang Technology University, 62 Nanyang Drive, N1.2-B3, Singapore, 637459, Singapore.
| | - Larry Sai Weng Loo
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sean Chia
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore
| | - Francesca Yi Teng Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hanry Yu
- Institute of Bioengineering and Bioimaging, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
- Interdisplinary Science and Engineering Program, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - Ian Walsh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
25
|
Cabezas F, Cabello-Verrugio C, González N, Salas J, Ramírez MJ, de la Vega E, Olguín HC. NEDD4-1 deficiency impairs satellite cell function during skeletal muscle regeneration. Biol Res 2023; 56:21. [PMID: 37147738 PMCID: PMC10161651 DOI: 10.1186/s40659-023-00432-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/17/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Satellite cells are tissue-specific stem cells primarily responsible for the regenerative capacity of skeletal muscle. Satellite cell function and maintenance are regulated by extrinsic and intrinsic mechanisms, including the ubiquitin-proteasome system, which is key for maintaining protein homeostasis. In this context, it has been shown that ubiquitin-ligase NEDD4-1 targets the transcription factor PAX7 for proteasome-dependent degradation, promoting muscle differentiation in vitro. Nonetheless, whether NEDD4-1 is required for satellite cell function in regenerating muscle remains to be determined. RESULTS Using conditional gene ablation, we show that NEDD4-1 loss, specifically in the satellite cell population, impairs muscle regeneration resulting in a significant reduction of whole-muscle size. At the cellular level, NEDD4-1-null muscle progenitors exhibit a significant decrease in the ability to proliferate and differentiate, contributing to the formation of myofibers with reduced diameter. CONCLUSIONS These results indicate that NEDD4-1 expression is critical for proper muscle regeneration in vivo and suggest that it may control satellite cell function at multiple levels.
Collapse
Affiliation(s)
- Felipe Cabezas
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, 7510157, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Natalia González
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jeremy Salas
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel J Ramírez
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo de la Vega
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hugo C Olguín
- Laboratory of Tissue Repair and Adult Stem Cells, Molecular and Cell Biology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
26
|
Muthamil S, Kim HY, Jang HJ, Lyu JH, Shin UC, Go Y, Park SH, Lee HG, Park JH. Understanding the relationship between cancer associated cachexia and hypoxia-inducible factor-1. Biomed Pharmacother 2023; 163:114802. [PMID: 37146421 DOI: 10.1016/j.biopha.2023.114802] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a multifactorial disorder characterized by an unrestricted loss of body weight as a result of muscle and adipose tissue atrophy. Cachexia is influenced by several factors, including decreased metabolic activity and food intake, an imbalance between energy uptake and expenditure, excessive catabolism, and inflammation. Cachexia is highly associated with all types of cancers responsible for more than half of cancer-related mortalities worldwide. In healthy individuals, adipose tissue significantly regulates energy balance and glucose homeostasis. However, in metastatic cancer patients, CAC occurs mainly because of an imbalance between muscle protein synthesis and degradation which are organized by certain extracellular ligands and associated signaling pathways. Under hypoxic conditions, hypoxia-inducible factor-1 (HIF-1α) accumulated and translocated to the nucleus and activate numerous genes involved in cell survival, invasion, angiogenesis, metastasis, metabolic reprogramming, and cancer stemness. On the other hand, the ubiquitination proteasome pathway is inhibited during low O2 levels which promote muscle wasting in cancer patients. Therefore, understanding the mechanism of the HIF-1 pathway and its metabolic adaptation to biomolecules is important for developing a novel therapeutic method for cancer and cachexia therapy. Even though many HIF inhibitors are already in a clinical trial, their mechanism of action remains unknown. With this background, this review summarizes the basic concepts of cachexia, the role of inflammatory cytokines, pathways connected with cachexia with special reference to the HIF-1 pathway and its regulation, metabolic changes, and inhibitors of HIFs.
Collapse
Affiliation(s)
- Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
27
|
Chen W, Perkins TJ, Rudnicki MA. Quantification of Muscle Satellite Stem Cell Divisions by High-Content Analysis. Methods Mol Biol 2023; 2587:537-553. [PMID: 36401049 DOI: 10.1007/978-1-0716-2772-3_29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
High-content screening is commonly performed on 2D cultured cells, which is high throughput but has low biological relevance. In contrast, single myofiber culture assay preserves the satellite cell niche between the basal lamina and sarcolemma and consequently has high biological relevance but is low throughput. We describe here a high-content screening method that utilizes single myofiber culture that addresses the caveats of both techniques. Our method utilizes the transgenic reporter allele Myf5-Cre:R26R-eYFP to differentiate stem and committed cells within a dividing couplet that can be quantified by high-content throughput immunodetection and bioinformatic analysis.
Collapse
Affiliation(s)
- William Chen
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Theodore J Perkins
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
28
|
Eliazer S, Sun X, Barruet E, Brack AS. Heterogeneous levels of delta-like 4 within a multinucleated niche cell maintains muscle stem cell diversity. eLife 2022; 11:68180. [PMID: 36583937 PMCID: PMC9803355 DOI: 10.7554/elife.68180] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 12/19/2022] [Indexed: 12/31/2022] Open
Abstract
The quiescent muscle stem cell (QSC) pool is heterogeneous and generally characterized by the presence and levels of intrinsic myogenic transcription factors. Whether extrinsic factors maintain the diversity of states across the QSC pool remains unknown. The muscle fiber is a multinucleated syncytium that serves as a niche to QSCs, raising the possibility that the muscle fiber regulates the diversity of states across the QSC pool. Here, we show that the muscle fiber maintains a continuum of quiescent states, through a gradient of Notch ligand, Dll4, produced by the fiber and captured by QSCs. The abundance of Dll4 captured by the QSC correlates with the protein levels of the stem cell (SC) identity marker, Pax7. Niche-specific loss of Dll4 decreases QSC diversity and shifts the continuum to cell states that are biased toward more proliferative and committed fates. We reveal that fiber-derived Mindbomb1 (Mib1), an E3 ubiquitin ligase activates Dll4 and controls the heterogeneous levels of Dll4. In response to injury, with a Dll4-replenished niche, the normal continuum and diversity of the SC pool is restored, demonstrating bidirectionality within the SC continuum. Our data show that a post-translational mechanism controls heterogeneity of Notch ligands in a multinucleated niche cell to maintain a continuum of metastable states within the SC pool during tissue homeostasis.
Collapse
Affiliation(s)
- Susan Eliazer
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Orthopedic Surgery, University of California San FranciscoSan FranciscoUnited States
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health SciencesGrand ForksUnited States
| | - Xuefeng Sun
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Orthopedic Surgery, University of California San FranciscoSan FranciscoUnited States
| | - Emilie Barruet
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Orthopedic Surgery, University of California San FranciscoSan FranciscoUnited States
- Departments of Surgery and Orofacial Sciences, Program in Craniofacial Biology, University of California San FranciscoSan FranciscoUnited States
| | - Andrew S Brack
- The Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Department of Orthopedic Surgery, University of California San FranciscoSan FranciscoUnited States
| |
Collapse
|
29
|
Skeletal Muscle Stem Cells in Aging: Asymmetric/Symmetric Division Switching. Symmetry (Basel) 2022. [DOI: 10.3390/sym14122676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In aged muscle, satellite cells’ symmetric and asymmetric divisions are impaired, and intrinsic and extrinsic complex mechanisms govern these processes. This review presents many updated aspects regarding muscle stem cells’ fate in normal and aging conditions. The balance between self-renewal and commitment divisions contributes to muscle regeneration, muscle homeostasis, aging, and disease. Stimulating muscle regeneration in aging could be a therapeutic target, but there is still a need to understand the many mechanisms that influence each other in satellite cells and their niche. We highlight here the general outlines regarding satellite cell divisions, the primary markers present in muscle stem cells, the aging aspects concerning signaling pathways involved in symmetric/asymmetric divisions, the regenerative capacity of satellite cells and their niche alteration in senescent muscle, genetics and epigenetics mechanisms implied in satellite cells aging and exercise effect on muscle regeneration in the elderly.
Collapse
|
30
|
Shao X, Gong W, Wang Q, Wang P, Shi T, Mahmut A, Qin J, Yao Y, Yan W, Chen D, Chen X, Jiang Q, Guo B. Atrophic skeletal muscle fibre-derived small extracellular vesicle miR-690 inhibits satellite cell differentiation during ageing. J Cachexia Sarcopenia Muscle 2022; 13:3163-3180. [PMID: 36237168 PMCID: PMC9745557 DOI: 10.1002/jcsm.13106] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Sarcopenia is a common and progressive skeletal muscle disorder characterized by atrophic muscle fibres and contractile dysfunction. Accumulating evidence shows that the number and function of satellite cells (SCs) decline and become impaired during ageing, which may contribute to impaired regenerative capacity. A series of myokines/small extracellular vesicles (sEVs) released from muscle fibres regulate metabolism in muscle and extramuscular tissues in an autocrine/paracrine/endocrine manner during muscle atrophy. It is still unclear whether myokines/sEVs derived from muscle fibres can affect satellite cell function during ageing. METHODS Aged mice were used to investigate changes in the myogenic capacity of SCs during ageing-induced muscle atrophy. The effects of atrophic myotube-derived sEVs on satellite cell differentiation were investigated by biochemical methods and immunofluorescence staining. Small RNA sequencing was performed to identify differentially expressed sEV microRNAs (miRNAs) between the control myotubes and atrophic myotubes. The target genes of the miRNA were predicted by bioinformatics analysis and verified by luciferase activity assays. The effects of identified miRNA on the myogenic capacity of SCs in vivo were investigated by intramuscular injection of adeno-associated virus (AAV) to overexpress or silence miRNA in skeletal muscle. RESULTS Our study showed that the myogenic capacity of SCs was significantly decreased (50%, n = 6, P < 0.001) in the tibialis anterior muscle of aged mice. We showed that atrophic myotube-derived sEVs inhibited satellite cell differentiation in vitro (n = 3, P < 0.001) and in vivo (35%, n = 6, P < 0.05). We also found that miR-690 was the most highly enriched miRNA among all the screened sEV miRNAs in atrophic myotubes [Log2 (Fold Change) = 7, P < 0.001], which was verified in the atrophic muscle of aged mice (threefold, n = 6, P < 0.001) and aged men with mean age of 71 ± 5.27 years (2.8-fold, n = 10, P < 0.001). MiR-690 can inhibit myogenic capacity of SCs by targeting myocyte enhancer factor 2, including Mef2a, Mef2c and Mef2d, in vitro (n = 3, P < 0.05) and in vivo (n = 6, P < 0.05). Specific silencing of miR-690 in the muscle can promote satellite cell differentiation (n = 6, P < 0.001) and alleviate muscle atrophy in aged mice (n = 6, P < 0.001). CONCLUSIONS Our study demonstrated that atrophic muscle fibre-derived sEV miR-690 may inhibit satellite cell differentiation by targeting myocyte enhancer factor 2 during ageing.
Collapse
Affiliation(s)
- Xiaoyan Shao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Wang Gong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qianjin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Pu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Abdurahman Mahmut
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jianghui Qin
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yao Yao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenjin Yan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Dongyang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China.,Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Baosheng Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University & Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| |
Collapse
|
31
|
Alfaqih MS, Tarawan VM, Sylviana N, Goenawan H, Lesmana R, Susianti S. Effects of Vitamin D on Satellite Cells: A Systematic Review of In Vivo Studies. Nutrients 2022; 14:4558. [PMID: 36364820 PMCID: PMC9657163 DOI: 10.3390/nu14214558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 08/30/2023] Open
Abstract
The non-classical role of vitamin D has been investigated in recent decades. One of which is related to its role in skeletal muscle. Satellite cells are skeletal muscle stem cells that play a pivotal role in skeletal muscle growth and regeneration. This systematic review aims to investigate the effect of vitamin D on satellite cells. A systematic search was performed in Scopus, MEDLINE, and Google Scholar. In vivo studies assessing the effect of vitamin D on satellite cells, published in English in the last ten years were included. Thirteen in vivo studies were analyzed in this review. Vitamin D increases the proliferation of satellite cells in the early life period. In acute muscle injury, vitamin D deficiency reduces satellite cells differentiation. However, administering high doses of vitamin D impairs skeletal muscle regeneration. Vitamin D may maintain satellite cell quiescence and prevent spontaneous differentiation in aging. Supplementation of vitamin D ameliorates decreased satellite cells' function in chronic disease. Overall, evidence suggests that vitamin D affects satellite cells' function in maintaining skeletal muscle homeostasis. Further research is needed to determine the most appropriate dose of vitamin D supplementation in a specific condition for the optimum satellite cells' function.
Collapse
Affiliation(s)
- Muhammad Subhan Alfaqih
- Biomedical Science Master Program, Faculty of Medicine, Universitas Padjadjaran, Jl. Prof Eyckman No.38, Bandung 45363, Indonesia
| | - Vita Murniati Tarawan
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Nova Sylviana
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Central Laboratory, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Hanna Goenawan
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Central Laboratory, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Ronny Lesmana
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor 45363, Indonesia
- Central Laboratory, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| | - Susianti Susianti
- Central Laboratory, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| |
Collapse
|
32
|
Go S, Sato C, Hane M, Go S, Kitajima K. Implication of N-glycolylneuraminic acid in regulation of cell adhesiveness of C2C12 myoblast cells during differentiation into myotube cells. Glycoconj J 2022; 39:619-631. [PMID: 35639196 DOI: 10.1007/s10719-022-10049-9] [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: 10/20/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 11/26/2022]
Abstract
A transition of sialic acid (Sia) species on GM3 ganglioside from N-acetylneuraminic acid (Neu5Ac) to N-glycolylneuraminic acid (Neu5Gc) takes place in mouse C2C12 myoblast cells during their differentiation into myotube cells. However, the meaning of this Sia transition remains unclear. This study thus aims to gain a functional insight into this phenomenon. The following lines of evidence show that the increased de novo synthesis of Neu5Gc residues in differentiating myoblast cells promotes adhesiveness of the cells, which is beneficial for promotion of differentiation. First, the Sia transition occurred even in the C2C12 cells cultured in serum-free medium, indicating that it happens through de novo synthesis of Neu5Gc. Second, GM3(Neu5Gc) was localized in myoblast cells, but not in myotube cells, and related to expression of the CMP-Neu5Ac hydroxylase (CMAH) gene. Notably, expression of CMAH precedes myotube formation not only in differentiating C2C12 cells, but also in mouse developing embryos. Since the myoblast cells were attached on the dish surface more strongly than the myotube cells, expression of GM3(Neu5Gc) may be related to the surface attachment of the myoblast cells. Third, exogenous Neu5Gc, but not Neu5Ac, promoted differentiation of C2C12 cells, thus increasing the number of cells committed to fuse with each other. Fourth, the CMAH-transfected C2C12 cells were attached on the gelatin-coated surface much more rapidly than the mock-cells, suggesting that the expression of CMAH promotes cell adhesiveness through the expression of Neu5Gc.
Collapse
Affiliation(s)
- Shiori Go
- Graduate School of Bioagricultural Sciences and Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Chihiro Sato
- Graduate School of Bioagricultural Sciences and Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Masaya Hane
- Graduate School of Bioagricultural Sciences and Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Shinji Go
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Ken Kitajima
- Graduate School of Bioagricultural Sciences and Bioscience and Biotechnology Center, Nagoya University, Nagoya, 464-8601, Japan.
- Institute for Glyco-Core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan.
| |
Collapse
|
33
|
Thomas ACQ, Brown A, Hatt AA, Manta K, Costa-Parke A, Kamal M, Joanisse S, McGlory C, Phillips SM, Kumbhare D, Parise G. Short-term aerobic conditioning prior to resistance training augments muscle hypertrophy and satellite cell content in healthy young men and women. FASEB J 2022; 36:e22500. [PMID: 35971745 DOI: 10.1096/fj.202200398rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/22/2022] [Accepted: 08/02/2022] [Indexed: 11/11/2022]
Abstract
Factors influencing inter-individual variability of responses to resistance training (RT) remain to be fully elucidated. We have proposed the importance of capillarization in skeletal muscle for the satellite cell (SC) response to RT-induced muscle hypertrophy, and hypothesized that aerobic conditioning (AC) would augment RT-induced adaptations. Fourteen healthy young (22 ± 2 years) men and women underwent AC via 6 weeks of unilateral cycling followed by 10 weeks of bilateral RT to investigate how AC alters SC content, activity, and muscle hypertrophy following RT. Muscle biopsies were taken at baseline (unilateral), post AC (bilateral), and post RT (bilateral) in the aerobically conditioned (AC + RT) and unconditioned (RT) legs. Immunofluorescence was used to determine muscle capillarization, fiber size, SC content, and activity. Type I and type II fiber cross-sectional area (CSA) increased following RT, and when legs were analyzed independently, AC + RT increased type I, type II, and mixed-fiber CSA, where the RT leg tended to increase type II (p = .05), but not type I or mixed-fiber CSA. SC content, activation, and differentiation increased with RT, where type I total and quiescent SC content was greater in AC + RT compared to the RT leg. Those with the greatest capillary-to-fiber perimeter exchange index before RT had the greatest change in CSA following RT and a significant relationship was observed between type II fiber capillarization and the change in type II-fiber CSA with RT (r = 0.35). This study demonstrates that AC prior to RT can augment RT-induced muscle adaptions and that these differences are associated with increases in capillarization.
Collapse
Affiliation(s)
- Aaron C Q Thomas
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Alex Brown
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Aidan A Hatt
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Katherine Manta
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Anamaria Costa-Parke
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Michael Kamal
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | - Sophie Joanisse
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada.,Musculoskeletal Sciences and Sport Medicine Research Centre, Department of Sport and Exercise Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK.,Manchester Metropolitan University Institute of Sport, Manchester, UK
| | - Chris McGlory
- School of Kinesiology and Health Studies, Queen's University, Kingston, Ontario, Canada.,Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stuart M Phillips
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| | | | - Gianni Parise
- Exercise Metabolism Research Group, Department of Kinesiology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
34
|
Downregulation of Sparc-like protein 1 during cisplatin-induced inhibition of myogenic differentiation of C2C12 myoblasts. Biochem Pharmacol 2022; 204:115234. [PMID: 36041542 DOI: 10.1016/j.bcp.2022.115234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 11/20/2022]
Abstract
Patients with cancer often experience muscle atrophy, which worsens their prognosis. Decreased muscle regenerative capacity plays an important role in the complex processes involved in muscle atrophy. Administration of cisplatin, a cancer chemotherapeutic agent, has been implicated as a cause of muscle atrophy. In this study, we examined whether cisplatin affects the differentiation of myoblasts into myotubes. We treated C2C12 myoblasts with a differentiation medium containing cisplatin and its vehicle during for 8 days and observed the changes in the expression of myosin heavy chain (MyHC) and myogenin in the myoblasts. Cisplatin was injected in mice for 4 consecutive days; on Day 5, the mice quadriceps muscles were sampled and examined. The expression of MyHCs increased and that of myogenin decreased after cisplatin treatment. The secretion of acidic cysteine-rich proteins (e.g., Sparc proteins) reportedly promotes C2C12 myoblast differentiation. Therefore, we investigated the Sparc family gene expression during myogenesis in C2C12 myoblasts after cisplatin treatment. Of all the genes investigated, Sparc-like protein 1 (Sparcl1) expression was significantly suppressed by cisplatin on Days 4-8. Simultaneous treatment with recombinant mouse Sparcl1 almost inhibited the cisplatin-induced suppression of total MyHC and myogenin protein levels. Moreover, Sparcl1 expression decreased in the skeletal muscles of mice, leading to cisplatin-induced muscle atrophy. Our results suggest that cisplatin-induced myogenesis suppression causes muscle atrophy and inhibits the expression of Sparcl1, which promotes C2C12 cell differentiation during myogenesis.
Collapse
|
35
|
Epigenetic Alterations in Sports-Related Injuries. Genes (Basel) 2022; 13:genes13081471. [PMID: 36011382 PMCID: PMC9408207 DOI: 10.3390/genes13081471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
It is a well-known fact that physical activity benefits people of all age groups. However, highly intensive training, maladaptation, improper equipment, and lack of sufficient rest lead to contusions and sports-related injuries. From the perspectives of sports professionals and those performing regular–amateur sports activities, it is important to maintain proper levels of training, without encountering frequent injuries. The bodily responses to physical stress and intensive physical activity are detected on many levels. Epigenetic modifications, including DNA methylation, histone protein methylation, acetylation, and miRNA expression occur in response to environmental changes and play fundamental roles in the regulation of cellular activities. In the current review, we summarise the available knowledge on epigenetic alterations present in tissues and organs (e.g., muscles, the brain, tendons, and bones) as a consequence of sports-related injuries. Epigenetic mechanism observations have the potential to become useful tools in sports medicine, as predictors of approaching pathophysiological alterations and injury biomarkers that have already taken place.
Collapse
|
36
|
Bidirectional roles of skeletal muscle fibro-adipogenic progenitors in homeostasis and disease. Ageing Res Rev 2022; 80:101682. [PMID: 35809776 DOI: 10.1016/j.arr.2022.101682] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/09/2022] [Accepted: 07/04/2022] [Indexed: 02/07/2023]
Abstract
Sarcopenia and myopathies cause progressive muscle weakness and degeneration, which are closely associated with fat infiltration and fibrosis in muscle. Recently, experimental research has shed light on fibro-adipogenic progenitors (FAPs), also known as muscle-resident mesenchymal progenitors with multiple differentiation potential for adipogenesis, fibrosis, osteogenesis and chondrogenesis. They are considered key regulators of muscle homeostasis and integrity. They play supportive roles in muscle development and repair by orchestrating the regulatory interplay between muscle stem cells (MuSCs) and immune cells. Interestingly, FAPs also contribute to intramuscular fat infiltration, fibrosis and other pathologies when the functional integrity of the network is compromised. In this review, we summarize recent insights into the roles of FAPs in maintenance of skeletal muscle homeostasis, and discuss the underlying mechanisms regulating FAPs behavior and fate, highlighting their roles in participating in efficient muscle repair and fat infiltrated muscle degeneration as well as during muscle atrophy. We suggest that controlling and predicting FAPs differentiation may become a promising strategy to improve muscle function and prevent irreparable muscle damage.
Collapse
|
37
|
Baig MH, Ahmad K, Moon JS, Park SY, Ho Lim J, Chun HJ, Qadri AF, Hwang YC, Jan AT, Ahmad SS, Ali S, Shaikh S, Lee EJ, Choi I. Myostatin and its Regulation: A Comprehensive Review of Myostatin Inhibiting Strategies. Front Physiol 2022; 13:876078. [PMID: 35812316 PMCID: PMC9259834 DOI: 10.3389/fphys.2022.876078] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022] Open
Abstract
Myostatin (MSTN) is a well-reported negative regulator of muscle growth and a member of the transforming growth factor (TGF) family. MSTN has important functions in skeletal muscle (SM), and its crucial involvement in several disorders has made it an important therapeutic target. Several strategies based on the use of natural compounds to inhibitory peptides are being used to inhibit the activity of MSTN. This review delivers an overview of the current state of knowledge about SM and myogenesis with particular emphasis on the structural characteristics and regulatory functions of MSTN during myogenesis and its involvements in various muscle related disorders. In addition, we review the diverse approaches used to inhibit the activity of MSTN, especially in silico approaches to the screening of natural compounds and the design of novel short peptides derived from proteins that typically interact with MSTN.
Collapse
Affiliation(s)
- Mohammad Hassan Baig
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Jun Sung Moon
- Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, South Korea
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Jeong Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Hee Jin Chun
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
| | - Afsha Fatima Qadri
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Ye Chan Hwang
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | - Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Shahid Ali
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- *Correspondence: Eun Ju Lee, ; Inho Choi,
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, South Korea
- *Correspondence: Eun Ju Lee, ; Inho Choi,
| |
Collapse
|
38
|
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
|
39
|
Wang J, Broer T, Chavez T, Zhou CJ, Tran S, Xiang Y, Khodabukus A, Diao Y, Bursac N. Myoblast deactivation within engineered human skeletal muscle creates a transcriptionally heterogeneous population of quiescent satellite-like cells. Biomaterials 2022; 284:121508. [PMID: 35421801 PMCID: PMC9289780 DOI: 10.1016/j.biomaterials.2022.121508] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 03/18/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022]
Abstract
Satellite cells (SCs), the adult Pax7-expressing stem cells of skeletal muscle, are essential for muscle repair. However, in vitro investigations of SC function are challenging due to isolation-induced SC activation, loss of native quiescent state, and differentiation to myoblasts. In the present study, we optimized methods to deactivate in vitro expanded human myoblasts within a 3D culture environment of engineered human skeletal muscle tissues ("myobundles"). Immunostaining and gene expression analyses revealed that a fraction of myoblasts within myobundles adopted a quiescent phenotype (3D-SCs) characterized by increased Pax7 expression, cell cycle exit, and activation of Notch signaling. Similar to native SCs, 3D-SC quiescence is regulated by Notch and Wnt signaling while loss of quiescence and reactivation of 3D-SCs can be induced by growth factors including bFGF. Myobundle injury with a bee toxin, melittin, induces robust myofiber fragmentation, functional decline, and 3D-SC proliferation. By applying single cell RNA-sequencing (scRNA-seq), we discover the existence of two 3D-SC subpopulations (quiescent and activated), identify deactivation-associated gene signature using trajectory inference between 2D myoblasts and 3D-SCs, and characterize the transcriptomic changes within reactivated 3D-SCs in response to melittin-induced injury. These results demonstrate the ability of an in vitro engineered 3D human skeletal muscle environment to support the formation of a quiescent and heterogeneous SC population recapitulating several aspects of the native SC phenotype, and provide a platform for future studies of human muscle regeneration and disease-associated SC dysfunction.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Torie Broer
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Taylor Chavez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Chris J Zhou
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sabrina Tran
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yu Xiang
- Department of Cell Biology, Duke University, Durham, NC, USA
| | | | - Yarui Diao
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
40
|
Jin CL, Ye M, Song ZW, Zhang ZM, Gao CQ, Yan HC, Wang XQ. Lysine Interacts with Frizzled7 to Activate β-Catenin in Satellite Cell-Participated Skeletal Muscle Growth. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3745-3756. [PMID: 35312309 DOI: 10.1021/acs.jafc.2c01027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
This work provided an interesting finding of lysine (Lys) control on skeletal muscle growth besides protein synthesis. According to the isobaric tag for relative and absolute quantitation and molecular docking analyses, we found both in in vivo skeletal muscle and in vitro muscle satellite cells (MuSCs) that the frizzled7 (FZD7) expression level was positively correlated with Lys levels and this was consistent with the activation of the Wnt/β-catenin pathway. On the other hand, FZD7 inhibition suppressed the Lys-rescued Wnt/β-catenin pathway, FZD7 knockdown caused cell proliferation, and Wnt/β-catenin pathway restrictions could not be compensated for by Lys or Wnt3a. Furthermore, the combination between Lys and recombinant pig frizzled7 (rpFZD7) protein was confirmed by isothermal titration calorimetry. This finding displayed concrete evidence that Lys is not only a molecular block of protein synthesis but is also a ligand for FZD7 to activate β-catenin to stimulate MuSCs in promoting skeletal muscle growth.
Collapse
Affiliation(s)
- Cheng-Long Jin
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong 510642, China
| | - Mao Ye
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong 510642, China
| | - Zhi-Wen Song
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong 510642, China
| | - Zong-Ming Zhang
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong 510642, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong 510642, China
| | - Hui-Chao Yan
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong 510642, China
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University/Guangdong Laboratory for Lingnan Modern Agriculture/Guangdong Provincial Key Laboratory of Animal Nutrition Control/National Engineering Research Center for Breeding Swine Industry, Guangzhou, Guangdong 510642, China
| |
Collapse
|
41
|
Su W, Liang L, Zhou L, Cao Y, Zhou X, Liu S, Wang Q, Zhang H. Macrophage Paired Immunoglobulin-Like Receptor B Deficiency Promotes Peripheral Atherosclerosis in Apolipoprotein E–Deficient Mice. Front Cell Dev Biol 2022; 9:783954. [PMID: 35321392 PMCID: PMC8936951 DOI: 10.3389/fcell.2021.783954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/13/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Peripheral atherosclerotic disease (PAD) is the narrowing or blockage of arteries that supply blood to the lower limbs. Given its complex nature, bioinformatics can help identify crucial genes involved in the progression of peripheral atherosclerosis. Materials and Methods: Raw human gene expression data for 462 PAD arterial plaque and 23 normal arterial samples were obtained from the GEO database. The data was analyzed using an integrated, multi-layer approach involving differentially-expressed gene analysis, KEGG pathway analysis, GO term enrichment analysis, weighted gene correlation network analysis, and protein-protein interaction analysis. The monocyte/macrophage-expressed leukocyte immunoglobulin-like receptor B2 (LILRB2) was strongly associated with the human PAD phenotype. To explore the role of the murine LILRB2 homologue PirB in vivo, we created a myeloid-specific PirB-knockout Apoe−/− murine model of PAD (PirBMΦKO) to analyze femoral atherosclerotic burden, plaque features of vulnerability, and monocyte recruitment to femoral atherosclerotic lesions. The phenotypes of PirBMΦKO macrophages under various stimuli were also investigated in vitro. Results:PirBMΦKO mice displayed increased femoral atherogenesis, a more vulnerable plaque phenotype, and enhanced monocyte recruitment into lesions. PirBMΦKO macrophages showed enhanced pro-inflammatory responses and a shift toward M1 over M2 polarization under interferon-γ and oxidized LDL exposure. PirBMΦKO macrophages also displayed enhanced efferocytosis and reduced lipid efflux under lipid exposure. Conclusion: Macrophage PirB reduces peripheral atherosclerotic burden, stabilizes peripheral plaque composition, and suppresses macrophage accumulation in peripheral lesions. Macrophage PirB inhibits pro-inflammatory activation, inhibits efferocytosis, and promotes lipid efflux, characteristics critical to suppressing peripheral atherogenesis.
Collapse
Affiliation(s)
- Wenhua Su
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
- Faculty of Life Science and Biotechnology, Kunming University of Science and Technology, Kunming, China
| | - Liwen Liang
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Liang Zhou
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Yu Cao
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
- Department of Cardiovascular Surgery, First People’s Hospital of Yunnan Province, Kunming, China
| | - Xiuli Zhou
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Shiqi Liu
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Qian Wang
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
| | - Hong Zhang
- Department of Cardiology, First People’s Hospital of Yunnan Province, Kunming, China
- *Correspondence: Hong Zhang,
| |
Collapse
|
42
|
McKee CM, Chapski DJ, Wehling-Henricks M, Rosa-Garrido M, Kuro-O M, Vondriska TM, Tidball JG. The anti-aging protein Klotho affects early postnatal myogenesis by downregulating Jmjd3 and the canonical Wnt pathway. FASEB J 2022; 36:e22192. [PMID: 35174906 PMCID: PMC9007106 DOI: 10.1096/fj.202101298r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/15/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022]
Abstract
Modulating the number of muscle stems cells, called satellite cells, during early postnatal development produces long-term effects on muscle growth. We tested the hypothesis that high expression levels of the anti-aging protein Klotho in early postnatal myogenesis increase satellite cell numbers by influencing the epigenetic regulation of genes that regulate myogenesis. Our findings show that elevated klotho expression caused a transient increase in satellite cell numbers and slowed muscle fiber growth, followed by a period of accelerated muscle growth that leads to larger fibers. Klotho also transcriptionally downregulated the H3K27 demethylase Jmjd3, leading to increased H3K27 methylation and decreased expression of genes in the canonical Wnt pathway, which was associated with a delay in muscle differentiation. In addition, Klotho stimulation and Jmjd3 downregulation produced similar but not additive reductions in the expression of Wnt4, Wnt9a, and Wnt10a in myogenic cells, indicating that inhibition occurred through a common pathway. Together, our results identify a novel pathway through which Klotho influences myogenesis by reducing the expression of Jmjd3, leading to reductions in the expression of Wnt genes and inhibition of canonical Wnt signaling.
Collapse
Affiliation(s)
- Cynthia M McKee
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA
| | - Douglas J Chapski
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Manuel Rosa-Garrido
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, USA
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Thomas M Vondriska
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA.,Departments of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - James G Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA.,Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA.,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| |
Collapse
|
43
|
CPEB1 directs muscle stem cell activation by reprogramming the translational landscape. Nat Commun 2022; 13:947. [PMID: 35177647 PMCID: PMC8854658 DOI: 10.1038/s41467-022-28612-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 01/14/2022] [Indexed: 11/08/2022] Open
Abstract
Skeletal muscle stem cells, also called Satellite Cells (SCs), are actively maintained in quiescence but can activate quickly upon extrinsic stimuli. However, the mechanisms of how quiescent SCs (QSCs) activate swiftly remain elusive. Here, using a whole mouse perfusion fixation approach to obtain bona fide QSCs, we identify massive proteomic changes during the quiescence-to-activation transition in pathways such as chromatin maintenance, metabolism, transcription, and translation. Discordant correlation of transcriptomic and proteomic changes reveals potential translational regulation upon SC activation. Importantly, we show Cytoplasmic Polyadenylation Element Binding protein 1 (CPEB1), post-transcriptionally affects protein translation during SC activation by binding to the 3' UTRs of different transcripts. We demonstrate phosphorylation-dependent CPEB1 promoted Myod1 protein synthesis by binding to the cytoplasmic polyadenylation elements (CPEs) within its 3' UTRs to regulate SC activation and muscle regeneration. Our study characterizes CPEB1 as a key regulator to reprogram the translational landscape directing SC activation and subsequent proliferation.
Collapse
|
44
|
Elashry MI, Kinde M, Klymiuk MC, Eldaey A, Wenisch S, Arnhold S. The effect of hypoxia on myogenic differentiation and multipotency of the skeletal muscle-derived stem cells in mice. Stem Cell Res Ther 2022; 13:56. [PMID: 35123554 PMCID: PMC8817503 DOI: 10.1186/s13287-022-02730-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/20/2022] [Indexed: 01/01/2023] Open
Abstract
Abstract
Background
Skeletal muscle-derived stem cells (SC) have become a promising approach for investigating myogenic differentiation and optimizing tissue regeneration. Muscle regeneration is performed by SC, a self-renewal cell population underlying the basal lamina of muscle fibers. Here, we examined the impact of hypoxia condition on the regenerative capacity of SC either in their native microenvironment or via isolation in a monolayer culture using ectopic differentiation inductions. Furthermore, the effect of low oxygen tension on myogenic differentiation protocols of the myoblasts cell line C2C12 was examined.
Methods
Hind limb muscles of wild type mice were processed for both SC/fiber isolation and myoblast extraction using magnetic beads. SC were induced for myogenic, adipogenic and osteogenic commitments under normoxic (21% O2) and hypoxic (3% O2) conditions. SC proliferation and differentiation were evaluated using histological staining, immunohistochemistry, morphometric analysis and RT-qPCR. The data were statistically analyzed using ANOVA.
Results
The data revealed enhanced SC proliferation and motility following differentiation induction after 48 h under hypoxia. Following myogenic induction, the number of undifferentiated cells positive for Pax7 were increased at 72 h under hypoxia. Hypoxia upregulated MyoD and downregulated Myogenin expression at day-7 post-myogenic induction. Hypoxia promoted both SC adipogenesis and osteogenesis under respective induction as shown by using Oil Red O and Alizarin Red S staining. The expression of adipogenic markers; peroxisome proliferator activated receptor gamma (PPARγ) and fatty acid-binding protein 4 (FABP4) were upregulated under hypoxia up to day 14 compared to normoxic condition. Enhanced osteogenic differentiation was detected under hypoxic condition via upregulation of osteocalcin and osteopontin expression up to day 14 as well as, increased calcium deposition at day 21. Hypoxia exposure increases the number of adipocytes and the size of fat vacuoles per adipocyte compared to normoxic culture. Combining the differentiation medium with dexamethasone under hypoxia improves the efficiency of the myogenic differentiation protocol of C2C12 by increasing the length of the myotubes.
Conclusions
Hypoxia exposure increases cell resources for clinical applications and promotes SC multipotency and thus beneficial for tissue regeneration.
Collapse
|
45
|
Kim JT, Roberts K, Dunlap G, Perry R, Washington T, Wolchok JC. Nandrolone supplementation does not improve functional recovery in an aged animal model of volumetric muscle loss injury. J Tissue Eng Regen Med 2022; 16:367-379. [PMID: 35113494 DOI: 10.1002/term.3286] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/12/2022] [Accepted: 01/20/2022] [Indexed: 11/12/2022]
Abstract
Aging hinders the effectiveness of regenerative medicine strategies targeting the repair of volumetric muscle loss (VML) injury. Anabolic steroids have been shown to improve several factors which contribute to the age-related decline in muscle's regenerative capacity. In this study, the impact of exogenous nandrolone decanoate (ND) administration on the effectiveness of a VML regenerative repair strategy was explored using an aged animal model. Unilateral tibialis anterior VML injuries were repaired in 18-month-aged animal models (male Fischer 344 rat) using decellularized human skeletal muscle scaffolds supplemented with autologous minced muscle. The contralateral limb was left untreated/uninjured. Following repair, ND(+) or a carrier control (ND-) was delivered via weekly injection for a period of 8 weeks. At 8 weeks, muscle isometric torque, gene expression, and tissue structure were assessed. ND(+) treatment did not improve contractile torque recovery following VML repair when compared to carrier only ND(-) injection controls. Peak isometric torque in the ND(+) VML repair group remained significantly below contralateral uninjured control values (4.69 ± 1.18vs. 7.46 ± 1.53 N mm/kg) and was statistically indistinguishable from carrier only ND(-) VML repair controls (4.47 ± 1.18 N mm/kg). Gene expression for key myogenic genes (Pax7, MyoD, MyoG, IGF-1) were not significantly elevated in response to ND injection, suggesting continued age related myogenic impairment even in the presence of ND(+) treatment. ND injection did reduce the histological appearance of fibrosis at the site of VML repair, and increased expression of the collagen III gene, suggesting some positive effects on repair site matrix regulation. Overall, the results presented in this study suggest that a decline in regenerative capacity with aging may present an obstacle to regenerative medicine strategies targeting VML injury and that the delivery of anabolic stimuli via ND administration was unable to overcome this decline.
Collapse
Affiliation(s)
- John T Kim
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Kevin Roberts
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Grady Dunlap
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| | - Richard Perry
- Department of Health, Human Performance, and Recreation, College of Education and Health Professions, University of Arkansas, Fayetteville, Arkansas, USA
| | - Tyrone Washington
- Department of Health, Human Performance, and Recreation, College of Education and Health Professions, University of Arkansas, Fayetteville, Arkansas, USA
| | - Jeffrey C Wolchok
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
46
|
Angleri V, Damas F, Phillips SM, Selistre-de-Araujo HS, Cornachione AS, Stotzer US, Santanielo N, Soligon SD, Costa LAR, Lixandrão ME, Conceição MS, Cassaro Vechin F, Ugrinowitsch C, Libardi CA. Resistance training variable manipulations is less relevant than intrinsic biology in affecting muscle fiber hypertrophy. Scand J Med Sci Sports 2022; 32:821-832. [PMID: 35092084 DOI: 10.1111/sms.14134] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/21/2021] [Accepted: 01/22/2022] [Indexed: 11/30/2022]
Abstract
We aimed to investigate whether muscle fiber cross-sectional area (fCSA) and associated molecular processes could be differently affected at the group and individual level by manipulating resistance training (RT) variables. Twenty resistance-trained subjects had each leg randomly allocated to either a standard RT (RT-CON: without specific variables manipulations) or a variable RT (RT-VAR: manipulation of load, volume, muscle action, and rest interval at each RT session). Muscle fCSA, satellite cell (SC) pool, myonuclei content, and gene expression were assessed before and after training (chronic effect). Gene expression was assessed 24h after the last training session (acute effect). RT-CON and RT-VAR increased fCSA and myonuclei domain in type I and II fibers after training (P < 0.05). SC and myonuclei content did not change for both conditions (P > 0.05). Pax-7, MyoD, MMP-2 and COL3A1 (chronic) and MGF, Pax-7, and MMP-9 (acute) increased similar for RT-CON and RT-VAR (P < 0.05). The increase in acute MyoG expression was significantly higher for the RT-VAR than RT-CON (P < 0.05). Significant correlation between RT-CON and RT-VAR for the fCSA changes (r = 0.89). fCSA changes were also correlated to satellite cells (r = 0.42) and myonuclei (r = 0.50) changes. Heatmap analyses showed coupled changes in fCSA, SC, and myonuclei responses at the individual level, regardless of the RT protocol. The high between and low within-subject variability regardless of RT protocol suggests that the intrinsic biological factors seem to be more important to explain the magnitude of fCSA gains in resistance-trained subjects.
Collapse
Affiliation(s)
- Vitor Angleri
- MUSCULAB - Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Felipe Damas
- MUSCULAB - Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Heloisa Sobreiro Selistre-de-Araujo
- LBBM - Laboratory of Biochemistry and Molecular Biology, Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Anabelle Silva Cornachione
- Muscle Physiology and Biophysics Laboratory, Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Uliana Sbeguen Stotzer
- LBBM - Laboratory of Biochemistry and Molecular Biology, Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Natalia Santanielo
- MUSCULAB - Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | - Samuel Domingos Soligon
- MUSCULAB - Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| | | | | | | | | | - Carlos Ugrinowitsch
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Cleiton Augusto Libardi
- MUSCULAB - Laboratory of Neuromuscular Adaptations to Resistance Training, Department of Physical Education, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
47
|
Yue F, Oprescu SN, Qiu J, Gu L, Zhang L, Chen J, Narayanan N, Deng M, Kuang S. Lipid droplet dynamics regulate adult muscle stem cell fate. Cell Rep 2022; 38:110267. [PMID: 35045287 PMCID: PMC9127130 DOI: 10.1016/j.celrep.2021.110267] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/31/2021] [Accepted: 12/21/2021] [Indexed: 11/03/2022] Open
Abstract
The lipid droplet (LD) is a central hub for fatty acid metabolism in cells. Here we define the dynamics and explore the role of LDs in skeletal muscle satellite cells (SCs), a stem cell population responsible for muscle regeneration. In newly divided SCs, LDs are unequally distributed in sister cells exhibiting asymmetric cell fates, as the LDLow cell self-renews while the LDHigh cell commits to differentiation. When transplanted into regenerating muscles, LDLow cells outperform LDHigh cells in self-renewal and regeneration in vivo. Pharmacological inhibition of LD biogenesis or genetic inhibition of LD catabolism through knockout of Pnpla2 (encoding ATGL, the rate-limiting enzyme for lipolysis) disrupts cell fate homeostasis and impairs the regenerative capacity of SCs. Dysfunction of Pnpla2-null SCs is associated with energy insufficiency and oxidative stress that can be partially rescued by antioxidant (N-acetylcysteine) treatment. These results establish a direct link between LD dynamics and stem cell fate determination.
Collapse
Affiliation(s)
- Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Stephanie N Oprescu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Lijie Gu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Lijia Zhang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jingjuan Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Naagarajan Narayanan
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Meng Deng
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
48
|
Jacques E, Kuang Y, Kann AP, Le Grand F, Krauss RS, Gilbert PM. The mini-IDLE 3D biomimetic culture assay enables interrogation of mechanisms governing muscle stem cell quiescence and niche repopulation. eLife 2022; 11:81738. [PMID: 36537758 PMCID: PMC9904761 DOI: 10.7554/elife.81738] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Adult skeletal muscle harbours a population of muscle stem cells (MuSCs) that are required for repair after tissue injury. In youth, MuSCs return to a reversible state of cell-cycle arrest termed 'quiescence' after injury resolution. Conversely, some MuSCs in aged muscle remain semi-activated, causing a premature response to injuries that results in incomplete repair and eventual stem cell depletion. Regulating this balance between MuSC quiescence and activation may hold the key to restoring tissue homeostasis with age, but is incompletely understood. To fill this gap, we developed a simple and tractable in vitro method, to rapidly inactivate MuSCs freshly isolated from young murine skeletal muscle, and return them to a quiescent-like state for at least 1-week, which we name mini-IDLE (Inactivation and Dormancy LEveraged in vitro). This was achieved by introducing MuSCs into a 3D bioartificial niche comprised of a thin sheet of mouse myotubes, which we demonstrate provides the minimal cues necessary to induce quiescence. With different starting numbers of MuSCs, the assay revealed cellular heterogeneity and population-level adaptations that converged on a common niche repopulation density; behaviours previously observed only in vivo. Quiescence-associated hallmarks included a Pax7+CalcR+DDX6+MyoD-c-FOS- signature, quiescent-like morphologies, and polarized niche markers. Leveraging high-content bioimaging pipelines, we demonstrate a relationship between morphology and cell fate signatures for possible real-time morphology-based screening. When using MuSCs from aged muscle, they displayed aberrant proliferative activities and delayed inactivation kinetics, among other quiescence-associated defects that we show are partially rescued by wortmannin treatment. Thus, the assay offers an unprecedented opportunity to systematically investigate long-standing queries in areas such as regulation of pool size and functional heterogeneity within the MuSC population, and to uncover quiescence regulators in youth and age.
Collapse
Affiliation(s)
- Erik Jacques
- Institute of Biomedical Engineering, University of TorontoTorontoCanada,Donnelly Centre, University of TorontoTorontoCanada
| | - Yinni Kuang
- Donnelly Centre, University of TorontoTorontoCanada,Department of Cell and Systems Biology, University of TorontoTorontoCanada
| | - Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Black Family Stem Cell Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Fabien Le Grand
- Université Claude Bernard Lyon 1, CNRS UMR 5261, INSERM U1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and MuscleLyonFrance
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Black Family Stem Cell Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of TorontoTorontoCanada,Donnelly Centre, University of TorontoTorontoCanada,Department of Cell and Systems Biology, University of TorontoTorontoCanada
| |
Collapse
|
49
|
Ahmad K, Lim JH, Lee EJ, Chun HJ, Ali S, Ahmad SS, Shaikh S, Choi I. Extracellular Matrix and the Production of Cultured Meat. Foods 2021; 10:foods10123116. [PMID: 34945667 PMCID: PMC8700801 DOI: 10.3390/foods10123116] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/05/2021] [Accepted: 12/13/2021] [Indexed: 12/28/2022] Open
Abstract
Cultured meat production is an evolving method of producing animal meat using tissue engineering techniques. Cells, chemical factors, and suitable biomaterials that serve as scaffolds are all essential for the cultivation of muscle tissue. Scaffolding is essential for the development of organized meat products resembling steaks because it provides the mechanical stability needed by cells to attach, differentiate, and mature. In in vivo settings, extracellular matrix (ECM) ensures substrates and scaffolds are provided for cells. The ECM of skeletal muscle (SM) maintains tissue elasticity, creates adhesion points for cells, provides a three-dimensional (3D) environment, and regulates biological processes. Consequently, creating mimics of native ECM is a difficult task. Animal-derived polymers like collagen are often regarded as the gold standard for producing scaffolds with ECM-like properties. Animal-free scaffolds are being investigated as a potential source of stable, chemically defined, low-cost materials for cultured meat production. In this review, we explore the influence of ECM on myogenesis and its role as a scaffold and vital component to improve the efficacy of the culture media used to produce cultured meat.
Collapse
Affiliation(s)
- Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (K.A.); (J.-H.L.); (E.-J.L.); (H.-J.C.); (S.A.); (S.S.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Jeong-Ho Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (K.A.); (J.-H.L.); (E.-J.L.); (H.-J.C.); (S.A.); (S.S.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Eun-Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (K.A.); (J.-H.L.); (E.-J.L.); (H.-J.C.); (S.A.); (S.S.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Hee-Jin Chun
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (K.A.); (J.-H.L.); (E.-J.L.); (H.-J.C.); (S.A.); (S.S.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Shahid Ali
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (K.A.); (J.-H.L.); (E.-J.L.); (H.-J.C.); (S.A.); (S.S.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Syed Sayeed Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (K.A.); (J.-H.L.); (E.-J.L.); (H.-J.C.); (S.A.); (S.S.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Sibhghatulla Shaikh
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (K.A.); (J.-H.L.); (E.-J.L.); (H.-J.C.); (S.A.); (S.S.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Korea; (K.A.); (J.-H.L.); (E.-J.L.); (H.-J.C.); (S.A.); (S.S.A.); (S.S.)
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea
- Correspondence:
| |
Collapse
|
50
|
Di X, Han W, Zhang R, Liu C, Zheng Y. C-reactive Protein, Free Fatty Acid, and Uric Acid as Predictors of Adverse Events after Endovascular Revascularization of Arterial Femoropopliteal Occlusion Lesions. Ann Vasc Surg 2021; 81:333-342. [PMID: 34775024 DOI: 10.1016/j.avsg.2021.09.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES This study aimed to investigate the relationship between pre-procedure high-sensitivity C-reactive protein (hsCRP), free fatty acid (FFA), and uric acid (UA) levels and post-procedure mortality and morbidity of endovascular revascularization of arterial femoropopliteal occlusion lesions. METHODS This was a retrospective review of clinical data retrieved from a prospectively held database in Peking Union Medical College Hospital. A total of 71 Patients who underwent endovascular treatment (EVT) for femoropopliteal occlusive disease between January 1, 2014 and November 1, 2017, were included in this study. Endpoints were defined as major adverse limb events (MALE; target vessel revascularization, amputation, or disease progression) and major adverse cardiovascular events (MACE; stroke, myocardial infarction, or all-cause death) during the entire follow-up period. Univariate and multivariate Cox proportional hazards regression models were used to evaluate the relationship of elevated biomarker levels (hsCRP, FFA and UA, measured by immunoturbidimetry assay, enzymatic assay and enzymatic assay, respectively) to MALE and MACE outcomes. RESULTS Seventy-one patients (72 limbs) with sufficient follow-up information were included in the analysis. The mean age was 69.7 ± 8.6 years; 21.1% were female. The Rutherford class of target limbs were ≥ 3. The median follow-up was 36 (range 18-59 months). Univariate analyses revealed that patients with elevated hsCRP levels had an increased risk of MALE (hazard ratio [HR], 2.682; 95% confidence interval [CI], 1.281-5.617, P = 0.009). High FFA levels were associated with an increased risk of MALE (HR, 2.658; 95% CI, 1.075-6.573; P = 0.034). Multivariate analyses demonstrated that elevated hsCRP values (HR, 4.015; 95% CI, 1.628-10.551; P = 0.003) and FFA value (HR, 3.034; 95% CI, 1.102-8.354; P = 0.032) were both significantly associated with increased MALE. Elevated UA levels predicted MACE in the presence of confounders (HR, 11.446; 95% CI, 1.367-95.801 P = 0.023). CONCLUSION Pre-procedure hsCRP and FFA levels could serve as predictors of adverse events after EVT in patients with arterial femoropopliteal occlusive disease. The role of UA in MACE may warrant further investigation, because the correlation is not as powerful as the other two in the study.
Collapse
Affiliation(s)
- Xiao Di
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei Han
- Department of Statistics, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Rui Zhang
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Changwei Liu
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | - Yuehong Zheng
- Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|