1
|
Seco-Cervera M, Ibáñez-Cabellos JS, Pallardo FV, García-Giménez JL, Aulinas A, Martel-Duguech L, Webb SM, Valassi E. Circulating miR-28-5p is overexpressed in patients with sarcopenia despite long-term remission of Cushing's syndrome: a pilot study. Front Endocrinol (Lausanne) 2024; 15:1410080. [PMID: 39086897 PMCID: PMC11289718 DOI: 10.3389/fendo.2024.1410080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/21/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction Patients with Cushing's syndrome (CS) in remission show sustained fatigue, myopathy, and an increased prevalence of sarcopenia. The mechanisms that determine these persistent muscle problems are not well known. We aimed to identify circulating microRNAs (miRNAs) with differential expression that could be potential biomarkers for the diagnosis and/or prognosis in CS. Patients and methods Thirty-six women in sustained remission for 13 ± 7 years (mean ± SD) from CS, with a median age (IQ range) of 51 (45.2-60) years and mean ± SD BMI of 27 ± 4 Kg/m2, and 36 matched healthy controls were investigated. In 7 patients sarcopenia was present according to the European Working Group on Sarcopenia in Older People (EWGSOP) criteria. Small RNA libraries were generated and indexed using a modified Illumina TruSeq small RNA-sequencing protocol. MiRNAs were identified in plasma using bioinformatic analysis, and validation was carried out using RT-qPCR. For the validation, Taqman probes were performed on QuantStudio 5 equipment (Applied Biosystems). Results In a first discovery group using RNA-sequencing, plasma samples of 18 CS patients and 18 healthy subjects were investigated; circulating miR-28-5p, miR-495-3p and miR-654-5p were upregulated in CS patients as compared with controls (p<0.05). In a validation study of the 3 upregulated miRNAs in 36 patients and 26 controls, no differences were observed by RT-qPCR; however, the expression of circulating miR-28-5p was upregulated in CS patients with sarcopenia as compared with those without (AUC for fold-change in the ROC analysis, 0.798; p=0.0156). The optimized cut-off value for miR-28-5p to identify CS patients with sarcopenia was 3.80, which yielded a sensitivity of 86% and a specificity of 69%. Conclusion MiR-28-5p, a muscle-specific microRNA involved in myotube proliferation and differentiation in vivo, may serve as an independent non-invasive biomarker for identifying CS patients at high-risk of sarcopenia despite biochemical remission.
Collapse
Affiliation(s)
- Marta Seco-Cervera
- Unit 733, Centre for Biomedical Network Research on Rare Diseases [CIBERER- Instituto de Salud Carlos III (ISCIII)], Madrid, Spain
- Mixed Unit for rare diseases INCLIVA-CIPF, INCLIVA Health Research Institute, Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | | | - Federico V. Pallardo
- Unit 733, Centre for Biomedical Network Research on Rare Diseases [CIBERER- Instituto de Salud Carlos III (ISCIII)], Madrid, Spain
- Mixed Unit for rare diseases INCLIVA-CIPF, INCLIVA Health Research Institute, Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - José-Luis García-Giménez
- Unit 733, Centre for Biomedical Network Research on Rare Diseases [CIBERER- Instituto de Salud Carlos III (ISCIII)], Madrid, Spain
- Mixed Unit for rare diseases INCLIVA-CIPF, INCLIVA Health Research Institute, Valencia, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Anna Aulinas
- Department of Endocrinology, Hospital S Pau, Research Center for Pituitary Diseases, Institut de Recerca Sant Pau (IIB-Sant Pau), Barcelona, Spain
- CIBERER Unit 747, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medicine, Universitat de Vic-Universitat Central de Catalunya, Vic, Spain
| | - Luciana Martel-Duguech
- Department of Endocrinology, Hospital S Pau, Research Center for Pituitary Diseases, Institut de Recerca Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Susan M. Webb
- Department of Endocrinology, Hospital S Pau, Research Center for Pituitary Diseases, Institut de Recerca Sant Pau (IIB-Sant Pau), Barcelona, Spain
- CIBERER Unit 747, Instituto de Salud Carlos III, Madrid, Spain
- Department of Medicine, Univ Autonoma Barcelona, Bellaterra, Spain
| | - Elena Valassi
- CIBERER Unit 747, Instituto de Salud Carlos III, Madrid, Spain
- Endocrinology and Nutrition Department, Germans Trias i Pujol Hospital and Research Institute, Badalona, Spain
- School of Medicine, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Barcelona, Spain
| |
Collapse
|
2
|
Luo W, Zhang H, Wan R, Cai Y, Liu Y, Wu Y, Yang Y, Chen J, Zhang D, Luo Z, Shang X. Biomaterials-Based Technologies in Skeletal Muscle Tissue Engineering. Adv Healthc Mater 2024; 13:e2304196. [PMID: 38712598 DOI: 10.1002/adhm.202304196] [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: 11/28/2023] [Revised: 04/26/2024] [Indexed: 05/08/2024]
Abstract
For many clinically prevalent severe injuries, the inherent regenerative capacity of skeletal muscle remains inadequate. Skeletal muscle tissue engineering (SMTE) seeks to meet this clinical demand. With continuous progress in biomedicine and related technologies including micro/nanotechnology and 3D printing, numerous studies have uncovered various intrinsic mechanisms regulating skeletal muscle regeneration and developed tailored biomaterial systems based on these understandings. Here, the skeletal muscle structure and regeneration process are discussed and the diverse biomaterial systems derived from various technologies are explored in detail. Biomaterials serve not merely as local niches for cell growth, but also as scaffolds endowed with structural or physicochemical properties that provide tissue regenerative cues such as topographical, electrical, and mechanical signals. They can also act as delivery systems for stem cells and bioactive molecules that have been shown as key participants in endogenous repair cascades. To achieve bench-to-bedside translation, the typical effect enabled by biomaterial systems and the potential underlying molecular mechanisms are also summarized. Insights into the roles of biomaterials in SMTE from cellular and molecular perspectives are provided. Finally, perspectives on the advancement of SMTE are provided, for which gene therapy, exosomes, and hybrid biomaterials may hold promise to make important contributions.
Collapse
Affiliation(s)
- Wei Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Hanli Zhang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Renwen Wan
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yuxi Cai
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yinuo Liu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yang Wu
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yimeng Yang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jiani Chen
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, Hong Kong
| | - Zhiwen Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Xiliang Shang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| |
Collapse
|
3
|
Rauen KA, Tidyman WE. RASopathies - what they reveal about RAS/MAPK signaling in skeletal muscle development. Dis Model Mech 2024; 17:dmm050609. [PMID: 38847227 PMCID: PMC11179721 DOI: 10.1242/dmm.050609] [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] [Indexed: 06/12/2024] Open
Abstract
RASopathies are rare developmental genetic syndromes caused by germline pathogenic variants in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) signal transduction pathway. Although the incidence of each RASopathy syndrome is rare, collectively, they represent one of the largest groups of multiple congenital anomaly syndromes and have severe developmental consequences. Here, we review our understanding of how RAS/MAPK dysregulation in RASopathies impacts skeletal muscle development and the importance of RAS/MAPK pathway regulation for embryonic myogenesis. We also discuss the complex interactions of this pathway with other intracellular signaling pathways in the regulation of skeletal muscle development and growth, and the opportunities that RASopathy animal models provide for exploring the use of pathway inhibitors, typically used for cancer treatment, to correct the unique skeletal myopathy caused by the dysregulation of this pathway.
Collapse
Affiliation(s)
- Katherine A Rauen
- Department of Pediatrics, Division of Genomic Medicine, University of California Davis, Sacramento, CA, 95817, USA
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| | - William E Tidyman
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| |
Collapse
|
4
|
Endo T. Postnatal skeletal muscle myogenesis governed by signal transduction networks: MAPKs and PI3K-Akt control multiple steps. Biochem Biophys Res Commun 2023; 682:223-243. [PMID: 37826946 DOI: 10.1016/j.bbrc.2023.09.048] [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: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
Skeletal muscle myogenesis represents one of the most intensively and extensively examined systems of cell differentiation, tissue formation, and regeneration. Muscle regeneration provides an in vivo model system of postnatal myogenesis. It comprises multiple steps including muscle stem cell (or satellite cell) quiescence, activation, migration, myogenic determination, myoblast proliferation, myocyte differentiation, myofiber maturation, and hypertrophy. A variety of extracellular signaling and subsequent intracellular signal transduction pathways or networks govern the individual steps of postnatal myogenesis. Among them, MAPK pathways (the ERK, JNK, p38 MAPK, and ERK5 pathways) and PI3K-Akt signaling regulate multiple steps of myogenesis. Ca2+, cytokine, and Wnt signaling also participate in several myogenesis steps. These signaling pathways often control cell cycle regulatory proteins or the muscle-specific MyoD family and the MEF2 family of transcription factors. This article comprehensively reviews molecular mechanisms of the individual steps of postnatal skeletal muscle myogenesis by focusing on signal transduction pathways or networks. Nevertheless, no or only a partial signaling molecules or pathways have been identified in some responses during myogenesis. The elucidation of these unidentified signaling molecules and pathways leads to an extensive understanding of the molecular mechanisms of myogenesis.
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
5
|
Cheng C, Zhang S, Gong Y, Wang X, Tang S, Wan J, Ding K, Yuan C, Sun W, Yao LH. Cordycepin inhibits myogenesis via activating the ERK1/2 MAPK signalling pathway in C2C12 cells. Biomed Pharmacother 2023; 165:115163. [PMID: 37453196 DOI: 10.1016/j.biopha.2023.115163] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
Cordycepin (with a molecular formula of C10H13N5O3), a natural adenosine isolated from Cordyceps militaris, has an important regulatory effect on skeletal muscle remodelling and quality maintenance. The aim of this study was to investigate the effect of cordycepin on myoblast differentiation and explore the underlying molecular mechanisms of this effect. Our results showed that cordycepin inhibited myogenesis by downregulating myogenic differentiation (MyoD) and myogenin (MyoG), preserved undifferentiated reserve cell pools by upregulating myogenic factor 5 (Myf5) and retinoblastoma-like protein p130 (p130), and enhanced energy reserves by decreasing intracellular reactive oxygen species (ROS) and enhancing mitochondrial membrane potential, mitochondrial mass, and ATP content. The effect of cordycepin on myogenesis was associated with increased phosphorylation of extracellular signal-regulated kinase 1/2 (p-ERK1/2). PD98059 (a specific inhibitor of p-ERK1/2) attenuated the inhibitory effect of cordycepin on C2C12 differentiation. The present study reveals that cordycepin inhibits myogenesis through ERK1/2 MAPK signalling activation accompanied by an increase in skeletal muscle energy reserves and improving skeletal muscle oxidative stress, which may have implications for its further application for the prevention and treatment of degenerative muscle diseases caused by the depletion of depleted muscle stem cells.
Collapse
Affiliation(s)
- Chunfang Cheng
- School of Sport Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Shasha Zhang
- School of Sport Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Yanchun Gong
- School of Sport Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China; School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Xuanyu Wang
- School of Sport Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Shan Tang
- School of Sport Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Juan Wan
- School of Sport Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Kaizhi Ding
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Chunhua Yuan
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Wei Sun
- School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China
| | - Li-Hua Yao
- School of Sport Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China; School of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi 330013, PR China.
| |
Collapse
|
6
|
Tidyman WE, Goodwin AF, Maeda Y, Klein OD, Rauen KA. MEK-inhibitor-mediated rescue of skeletal myopathy caused by activating Hras mutation in a Costello syndrome mouse model. Dis Model Mech 2022; 15:272258. [PMID: 34553752 PMCID: PMC8617311 DOI: 10.1242/dmm.049166] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 09/13/2021] [Indexed: 11/20/2022] Open
Abstract
Costello syndrome (CS) is a congenital disorder caused by heterozygous activating germline HRAS mutations in the canonical Ras/mitogen-activated protein kinase (Ras/MAPK) pathway. CS is one of the RASopathies, a large group of syndromes caused by mutations within various components of the Ras/MAPK pathway. An important part of the phenotype that greatly impacts quality of life is hypotonia. To gain a better understanding of the mechanisms underlying hypotonia in CS, a mouse model with an activating HrasG12V allele was utilized. We identified a skeletal myopathy that was due, in part, to inhibition of embryonic myogenesis and myofiber formation, resulting in a reduction in myofiber size and number that led to reduced muscle mass and strength. In addition to hyperactivation of the Ras/MAPK and PI3K/AKT pathways, there was a significant reduction in p38 signaling, as well as global transcriptional alterations consistent with the myopathic phenotype. Inhibition of Ras/MAPK pathway signaling using a MEK inhibitor rescued the HrasG12V myopathy phenotype both in vitro and in vivo, demonstrating that increased MAPK signaling is the main cause of the muscle phenotype in CS. Summary: A Costello syndrome (CS) mouse model carrying a heterozygous Hras p.G12V mutation was utilized to investigate Ras pathway dysregulation, revealing that increased MAPK signaling is the main cause of the muscle phenotype in CS.
Collapse
Affiliation(s)
- William E Tidyman
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA.,UC Davis MIND Institute, Sacramento, CA 95817, USA
| | - Alice F Goodwin
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA 94143, USA
| | - Yoshiko Maeda
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA.,UC Davis MIND Institute, Sacramento, CA 95817, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA 94143, USA.,Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA 94143, USA
| | - Katherine A Rauen
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA.,UC Davis MIND Institute, Sacramento, CA 95817, USA
| |
Collapse
|
7
|
Sabouri M, Taghibeikzadehbadr P, Shabkhiz F, Izanloo Z, Shaghaghi FA. Effect of eccentric and concentric contraction mode on myogenic regulatory factors expression in human vastus lateralis muscle. J Muscle Res Cell Motil 2022; 43:9-20. [PMID: 35018575 DOI: 10.1007/s10974-021-09613-x] [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: 07/05/2021] [Accepted: 12/01/2021] [Indexed: 11/26/2022]
Abstract
Skeletal muscle contractions are caused to release myokines by muscle fiber. This study investigated the myogenic regulatory factors, as MHC I, IIA, IIX, Myo-D, MRF4, Murf, Atrogin-1, Decorin, Myonection, and IL-15 mRNA expression in the response of eccentric vs concentric contraction. Eighteen healthy men were randomly divided into two eccentric and concentric groups, each of 9 persons. Isokinetic contraction protocols included maximal single-leg eccentric or concentric knee extension tasks at 60°/s with the dominant leg. Contractions consisted of a maximum of 12 sets of 10 reps, and the rest time between each set was 30 s. The baseline biopsy was performed 4 weeks before the study, and post-test biopsies were taken immediately after exercise protocols from the vastus lateralis muscle. The gene expression levels were evaluated using Real-Time PCR methods. The eccentric group showed a significantly lower RPE score than the concentric group (P ≤ 0.05). A significant difference in MyoD, MRF4, Myonection, and Decorin mRNA, were observed following eccentric or concentric contractions (P ≤ 0.05). The MHC I, MHC IIA, IL-15 mRNA has been changed significantly compared to the pre-exercise in the concentric group (P ≤ 0.05). While only MHC IIX and Atrogin-1 mRNA changed significantly in the eccentric group (P ≤ 0.05). Additionally, the results showed a significant difference in MyoD, MRF4, IL-15, and Decorin at the follow-up values between eccentric or concentric groups (P ≤ 0.05). Our findings highlight the growing importance of elucidating the different responses of muscle growth factors associated with a myogenic activity such as MHC IIA, Decorin, IL-15, Myonectin, Decorin, MuRF1, and MHC IIX mRNA in following various types of exercise.
Collapse
Affiliation(s)
- Mostafa Sabouri
- Department of Exercise Physiology & Health Science, University of Tehran, Tehran, Iran.
| | | | - Fatemeh Shabkhiz
- Department of Exercise Physiology & Health Science, University of Tehran, Tehran, Iran
| | - Zahra Izanloo
- Department of Sport Science, Faculty of Human Science, University of Bojnord, Bojnord, Iran
| | | |
Collapse
|
8
|
Eugenis I, Wu D, Rando TA. Cells, scaffolds, and bioactive factors: Engineering strategies for improving regeneration following volumetric muscle loss. Biomaterials 2021; 278:121173. [PMID: 34619561 PMCID: PMC8556323 DOI: 10.1016/j.biomaterials.2021.121173] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/01/2021] [Accepted: 08/14/2021] [Indexed: 12/20/2022]
Abstract
Severe traumatic skeletal muscle injuries, such as volumetric muscle loss (VML), result in the obliteration of large amounts of skeletal muscle and lead to permanent functional impairment. Current clinical treatments are limited in their capacity to regenerate damaged muscle and restore tissue function, promoting the need for novel muscle regeneration strategies. Advances in tissue engineering, including cell therapy, scaffold design, and bioactive factor delivery, are promising solutions for VML therapy. Herein, we review tissue engineering strategies for regeneration of skeletal muscle, development of vasculature and nerve within the damaged muscle, and achievements in immunomodulation following VML. In addition, we discuss the limitations of current state of the art technologies and perspectives of tissue-engineered bioconstructs for muscle regeneration and functional recovery following VML.
Collapse
Affiliation(s)
- Ioannis Eugenis
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Di Wu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Center for Tissue Regeneration, Repair, and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
9
|
Masuzawa R, Takahashi K, Takano K, Nishino I, Sakai T, Endo T. DA-Raf and the MEK inhibitor trametinib reverse skeletal myocyte differentiation inhibition or muscle atrophy caused by myostatin and GDF11 through the non-Smad Ras-ERK pathway. J Biochem 2021; 171:109-122. [PMID: 34676394 DOI: 10.1093/jb/mvab116] [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: 05/26/2021] [Accepted: 10/18/2021] [Indexed: 12/25/2022] Open
Abstract
Myostatin (Mstn) and GDF11 are critical factors that are involved in muscle atrophy in the young and sarcopenia in the elderly, respectively. These TGF-β superfamily proteins activate not only Smad signaling but also non-Smad signaling including the Ras-mediated ERK pathway (Raf-MEK-ERK phosphorylation cascade). Although Mstn and GDF11 have been shown to induce muscle atrophy or sarcopenia by Smad2/3-mediated Akt inhibition, participation of the non-Smad Ras-ERK pathway in atrophy and sarcopenia has not been well determined. We show here that both Mstn and GDF11 prevented skeletal myocyte differentiation but that the MEK inhibitor U0126 or trametinib restored differentiation in Mstn- or GDF11-treated myocytes. These MEK inhibitors induced the expression of DA-Raf1 (DA-Raf), which is a dominant-negative antagonist of the Ras-ERK pathway. Exogenous expression of DA-Raf in Mstn- or GDF11-treated myocytes restored differentiation. Furthermore, administration of trametinib to aged mice resulted in an increase in myofiber size, or recovery from muscle atrophy. The trametinib administration downregulated ERK activity in these muscles. These results imply that the Mstn/GDF11-induced Ras-ERK pathway plays critical roles in the inhibition of myocyte differentiation and muscle regeneration, which leads to muscle atrophy. Trametinib and similar approved drugs might be applicable to the treatment of muscle atrophy in sarcopenia or cachexia.
Collapse
Affiliation(s)
- Ryuichi Masuzawa
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan
| | - Kazuya Takahashi
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan
| | - Kazunori Takano
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo 187-8502, Japan
| | - Toshiyuki Sakai
- Drug Discovery Center and Department of Drug Discovery Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan
| |
Collapse
|
10
|
Cai X, Yang S, Peng Y, Huang Y, Chen H, Wu X. Screening of key genes during early embryonic development of Nile tilapia (Oreochromis niloticus). GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
11
|
Myogenic Differentiation of iPS Cells Shows Different Efficiency in Simultaneous Comparison of Protocols. Cells 2021; 10:cells10071671. [PMID: 34359837 PMCID: PMC8307201 DOI: 10.3390/cells10071671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/17/2021] [Accepted: 06/28/2021] [Indexed: 11/17/2022] Open
Abstract
Induced pluripotent stem (iPS) cells constitute a perfect tool to study human embryo development processes such as myogenesis, thanks to their ability to differentiate into three germ layers. Currently, many protocols to obtain myogenic cells have been described in the literature. They differ in many aspects, such as media components, including signaling modulators, feeder layer constituents, and duration of culture. In our study, we compared three different myogenic differentiation protocols to verify, side by side, their efficiency. Protocol I was based on embryonic bodies differentiation induction, ITS addition, and selection with adhesion to collagen I type. Protocol II was based on strong myogenic induction at the embryonic bodies step with BIO, forskolin, and bFGF, whereas cells in Protocol III were cultured in monolayers in three special media, leading to WNT activation and TGF-β and BMP signaling inhibition. Myogenic induction was confirmed by the hierarchical expression of myogenic regulatory factors MYF5, MYOD, MYF6 and MYOG, as well as the expression of myotubes markers MYH3 and MYH2, in each protocol. Our results revealed that Protocol III is the most efficient in obtaining myogenic cells. Furthermore, our results indicated that CD56 is not a specific marker for the evaluation of myogenic differentiation.
Collapse
|
12
|
Maeda Y, Tidyman WE, Ander BP, Pritchard CA, Rauen KA. Ras/MAPK dysregulation in development causes a skeletal myopathy in an activating Braf L597V mouse model for cardio-facio-cutaneous syndrome. Dev Dyn 2021; 250:1074-1095. [PMID: 33522658 DOI: 10.1002/dvdy.309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/03/2021] [Accepted: 01/19/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cardio-facio-cutaneous (CFC) syndrome is a human multiple congenital anomaly syndrome that is caused by activating heterozygous mutations in either BRAF, MEK1, or MEK2, three protein kinases of the Ras/mitogen-activated protein kinase (MAPK) pathway. CFC belongs to a group of syndromes known as RASopathies. Skeletal muscle hypotonia is a ubiquitous phenotype of RASopathies, especially in CFC syndrome. To better understand the underlying mechanisms for the skeletal myopathy in CFC, a mouse model with an activating BrafL597V allele was utilized. RESULTS The activating BrafL597V allele resulted in phenotypic alterations in skeletal muscle characterized by a reduction in fiber size which leads to a reduction in muscle size which are functionally weaker. MAPK pathway activation caused inhibition of myofiber differentiation during embryonic myogenesis and global transcriptional dysregulation of developmental pathways. Inhibition in differentiation can be rescued by MEK inhibition. CONCLUSIONS A skeletal myopathy was identified in the CFC BrafL597V mouse validating the use of models to study the effect of Ras/MAPK dysregulation on skeletal myogenesis. RASopathies present a novel opportunity to identify new paradigms of myogenesis and further our understanding of Ras in development. Rescue of the phenotype by inhibitors may help advance the development of therapeutic options for RASopathy patients.
Collapse
Affiliation(s)
- Yoshiko Maeda
- Department of Pediatrics, University of California Davis, Sacramento, California, USA.,UC Davis MIND Institute, Sacramento, California, USA
| | - William E Tidyman
- Department of Pediatrics, University of California Davis, Sacramento, California, USA.,UC Davis MIND Institute, Sacramento, California, USA
| | - Bradley P Ander
- UC Davis MIND Institute, Sacramento, California, USA.,Department of Neurology, University of California Davis, Sacramento, California, USA
| | - Catrin A Pritchard
- Leicester Cancer Research Centre, University of Leicester, Leicester, United Kingdom
| | - Katherine A Rauen
- Department of Pediatrics, University of California Davis, Sacramento, California, USA.,UC Davis MIND Institute, Sacramento, California, USA
| |
Collapse
|
13
|
Yang Y, Zhu X, Jia X, Hou W, Zhou G, Ma Z, Yu B, Pi Y, Zhang X, Wang J, Wang G. Phosphorylation of Msx1 promotes cell proliferation through the Fgf9/18-MAPK signaling pathway during embryonic limb development. Nucleic Acids Res 2020; 48:11452-11467. [PMID: 33080014 PMCID: PMC7672426 DOI: 10.1093/nar/gkaa905] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/26/2020] [Accepted: 10/08/2020] [Indexed: 11/25/2022] Open
Abstract
Msh homeobox (Msx) is a subclass of homeobox transcriptional regulators that control cell lineage development, including the early stage of vertebrate limb development, although the underlying mechanisms are not clear. Here, we demonstrate that Msx1 promotes the proliferation of myoblasts and mesenchymal stem cells (MSCs) by enhancing mitogen-activated protein kinase (MAPK) signaling. Msx1 directly binds to and upregulates the expression of fibroblast growth factor 9 (Fgf9) and Fgf18. Accordingly, knockdown or antibody neutralization of Fgf9/18 inhibits Msx1-activated extracellular signal-regulated kinase 1/2 (Erk1/2) phosphorylation. Mechanistically, we determined that the phosphorylation of Msx1 at Ser136 is critical for enhancing Fgf9 and Fgf18 expression and cell proliferation, and cyclin-dependent kinase 1 (CDK1) is apparently responsible for Ser136 phosphorylation. Furthermore, mesenchymal deletion of Msx1/2 results in decreased Fgf9 and Fgf18 expression and Erk1/2 phosphorylation, which leads to serious defects in limb development in mice. Collectively, our findings established an important function of the Msx1-Fgf-MAPK signaling axis in promoting cell proliferation, thus providing a new mechanistic insight into limb development.
Collapse
Affiliation(s)
- Yenan Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Xiaoli Zhu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui 230001, China
| | - Xiang Jia
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Wanwan Hou
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Guoqiang Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Zhangjing Ma
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Bin Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yan Pi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Xumin Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Jingqiang Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Gang Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Zhongshan Hospital, Fudan University, Shanghai 200438, China
| |
Collapse
|
14
|
Skeletal Muscle and the Effects of Ammonia Toxicity in Fish, Mammalian, and Avian Species: A Comparative Review Based on Molecular Research. Int J Mol Sci 2020; 21:ijms21134641. [PMID: 32629824 PMCID: PMC7370143 DOI: 10.3390/ijms21134641] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022] Open
Abstract
Typically, mammalian and avian models have been used to examine the effects of ammonia on skeletal muscle. Hyperammonemia causes sarcopenia or muscle wasting, in mammals and has been linked to sarcopenia in liver disease patients. Avian models of skeletal muscle have responded positively to hyperammonemia, differing from the mammalian response. Fish skeletal muscle has not been examined as extensively as mammalian and avian muscle. Fish skeletal muscle shares similarities with avian and mammalian muscle but has notable differences in growth, fiber distribution, and response to the environment. The wide array of body sizes and locomotion needs of fish also leads to greater diversity in muscle fiber distribution and growth between different fish species. The response of fish muscle to high levels of ammonia is important for aquaculture and quality food production but has not been extensively studied to date. Understanding the differences between fish, mammalian and avian species’ myogenic response to hyperammonemia could lead to new therapies for muscle wasting due to a greater understanding of the mechanisms behind skeletal muscle regulation and how ammonia effects these mechanisms. This paper provides an overview of fish skeletal muscle and ammonia excretion and toxicity in fish, as well as a comparison to avian and mammalian species.
Collapse
|
15
|
Wierzbicki M, Hotowy A, Kutwin M, Jaworski S, Bałaban J, Sosnowska M, Wójcik B, Wędzińska A, Chwalibog A, Sawosz E. Graphene Oxide Scaffold Stimulates Differentiation and Proangiogenic Activities of Myogenic Progenitor Cells. Int J Mol Sci 2020; 21:ijms21114173. [PMID: 32545308 PMCID: PMC7311992 DOI: 10.3390/ijms21114173] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 05/26/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
The physiological process of muscle regeneration is quite limited due to low satellite cell quantity and also the inability to regenerate and reconstruct niche tissue. The purpose of the study was to examine whether a graphene oxide scaffold is able to stimulate myogenic progenitor cell proliferation and the endocrine functions of differentiating cells, and therefore, their active participation in the construction of muscle tissue. Studies were carried out using mesenchymal cells taken from 6-day-old chicken embryos and human umbilical vein endothelial cells (HUVEC) were used to assess angiogenesis. The graphene scaffold was readily colonized by myogenic progenitor cells and the cells dissected from heart, brain, eye, and blood vessels did not avoid the scaffold. The scaffold strongly induced myogenic progenitor cell signaling pathways and simultaneously activated proangiogenic signaling pathways via exocrine vascular endothelial growth factor (VEGF) secretion. The present study revealed that the graphene oxide (GO) scaffold initiates the processes of muscle cell differentiation due to mechanical interaction with myogenic progenitor cell.
Collapse
Affiliation(s)
- Mateusz Wierzbicki
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.H.); (M.K.); (S.J.); (J.B.); (M.S.); (B.W.); (A.W.); (E.S.)
- Correspondence:
| | - Anna Hotowy
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.H.); (M.K.); (S.J.); (J.B.); (M.S.); (B.W.); (A.W.); (E.S.)
| | - Marta Kutwin
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.H.); (M.K.); (S.J.); (J.B.); (M.S.); (B.W.); (A.W.); (E.S.)
| | - Sławomir Jaworski
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.H.); (M.K.); (S.J.); (J.B.); (M.S.); (B.W.); (A.W.); (E.S.)
| | - Jaśmina Bałaban
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.H.); (M.K.); (S.J.); (J.B.); (M.S.); (B.W.); (A.W.); (E.S.)
| | - Malwina Sosnowska
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.H.); (M.K.); (S.J.); (J.B.); (M.S.); (B.W.); (A.W.); (E.S.)
| | - Barbara Wójcik
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.H.); (M.K.); (S.J.); (J.B.); (M.S.); (B.W.); (A.W.); (E.S.)
| | - Aleksandra Wędzińska
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.H.); (M.K.); (S.J.); (J.B.); (M.S.); (B.W.); (A.W.); (E.S.)
| | - André Chwalibog
- Department of Veterinary and Animal Sciences, University of Copenhagen, Groennegaardsvej 3, 1870 Frederiksberg, Denmark;
| | - Ewa Sawosz
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland; (A.H.); (M.K.); (S.J.); (J.B.); (M.S.); (B.W.); (A.W.); (E.S.)
| |
Collapse
|
16
|
Endo T. Dominant-negative antagonists of the Ras-ERK pathway: DA-Raf and its related proteins generated by alternative splicing of Raf. Exp Cell Res 2019; 387:111775. [PMID: 31843497 DOI: 10.1016/j.yexcr.2019.111775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022]
Abstract
The Ras-ERK pathway regulates a variety of cellular and physiological responses, including cell proliferation, differentiation, morphogenesis during animal development, and homeostasis in adults. Deregulated activation of this pathway leads to cellular transformation and tumorigenesis as well as RASopathies. Several negative regulators of this pathway have been documented. Each of these proteins acts at particular points of the pathway, and they exert specific cellular and physiological functions. Among them, DA-Raf1 (DA-Raf), which is a splicing isoform of A-Raf and contains the Ras-binding domain but lacks the kinase domain, antagonizes the Ras-ERK pathway in a dominant-negative manner. DA-Raf induces apoptosis, skeletal myocyte differentiation, lung alveolarization, and fulfills tumor suppressor functions by interfering with the Ras-ERK pathway. After the findings of DA-Raf, several kinase-domain-truncated splicing variants of Raf proteins have also been reported. The family of these truncated proteins represents the concept that alternative splicing can generate antagonistic proteins to their full-length counterparts.
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Biology, Graduate School of Science, Chiba University, 1-33 Yayoicho, Inageku, Chiba, Chiba 263-8522, Japan.
| |
Collapse
|
17
|
Soundharrajan I, Kim DH, Kuppusamy P, Choi KC. Modulation of osteogenic and myogenic differentiation by a phytoestrogen formononetin via p38MAPK-dependent JAK-STAT and Smad-1/5/8 signaling pathways in mouse myogenic progenitor cells. Sci Rep 2019; 9:9307. [PMID: 31243298 PMCID: PMC6594940 DOI: 10.1038/s41598-019-45793-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 06/12/2019] [Indexed: 11/09/2022] Open
Abstract
Formononetin (FN), a typical phytoestrogen has attracted substantial attention as a novel agent because of its diverse biological activities including, osteogenic differentiation. However, the molecular mechanisms underlying osteogenic and myogenic differentiation by FN in C2C12 progenitor cells remain unknown. Therefore the objective of the current study was to investigate the action of FN on myogenic and osteogenic differentiation and its impact on signaling pathways in C2C12 cells. FN significantly increased myogenic markers such as Myogenin, myosin heavy chains, and myogenic differentiation 1 (MyoD). In addition, the expression of osteogenic specific genes alkaline phosphatase (ALP), Run-related transcription factor 2(RUNX2), and osteocalcin (OCN) were up-regulated by FN treatment. Moreover, FN enhanced the ALP level, calcium deposition and the expression of bone morphogenetic protein isoform (BMPs). Signal transduction pathways mediated by p38 mitogen-activated protein kinase (p38MAPK), extracellular signal-related kinases (ERKs), protein kinase B (Akt), Janus kinases (JAKs), and signal transducer activator of transcription proteins (STATs) in myogenic and osteogenic differentiation after FN treatment were also examined. FN treatment activates myogenic differentiation by increasing p38MAPK and decreasing JAK1-STAT1 phosphorylation levels, while osteogenic induction was enhanced by p38MAPK dependent Smad, 1/5/8 signaling pathways in C2C12 progenitor cells.
Collapse
Affiliation(s)
- Ilavenil Soundharrajan
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Da Hye Kim
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, 1095 VA Drive, Lexington, KY, 40536, USA
| | - Palaniselvam Kuppusamy
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea
| | - Ki Choon Choi
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan, 31000, Republic of Korea.
| |
Collapse
|
18
|
Adhikary S, Choudhary D, Tripathi AK, Karvande A, Ahmad N, Kothari P, Trivedi R. FGF-2 targets sclerostin in bone and myostatin in skeletal muscle to mitigate the deleterious effects of glucocorticoid on musculoskeletal degradation. Life Sci 2019; 229:261-276. [PMID: 31082400 DOI: 10.1016/j.lfs.2019.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 10/26/2022]
Abstract
AIM Myokines are associated with regulation of bone and muscle mass. However, limited information is available regarding the impact of myokines on glucocorticoid (GC) mediated adverse effects on the musculoskeletal system. This study investigates the role of myokine fibroblast growth factor-2 (FGF-2) in regulating GC-induced deleterious effects on bone and skeletal muscle. METHODS Primary osteoblast cells and C2C12 myoblast cell line were treated with FGF-2 and then exposed to dexamethasone (GC). FGF-2 mediated attenuation of the inhibitory effect of GC on osteoblast and myoblast differentiation and muscle atrophy was assessed through quantitative PCR and western blot analysis. Further, FGF-2 was administered subcutaneously to dexamethasone treated mice to collect bone and skeletal muscle tissue for in vivo analysis of bone microarchitecture, mechanical strength, histomorphometry and for histological alterations in treated tissue samples. KEY FINDINGS FGF-2 abrogated the dexamethasone induced inhibitory effect on osteoblast differentiation by modulating BMP-2 pathway and inhibiting Wnt antagonist sclerostin. Further, dexamethasone induced atrophy in C2C12 cells was mitigated by FGF-2 as evident from down regulation of atrogenes expression. FGF-2 prevented GC-induced impairment of mineral density, biomechanical strength, trabecular bone volume, cortical thickness and bone formation rate in mice. Additionally, skeletal muscle tissue from GC treated mice displayed weak myostatin immunostaining and reduced expression of atrogenes following FGF-2 treatment. SIGNIFICANCE FGF-2 mitigated GC induced effects through inhibition of sclerostin and myostatin expression in bone and muscle respectively. Taken together, this study exhibited the role of exogenous FGF-2 in sustaining osteoblastogenesis and inhibiting muscle atrophy in presence of glucocorticoid.
Collapse
Affiliation(s)
- Sulekha Adhikary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Dharmendra Choudhary
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ashish Kumar Tripathi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anirudha Karvande
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Naseer Ahmad
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Priyanka Kothari
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ritu Trivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
19
|
Takahashi K, Itakura E, Takano K, Endo T. DA-Raf, a dominant-negative regulator of the Ras–ERK pathway, is essential for skeletal myocyte differentiation including myoblast fusion and apoptosis. Exp Cell Res 2019; 376:168-180. [DOI: 10.1016/j.yexcr.2019.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/19/2019] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
|
20
|
Avin KG, Vallejo JA, Chen NX, Wang K, Touchberry CD, Brotto M, Dallas SL, Moe SM, Wacker MJ. Fibroblast growth factor 23 does not directly influence skeletal muscle cell proliferation and differentiation or ex vivo muscle contractility. Am J Physiol Endocrinol Metab 2018; 315:E594-E604. [PMID: 29558205 PMCID: PMC6230710 DOI: 10.1152/ajpendo.00343.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/29/2018] [Accepted: 02/15/2018] [Indexed: 02/06/2023]
Abstract
Skeletal muscle dysfunction accompanies the clinical disorders of chronic kidney disease (CKD) and hereditary hypophosphatemic rickets. In both disorders, fibroblast growth factor 23 (FGF23), a bone-derived hormone regulating phosphate and vitamin D metabolism, becomes chronically elevated. FGF23 has been shown to play a direct role in cardiac muscle dysfunction; however, it is unknown whether FGF23 signaling can also directly induce skeletal muscle dysfunction. We found expression of potential FGF23 receptors ( Fgfr1-4) and α-Klotho in muscles of two animal models (CD-1 and Cy/+ rat, a naturally occurring rat model of chronic kidney disease-mineral bone disorder) as well as C2C12 myoblasts and myotubes. C2C12 proliferation, myogenic gene expression, oxidative stress marker 8-OHdG, intracellular Ca2+ ([Ca2+]i), and ex vivo contractility of extensor digitorum longus (EDL) or soleus muscles were assessed after treatment with various amounts of FGF23. FGF23 (2-100 ng/ml) did not alter C2C12 proliferation, expression of myogenic genes, or oxidative stress after 24- to 72-h treatment. Acute or prolonged FGF23 treatment up to 6 days did not alter C2C12 [Ca2+]i handling, nor did acute treatment with FGF23 (9-100 ng/ml) affect EDL and soleus muscle contractility. In conclusion, although skeletal muscles express the receptors involved in FGF23-mediated signaling, in vitro FGF23 treatments failed to directly alter skeletal muscle development or function under the conditions tested. We hypothesize that other endogenous substances may be required to act in concert with FGF23 or apart from FGF23 to promote muscle dysfunction in hereditary hypophosphatemic rickets and CKD.
Collapse
Affiliation(s)
- Keith G Avin
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University , Indianapolis, Indiana
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Julian A Vallejo
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City , Kansas City, Missouri
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City , Kansas City, Missouri
| | - Neal X Chen
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
| | - Kun Wang
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City , Kansas City, Missouri
| | - Chad D Touchberry
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City , Kansas City, Missouri
| | - Marco Brotto
- College of Nursing and Health Innovation, Bone-Muscle Collaborative Sciences, University of Texas-Arlington , Arlington, Texas
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City , Kansas City, Missouri
| | - Sharon M Moe
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine , Indianapolis, Indiana
- Roudebush Veterans Administration Medical Center , Indianapolis, Indiana
| | - Michael J Wacker
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City , Kansas City, Missouri
| |
Collapse
|
21
|
Current Progress and Challenges for Skeletal Muscle Differentiation from Human Pluripotent Stem Cells Using Transgene-Free Approaches. Stem Cells Int 2018; 2018:6241681. [PMID: 29760730 PMCID: PMC5924987 DOI: 10.1155/2018/6241681] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/11/2018] [Accepted: 02/18/2018] [Indexed: 12/13/2022] Open
Abstract
Neuromuscular diseases are caused by functional defects of skeletal muscles, directly via muscle pathology or indirectly via disruption of the nervous system. Extensive studies have been performed to improve the outcomes of therapies; however, effective treatment strategies have not been fully established for any major neuromuscular disease. Human pluripotent stem cells have a great capacity to differentiate into myogenic progenitors and skeletal myocytes for use in treating and modeling neuromuscular diseases. Recent advances have allowed the creation of patient-derived stem cells, which can be used as a unique platform for comprehensive study of disease mechanisms, in vitro drug screening, and potential new cell-based therapies. In the last decade, a number of methods have been developed to derive skeletal muscle cells from human pluripotent stem cells. By controlling the process of myogenesis using transcription factors and signaling molecules, human pluripotent stem cells can be directed to differentiate into cell types observed during muscle development. In this review, we highlight signaling pathways relevant to the formation of muscle tissue during embryonic development. We then summarize current methods to differentiate human pluripotent stem cells toward the myogenic lineage, specifically focusing on transgene-free approaches. Lastly, we discuss existing challenges for deriving skeletal myocytes and myogenic progenitors from human pluripotent stem cells.
Collapse
|
22
|
Reis EPD, Paixão DM, Brustolini OJB, Silva FFE, Silva W, Araújo FMGD, Salim ACDM, Oliveira G, Guimarães SEF. Expression of myogenes in longissimus dorsi muscle during prenatal development in commercial and local Piau pigs. Genet Mol Biol 2016; 39:589-599. [PMID: 27801482 PMCID: PMC5127148 DOI: 10.1590/1678-4685-gmb-2015-0295] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 02/20/2016] [Indexed: 11/22/2022] Open
Abstract
This study used qRT-PCR to examine variation in the expression of 13 myogenes during muscle development in four prenatal periods (21, 40, 70 and 90 days post-insemination) in commercial (the three-way Duroc, Landrace and Large-White cross) and local Piau pig breeds that differ in muscle mass. There was no variation in the expression of the CHD8, EID2B, HIF1AN, IKBKB, RSPO3, SOX7 and SUFU genes at the various prenatal ages or between breeds. The MAP2K1 and RBM24 genes showed similar expression between commercial and Piau pigs but greater expression (p < 0.05) in at least one prenatal period. Pair-wise comparisons of prenatal periods in each breed showed that only the CSRP3, LEF1, MRAS and MYOG genes had higher expression (p < 0.05) in at least one prenatal period in commercial and Piau pigs. Overall, these results identified the LEF1 gene as a primary candidate to account for differences in muscle mass between the pig breeds since activation of this gene may lead to greater myoblast fusion in the commercial breed compared to Piau pigs. Such fusion could explain the different muscularity between breeds in the postnatal periods.
Collapse
Affiliation(s)
| | | | | | | | - Walmir Silva
- Departamento de Zootecnia, Universidade Federal de Viçosa (UFV), Viçosa, MG, Brazil
| | | | | | | | | |
Collapse
|
23
|
Soltani L, Rahmani HR, Daliri Joupari M, Ghaneialvar H, Mahdavi AH, Shamsara M. Ovine fetal mesenchymal stem cell differentiation to cardiomyocytes, effects of co-culture, role of small molecules; reversine and 5-azacytidine. Cell Biochem Funct 2016; 34:250-61. [DOI: 10.1002/cbf.3187] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 03/09/2016] [Accepted: 03/29/2016] [Indexed: 02/06/2023]
Affiliation(s)
- Leila Soltani
- Department of Animal Science, Faculty of Agriculture; Isfahan University of Technology; Isfahan Iran
| | - Hamid Reza Rahmani
- Department of Animal Science, Faculty of Agriculture; Isfahan University of Technology; Isfahan Iran
| | - Morteza Daliri Joupari
- Department of Animal Biotechnology; National Institute of Genetic Engineering and Biotechnology (NIGEB); Tehran Iran
| | - Hori Ghaneialvar
- Department of Clinical Biochemistry, Faculty of Medical Science; Tarbiat Modares University; Tehran Iran
| | - Amir Hossein Mahdavi
- Department of Animal Science, Faculty of Agriculture; Isfahan University of Technology; Isfahan Iran
| | - Mehdi Shamsara
- Department of Animal Biotechnology; National Institute of Genetic Engineering and Biotechnology (NIGEB); Tehran Iran
| |
Collapse
|
24
|
Kim WK, Oh KJ, Choi HR, Park A, Han BS, Chi SW, Kim SJ, Bae KH, Lee SC. MAP kinase phosphatase 3 inhibits brown adipocyte differentiation via regulation of Erk phosphorylation. Mol Cell Endocrinol 2015; 416:70-6. [PMID: 26325440 DOI: 10.1016/j.mce.2015.08.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/10/2015] [Accepted: 08/25/2015] [Indexed: 12/29/2022]
Abstract
Brown fat has been highlight as a new therapeutic target for treatment of obesity and diabetes. However, molecular mechanism underlying brown adipogenesis are not fully understood. Here, we identified that MAP kinase phosphatase 3 (MKP3) has a novel role as regulator of brown adipocyte differentiation. The expression of MKP3 was significantly decreased during the early stage(s) of brown adipocyte differentiation in HIB-1B cells and primary cells. Ectopic expression of MKP3 led to reduced brown adipocyte differentiation, whereas depletion of MKP3 significantly enhanced the differentiation of primary brown preadipocytes. Consistently, we found an increased brown adipocyte differentiation in MKP3-null MEF cells. These inhibitory effects of MKP3 could be resulted via the temporal regulation of Erk activation. In recent, it was reported that MKP3 deficient mice are resistant to diet-induced obesity, and display enhanced energy expenditure. Taken together, we suggest that MKP3 could be an important factor in the regulation of brown adipocyte differentiation.
Collapse
Affiliation(s)
- Won Kon Kim
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology of Korea, Daejeon, 305-806, Republic of Korea
| | - Kyoung-Jin Oh
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Hye-Ryung Choi
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Anna Park
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Baek Soo Han
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology of Korea, Daejeon, 305-806, Republic of Korea
| | - Seung-Wook Chi
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Seung Jun Kim
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea
| | - Kwang-Hee Bae
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology of Korea, Daejeon, 305-806, Republic of Korea.
| | - Sang Chul Lee
- Functional Genomics Research Center, KRIBB, Daejeon, 305-806, Republic of Korea; Department of Functional Genomics, University of Science and Technology of Korea, Daejeon, 305-806, Republic of Korea.
| |
Collapse
|
25
|
Abstract
The developmental mechanisms that control head muscle formation are distinct from those that operate in the trunk. Head and neck muscles derive from various mesoderm populations in the embryo and are regulated by distinct transcription factors and signaling molecules. Throughout the last decade, developmental, and lineage studies in vertebrates and invertebrates have revealed the peculiar nature of the pharyngeal mesoderm that forms certain head muscles and parts of the heart. Studies in chordates, the ancestors of vertebrates, revealed an evolutionarily conserved cardiopharyngeal field that progressively facilitates the development of both heart and craniofacial structures during vertebrate evolution. This ancient regulatory circuitry preceded and facilitated the emergence of myogenic cell types and hierarchies that exist in vertebrates. This chapter summarizes studies related to the origins, signaling circuits, genetics, and evolution of the head musculature, highlighting its heterogeneous characteristics in all these aspects, with a special focus on the FGF-ERK pathway. Additionally, we address the processes of head muscle regeneration, and the development of stem cell-based therapies for treatment of muscle disorders.
Collapse
Affiliation(s)
- Inbal Michailovici
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Eigler
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Tzahor
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
26
|
Jang YN, Baik EJ. JAK-STAT pathway and myogenic differentiation. JAKSTAT 2014; 2:e23282. [PMID: 24058805 PMCID: PMC3710318 DOI: 10.4161/jkst.23282] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 12/13/2012] [Accepted: 12/14/2012] [Indexed: 11/19/2022] Open
Abstract
Myogenic differentiation plays an important role in muscle regeneration and is regulated by two transcription factor families, MRFs and MEF2, which induce differentiation of myoblasts through expression of the muscle-specific gene, myogenin. In addition, many intracellular signaling pathways are also involved in myogenic differentiation, including p38 MAPK, ERK/MAPK and PI3K/AKT. The JAK-STAT pathway is activated by various cytokines and positively or negatively regulates the differentiation of myoblasts. JAK1 plays a notable role in proliferation; whereas, JAK2 and JAK3 function mainly in differentiation. The STATs, molecules downstream of JAK, regulate myogenesis. With JAK1, STAT1 promotes proliferation, while STAT3 has a dual effect on proliferation and differentiation. The JAK-STAT negative regulator, SOCS, is also associated with myogenesis; although, its role is controversial. In this review, we will discuss the role of the JAK-STAT pathway on myogenic differentiation.
Collapse
Affiliation(s)
- You-Na Jang
- Department of Physiology; Chronic Inflammatory Disease Research Center; Ajou University School of Medicine; Suwon, Korea
| | | |
Collapse
|
27
|
Implant of Polymer Containing Pentacyclic Triterpenes from Eugenia punicifolia Inhibits Inflammation and Activates Skeletal Muscle Remodeling. Arch Immunol Ther Exp (Warsz) 2014; 62:483-91. [DOI: 10.1007/s00005-014-0291-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/24/2013] [Indexed: 10/25/2022]
|
28
|
Muir LA, Nguyen QG, Hauschka SD, Chamberlain JS. Engraftment potential of dermal fibroblasts following in vivo myogenic conversion in immunocompetent dystrophic skeletal muscle. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14025. [PMID: 25558461 PMCID: PMC4280788 DOI: 10.1038/mtm.2014.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Autologous dermal fibroblasts (dFbs) are promising candidates for enhancing muscle regeneration in Duchenne muscular dystrophy (DMD) due to their ease of isolation, immunological compatibility, and greater proliferative potential than DMD satellite cells. We previously showed that mouse fibroblasts, after MyoD-mediated myogenic reprogramming in vivo, engraft in skeletal muscle and supply dystrophin. Assessing the therapeutic utility of this system requires optimization of conversion and transplantation conditions and quantitation of engraftment so that these parameters can be correlated with possible functional improvements. Here, we derived dFbs from transgenic mice carrying mini-dystrophin, transduced them by lentivirus carrying tamoxifen-inducible MyoD, and characterized their myogenic and engraftment potential. After cell transplantation into the muscles of immunocompetent dystrophic mdx4cv mice, tamoxifen treatment drove myogenic conversion and fusion into myofibers that expressed high levels of mini-dystrophin. Injecting 50,000 cells/µl (1 × 106 total cells) resulted in a peak of ~600 mini-dystrophin positive myofibers in tibialis anterior muscle single cross-sections. However, extensor digitorum longus muscles with up to 30% regional engraftment showed no functional improvements; similar limitations were obtained with whole muscle mononuclear cells. Despite the current lack of physiological improvement, this study suggests a viable initial strategy for using a patient-accessible dermal cell population to enhance skeletal muscle regeneration in DMD.
Collapse
Affiliation(s)
- Lindsey A Muir
- Program in Molecular and Cellular Biology, University of Washington ; Department of Neurology, University of Washington
| | | | | | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington ; Department of Biochemistry, University of Washington ; Department of Medicine, University of Washington
| |
Collapse
|
29
|
McClure MJ, Garg K, Simpson DG, Ryan JJ, Sell SA, Bowlin GL, Ericksen JJ. The influence of platelet-rich plasma on myogenic differentiation. J Tissue Eng Regen Med 2013; 10:E239-49. [PMID: 23868863 DOI: 10.1002/term.1755] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 03/04/2013] [Accepted: 03/25/2013] [Indexed: 01/24/2023]
Abstract
The ability to expand and direct both precursor and stem cells towards a differential fate is considered extremely advantageous in tissue engineering. Platelet-rich plasma (PRP) possesses a milieu of growth factors and cytokines, which have the potential to have either a differentiative or proliferative influence on the cell type tested. Here, we investigated the effect of PRP on C2C12 myoblasts. A range of PRP concentrations in differentiation media was used to determine whether a concentration dependence existed, while PRP embedded in fibres of aligned electrospun polydioxanone and polycaprolactone was used to determine whether this presence of fibres would cause any differences in response. In both cases, it was found that late myogenic markers were suppressed after 7 days in culture. However, an early differentiation marker, MyoD, was upregulated during this same time period. The results from this study represent the ability of PRP to have an influence over both myogenic proliferation and differentiation, a factor which could prove useful in future studies involved with skeletal muscle tissue engineering.
Collapse
Affiliation(s)
- Michael J McClure
- Physical Medicine and Rehabilitation Service, Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA.,Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Koyal Garg
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - David G Simpson
- Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA, USA
| | - Scott A Sell
- Department of Biomedical Engineering, Parks College of Engineering, Aviation and Technology, Saint Louis University, St. Louis, MO, USA
| | - Gary L Bowlin
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Jeffery J Ericksen
- Physical Medicine and Rehabilitation Service, Hunter Holmes McGuire VA Medical Center, Richmond, VA, USA
| |
Collapse
|
30
|
Abstract
Myoblast fusion is a critical process that contributes to the growth of muscle during development and to the regeneration of myofibers upon injury. Myoblasts fuse with each other as well as with multinucleated myotubes to enlarge the myofiber. Initial studies demonstrated that myoblast fusion requires extracellular calcium and changes in cell membrane topography and cytoskeletal organization. More recent studies have identified several cell-surface and intracellular proteins that mediate myoblast fusion. Furthermore, emerging evidence suggests that myoblast fusion is also regulated by the activation of specific cell-signaling pathways that lead to the expression of genes whose products are essential for the fusion process and for modulating the activity of molecules that are involved in cytoskeletal rearrangement. Here, we review the roles of the major signaling pathways in mammalian myoblast fusion.
Collapse
Affiliation(s)
- Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | | | | |
Collapse
|
31
|
Li B, Qu C, Chen C, Liu Y, Akiyama K, Yang R, Chen F, Zhao Y, Shi S. Basic fibroblast growth factor inhibits osteogenic differentiation of stem cells from human exfoliated deciduous teeth through ERK signaling. Oral Dis 2011; 18:285-92. [PMID: 22151351 DOI: 10.1111/j.1601-0825.2011.01878.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Stem cells from human exfoliated deciduous teeth (SHED) are a unique postnatal stem cell population capable of regenerating mineralized tissue and treating immune disorders. However, the mechanism that controls SHED differentiation is not fully understood. Here, we showed that basic fibroblast growth factor (bFGF) treatment attenuated SHED-mediated mineralized tissue regeneration through activation of the extracellular signal-regulated kinase (ERK) 1/2 pathway. MATERIAL AND METHOD The level of mineralized nodule formation was assessed by alizarin red staining. Expression levels of osteogenic genes, osteocalcin and runt-related transcription factor 2, were examined by RT-PCR. Subcutaneous implantation approach was used to assess in vivo bone formation. Downstream signaling pathways of bFGF were examined by Western blotting. RESULT Activation of ERK1/2 signaling by bFGF treatment inhibited WNT/β-catenin pathway, leading to osteogenic deficiency of SHED. ERK1/2 inhibitor treatment rescued bFGF-induced osteogenic differentiation deficiency. CONCLUSION These data suggest that bFGF inhibits osteogenic differentiation of SHED via ERK1/2 pathway. Blockade ERK1/2 signaling by small molecular inhibitor treatment improves bone formation of SHED after bFGF treatment.
Collapse
Affiliation(s)
- B Li
- Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Shaanxi, China
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Jang YN, Lee IJ, Park MC, Baik EJ. Role of JAK3 in myogenic differentiation. Cell Signal 2011; 24:742-9. [PMID: 22120524 DOI: 10.1016/j.cellsig.2011.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 11/08/2011] [Accepted: 11/08/2011] [Indexed: 01/06/2023]
Abstract
Skeletal muscle differentiation is regulated by transcription factors, including members of the myogenic regulatory factor (MRF) family and many signaling pathways. The JAK1 and JAK2 pathways are known to each have different effects on myoblast proliferation and differentiation; however, the role of JAK3 in myoblast differentiation remains unclear. In this study, we investigated the effect of JAK3 inhibition on myogenic differentiation in the C2C12 mouse myoblast cell line. During myogenic differentiation, treatment with the JAK3 inhibitor WHIp154 significantly increased the number of MHC-positive multinucleated myotubes and the expressions of myosin heavy chain (MHC), myogenin (MGN), MyoD, and myogenic enhancer factor 2 (MEF2). Knockdown of the JAK3 gene using siJAK3 also significantly increased MHC, MGN and MyoD mRNA expressions as well as insulin-like growth factor-II (IGF-II) gene expression. During differentiation, JAK3 was initially activated and later decreased. Differentiation decreased STAT1, which was further decreased by WHIp154. In contrast, STAT3 gradually was elevated during differentiation, and was increased by JAK3 inhibition. Moreover, we found that up-regulation of AKT activity and down-regulation of ERK activity cooperated to accelerate myogenic differentiation. Taken together, these data indicate that JAK3 inhibition potently facilitates myoblast differentiation through antagonistic STAT1/STAT3 activities. Additionally, JAK3 inhibition induced precocious differentiation and played important roles for terminal differentiation, including fusion, which is involved with regulation of AKT and ERK pathways.
Collapse
Affiliation(s)
- You-Na Jang
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | | | | | | |
Collapse
|
33
|
Burks TN, Andres-Mateos E, Marx R, Mejias R, Van Erp C, Simmers JL, Walston JD, Ward CW, Cohn RD. Losartan restores skeletal muscle remodeling and protects against disuse atrophy in sarcopenia. Sci Transl Med 2011; 3:82ra37. [PMID: 21562229 DOI: 10.1126/scitranslmed.3002227] [Citation(s) in RCA: 179] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sarcopenia, a critical loss of muscle mass and function because of the physiological process of aging, contributes to disability and mortality in older adults. It increases the incidence of pathologic fractures, causing prolonged periods of hospitalization and rehabilitation. The molecular mechanisms underlying sarcopenia are poorly understood, but recent evidence suggests that increased transforming growth factor-β (TGF-β) signaling contributes to impaired satellite cell function and muscle repair in aged skeletal muscle. We therefore evaluated whether antagonism of TGF-β signaling via losartan, an angiotensin II receptor antagonist commonly used to treat high blood pressure, had a beneficial impact on the muscle remodeling process of sarcopenic mice. We demonstrated that mice treated with losartan developed significantly less fibrosis and exhibited improved in vivo muscle function after cardiotoxin-induced injury. We found that losartan not only blunted the canonical TGF-β signaling cascade but also modulated the noncanonical TGF-β mitogen-activated protein kinase pathway. We next assessed whether losartan was able to combat disuse atrophy in aged mice that were subjected to hindlimb immobilization. We showed that immobilized mice treated with losartan were protected against loss of muscle mass. Unexpectedly, this protective mechanism was not mediated by TGF-β signaling but was due to an increased activation of the insulin-like growth factor 1 (IGF-1)/Akt/mammalian target of rapamycin (mTOR) pathway. Thus, blockade of the AT1 (angiotensin II type I) receptor improved muscle remodeling and protected against disuse atrophy by differentially regulating the TGF-β and IGF-1/Akt/mTOR signaling cascades, two pathways critical for skeletal muscle homeostasis. Thus, losartan, a Food and Drug Administration-approved drug, may prove to have clinical benefits to combat injury-related muscle remodeling and provide protection against disuse atrophy in humans with sarcopenia.
Collapse
Affiliation(s)
- Tyesha N Burks
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
The myogenic kinome: protein kinases critical to mammalian skeletal myogenesis. Skelet Muscle 2011; 1:29. [PMID: 21902831 PMCID: PMC3180440 DOI: 10.1186/2044-5040-1-29] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 09/08/2011] [Indexed: 12/13/2022] Open
Abstract
Myogenesis is a complex and tightly regulated process, the end result of which is the formation of a multinucleated myofibre with contractile capability. Typically, this process is described as being regulated by a coordinated transcriptional hierarchy. However, like any cellular process, myogenesis is also controlled by members of the protein kinase family, which transmit and execute signals initiated by promyogenic stimuli. In this review, we describe the various kinases involved in mammalian skeletal myogenesis: which step of myogenesis a particular kinase regulates, how it is activated (if known) and what its downstream effects are. We present a scheme of protein kinase activity, similar to that which exists for the myogenic transcription factors, to better clarify the complex signalling that underlies muscle development.
Collapse
|
35
|
Murray J, Huss JM. Estrogen-related receptor α regulates skeletal myocyte differentiation via modulation of the ERK MAP kinase pathway. Am J Physiol Cell Physiol 2011; 301:C630-45. [PMID: 21562305 PMCID: PMC3174569 DOI: 10.1152/ajpcell.00033.2011] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 05/06/2011] [Indexed: 02/06/2023]
Abstract
Myocyte differentiation involves complex interactions between signal transduction pathways and transcription factors. The estrogen-related receptors (ERRs) regulate energy substrate uptake, mitochondrial respiration, and biogenesis and may target structural gene programs in striated muscle. However, ERRα's role in regulating myocyte differentiation is not known. ERRα and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) are coordinately upregulated with metabolic and skeletal muscle-specific genes early in myogenesis. We analyzed effects of ERRα overexpression and loss of function in myogenic models. In C2C12 myocytes ERRα overexpression accelerated differentiation, whereas XCT790 treatment delayed myogenesis and resulted in myotubes with fewer mitochondria and disorganized sarcomeres. ERRα-/- primary myocytes showed delayed myogenesis, resulting in structurally immature myotubes with reduced sarcomeric assembly and mitochondrial function. However, sarcomeric and metabolic gene expression was unaffected or upregulated in ERRα-/- cells. Instead, ERRα-/- myocytes exhibited aberrant ERK activation early in myogenesis, consistent with delayed myotube formation. XCT790 treatment also increased ERK phosphorylation in C2C12, whereas ERRα overexpression decreased early ERK activation, consistent with the opposing effects of these treatments on differentiation. The transient induction of MAP kinase phosphatase-1 (MKP-1), which mediates ERK dephosphorylation at the onset of myogenesis, was lost in ERRα-/- myocytes and in XCT790-treated C2C12. The ERRα-PGC-1α complex activates the Dusp1 gene, which encodes MKP-1, and ERRα occupies the proximal 5' regulatory region during early differentiation in C2C12 myocytes. Finally, treatment of ERRα-/- myocytes with MEK inhibitors rescued normal ERK signaling and myogenesis. Collectively, these data demonstrate that ERRα is required for normal skeletal myocyte differentiation via modulation of MAP kinase signaling.
Collapse
MESH Headings
- Animals
- Butadienes/pharmacology
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Creatine Kinase, Mitochondrial Form/genetics
- Dual Specificity Phosphatase 1/genetics
- Dual Specificity Phosphatase 1/metabolism
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Flavonoids/pharmacology
- Gene Expression/drug effects
- Gene Expression/genetics
- Kinetics
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria/metabolism
- Mitochondria/pathology
- Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Muscle Development/drug effects
- Muscle Development/physiology
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/enzymology
- Muscle Fibers, Skeletal/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Myoblasts, Skeletal/cytology
- Myoblasts, Skeletal/drug effects
- Myoblasts, Skeletal/enzymology
- Myoblasts, Skeletal/metabolism
- Myogenin/genetics
- Myosin Heavy Chains/genetics
- Nitriles/pharmacology
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
- Phosphorylation/drug effects
- Protein Binding/genetics
- Protein Serine-Threonine Kinases/genetics
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/deficiency
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Sarcomeres/pathology
- Thiazoles/pharmacology
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors
- Transduction, Genetic
- Troponin I/genetics
- Troponin I/metabolism
- Up-Regulation/drug effects
- Up-Regulation/genetics
- p38 Mitogen-Activated Protein Kinases/metabolism
- ERRalpha Estrogen-Related Receptor
Collapse
Affiliation(s)
- Jennifer Murray
- Division of Gene Regulation and Drug Discovery, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Duarte, California, USA
| | | |
Collapse
|
36
|
Minami M, Koyama T, Wakayama Y, Fukuhara S, Mochizuki N. EphrinA/EphA signal facilitates insulin-like growth factor-I-induced myogenic differentiation through suppression of the Ras/extracellular signal-regulated kinase 1/2 cascade in myoblast cell lines. Mol Biol Cell 2011; 22:3508-19. [PMID: 21795402 PMCID: PMC3172274 DOI: 10.1091/mbc.e11-03-0183] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Insulin-like growth factor-I (IGF-I) activates not only the phosphatidylinositol 3-kinase (PI3K)-AKT cascade that is essential for myogenic differentiation but also the extracellular signal-regulated kinase (ERK) 1/2 cascade that inhibits myogenesis. We hypothesized that there must be a signal that inhibits ERK1/2 upon cell-cell contact required for skeletal myogenesis. Cell-cell contact-induced engagement of ephrin ligands and Eph receptors leads to downregulation of the Ras-ERK1/2 pathway through p120 Ras GTPase-activating protein (p120RasGAP). We therefore investigated the significance of the ephrin/Eph signal in IGF-I-induced myogenesis. EphrinA1-Fc suppressed IGF-I-induced activation of Ras and ERK1/2, but not that of AKT, in C2C12 myoblasts, whereas ephrinB1-Fc affected neither ERK1/2 nor AKT activated by IGF-I. IGF-I-dependent myogenic differentiation of C2C12 myoblasts was potentiated by ephrinA1-Fc. In p120RasGAP-depleted cells, ephrinA1-Fc failed to suppress the Ras-ERK1/2 cascade by IGF-I and to promote IGF-I-mediated myogenesis. EphrinA1-Fc did not promote IGF-I-dependent myogenesis when the ERK1/2 was constitutively activated. Furthermore, a dominant-negative EphA receptor blunted IGF-I-induced myogenesis in C2C12 and L6 myoblasts. However, the inhibition of IGF-I-mediated myogenesis by down-regulation of ephrinA/EphA signal was canceled by inactivation of the ERK1/2 pathway. Collectively, these findings demonstrate that the ephrinA/EphA signal facilitates IGF-I-induced myogenesis by suppressing the Ras-ERK1/2 cascade through p120RasGAP in myoblast cell lines.
Collapse
Affiliation(s)
- Masayoshi Minami
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka 565-8565, Japan
| | | | | | | | | |
Collapse
|
37
|
Qian Q, Qian H, Zhang X, Zhu W, Yan Y, Ye S, Peng X, Li W, Xu Z, Sun L, Xu W. 5-Azacytidine induces cardiac differentiation of human umbilical cord-derived mesenchymal stem cells by activating extracellular regulated kinase. Stem Cells Dev 2011; 21:67-75. [PMID: 21476855 DOI: 10.1089/scd.2010.0519] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
5-Azacytidine (5-Aza) induces differentiation of mesenchymal stem cells (MSCs) into cardiomyocytes. However, the underlying mechanisms are not well understood. Our previous work showed that 5-Aza induces human bone marrow-derived MSCs to differentiate into cardiomyocytes. Here, we demonstrated that 5-Aza induced cardiac differentiation of human umbilical cord-derived MSCs (hucMSCs) and explored the potential signaling pathway. Our results showed that hucMSCs had cardiomyocyte phenotypes after 5-Aza treatment. In addition, myogenic cells differentiated from hucMSCs were positive for mRNA and protein of desmin, β-myosin heavy chain, cardiac troponin T, A-type natriuretic peptide, and Nkx2.5. Human diploid lung fibroblasts treated with 5-Aza expressed no cardiac-specific genes. 5-Aza did not induce hucMSCs to differentiate into osteoblasts. Further study revealed that 5-Aza treatment activated extracellular signal related kinases (ERK) in hucMSCs, but protein kinase C showed no response to 5-Aza administration. U0126, a specific inhibitor of ERK, could inhibit 5-Aza-induced expression of cardiac-specific genes and proteins in hucMSCs. Increased phosphorylation of signal transducers and activators of transcription 3, and up-regulation of myocyte enhancer-binding factor-2c and myogenic differentiation antigen in 5-Aza-treated hucMSCs were also suppressed by U0126. Taken together, these results suggested that sustained activation of ERK by 5-Aza contributed to the induction of the differentiation of hucMSCs into cardiomyocytes in vitro.
Collapse
Affiliation(s)
- Qian Qian
- School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Halwani R, Al-Abri J, Beland M, Al-Jahdali H, Halayko AJ, Lee TH, Al-Muhsen S, Hamid Q. CC and CXC chemokines induce airway smooth muscle proliferation and survival. THE JOURNAL OF IMMUNOLOGY 2011; 186:4156-63. [PMID: 21368236 DOI: 10.4049/jimmunol.1001210] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The increase in airway smooth muscle (ASM) mass is a major structural change in asthma. This increase has been attributed to ASM cell (ASMC) hyperplasia and hypertrophy. The distance between ASMC and the epithelium is reduced, suggesting migration of smooth muscle cells toward the epithelium. Recent studies have suggested a role of chemokines in ASMC migration toward the epithelium; however, chemokines have other biological effects. The objective of the current study is to test the hypothesis that chemokines (eotaxin, RANTES, IL-8, and MIP-1α) can directly influence ASMC mass by increasing the rate of proliferation or enhancing the survival of these cells. Human ASMCs were exposed to different concentrations of eotaxin, RANTES, IL-8, or MIP-1α. To test for proliferation, matched control and stimulated ASMC were pulsed with [(3)H]thymidine, or ASMCs were stained with BrdU and then analyzed with flow cytometry. Apoptosis was measured using Annexin V staining and flow cytometry. Expression of phosphorylated p42/p44 and MAPKs was assessed by Western blot. In a concentration-dependent manner, chemokines including eotaxin, RANTES, IL-8, and MIP-1α increased ASMC's [(3)H]thymidine incorporation and DNA synthesis. IL-8, eotaxin, and MIP-1α decreased the rate of apoptosis of ASMCs compared with the matched controls. A significant increase in phosphorylated p42/p44 MAPKs was seen after treating ASMCs with RANTES and eotaxin. Moreover, inhibition of p42/p44 MAPK phosphorylation reduced the level of chemokine-induced ASM proliferation. We conclude that chemokines might contribute to airway remodeling seen in asthma by enhancing the number and survival of ASMCs.
Collapse
Affiliation(s)
- Rabih Halwani
- Prince Naif Center for Immunology Research, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Sangaj N, Kyriakakis P, Yang D, Chang CW, Arya G, Varghese S. Heparin mimicking polymer promotes myogenic differentiation of muscle progenitor cells. Biomacromolecules 2010; 11:3294-300. [PMID: 21058638 PMCID: PMC4136807 DOI: 10.1021/bm101041f] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Heparin and heparan sulfate mediated basic fibroblast growth factor (bFGF) signaling plays an important role in skeletal muscle homeostasis by maintaining a balance between proliferation and differentiation of muscle progenitor cells. In this study we investigate the role of a synthetic mimic of heparin, poly(sodium-4-styrenesulfonate) (PSS), on myogenic differentiation of C2C12 cells. Exogenous supplementation of PSS increased the differentiation of C2C12 cells in a dose-dependent manner, while the formation of multinucleated myotubes exhibited a nonmonotonic dependence with the concentration of PSS. Our results further suggest that one possible mechanism by which PSS promotes myogenic differentiation is by downregulating the mitogen activated extracellular regulated signaling kinase (MAPK/ERK) pathway. The binding ability of PSS to bFGF was found to be comparable to heparin through molecular docking calculations and by native PAGE. Such synthetic heparin mimics could offer a cost-effective alternative to heparin and also reduce the risk associated with batch-to-batch variation and contamination of heparin.
Collapse
Affiliation(s)
- Nivedita Sangaj
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Phillip Kyriakakis
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Darren Yang
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chien-Wen Chang
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gaurav Arya
- Department of Nanoengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Shyni Varghese
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
40
|
Gutiérrez J, Brandan E. A novel mechanism of sequestering fibroblast growth factor 2 by glypican in lipid rafts, allowing skeletal muscle differentiation. Mol Cell Biol 2010; 30:1634-49. [PMID: 20100867 PMCID: PMC2838066 DOI: 10.1128/mcb.01164-09] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 09/25/2009] [Accepted: 01/14/2010] [Indexed: 12/14/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are critical modulators of growth factor activities. Skeletal muscle differentiation is strongly inhibited by fibroblast growth factor 2 (FGF-2). We have shown that HSPGs present at the plasma membrane are expressed in myoblasts and are downregulated during muscle differentiation. An exception is glypican-1, which is present throughout the myogenic process. Myoblasts that do not express glypican-1 exhibit defective differentiation, with an increase in the receptor binding of FGF-2, concomitant with increased signaling. Glypican-1-deficient myoblasts show decreased expression of myogenin, the master gene that controls myogenesis, myosin, and the myoblast fusion index. Reversion of these defects was induced by expression of rat glypican-1. Glypican-1 is the only HSPG localized in lipid raft domains in myoblasts, resulting in the sequestration of FGF-2 away from FGF-2 receptors (FGFRs) located in nonraft domains. A chimeric glypican-1, containing syndecan-1 transmembrane and cytoplasmic domains, is located in nonraft domains interacting with FGFR-IV- and enhanced FGF-2-dependent signaling. Thus, glypican-1 acts as a positive regulator of muscle differentiation by sequestering FGF-2 in lipid rafts and preventing its binding and dependent signaling.
Collapse
Affiliation(s)
- Jaime Gutiérrez
- Centro de Regulación Celular y Patología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, MIFAB, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrique Brandan
- Centro de Regulación Celular y Patología (CRCP), Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, MIFAB, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
41
|
The stimulative effects of endogenous opioids on endothelial cell proliferation, migration and angiogenesis in vitro. Eur J Pharmacol 2010; 628:42-50. [DOI: 10.1016/j.ejphar.2009.11.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 11/03/2009] [Accepted: 11/16/2009] [Indexed: 12/26/2022]
|
42
|
Makino T, Jinnin M, Muchemwa F, Fukushima S, Kogushi-Nishi H, Moriya C, Igata T, Fujisawa A, Johno T, Ihn H. Basic fibroblast growth factor stimulates the proliferation of human dermal fibroblasts via the ERK1/2 and JNK pathways. Br J Dermatol 2009; 162:717-23. [DOI: 10.1111/j.1365-2133.2009.09581.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
43
|
MEK1 plays contrary stage-specific roles in skeletal myogenic differentiation. Cell Signal 2009; 21:1910-7. [DOI: 10.1016/j.cellsig.2009.08.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 08/14/2009] [Accepted: 08/21/2009] [Indexed: 12/11/2022]
|
44
|
Abe S, Rhee S, Iwanuma O, Hiroki E, Yanagisawa N, Sakiyama K, Ide Y. Effect of Mechanical Stretching on Expressions of Muscle Specific Transcription Factors MyoD, Myf-5, Myogenin and MRF4 in Proliferated Myoblasts. Anat Histol Embryol 2009; 38:305-10. [DOI: 10.1111/j.1439-0264.2009.00945.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
45
|
Yang Y, Xu Y, Li W, Wang G, Song Y, Yang G, Han X, Du Z, Sun L, Ma K. STAT3 induces muscle stem cell differentiation by interaction with myoD. Cytokine 2009; 46:137-41. [PMID: 19223199 DOI: 10.1016/j.cyto.2008.12.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2008] [Accepted: 12/29/2008] [Indexed: 11/17/2022]
Abstract
Signal transducers and activators of transcription (STAT) family proteins transduce pivotal biological effects of various cytokines and hormones. STAT3 proteins are known to play a central role in the regulation of growth, differentiation, and survival of many types of cells. However, the function of STAT3 in myogenesis still remains largely unknown. We now provided direct evidence that STAT3 could induce myogenic differentiation and this effect might be mediated by interaction with MyoD--the essential transcription factor during myogenic differentiation. Furthermore, leukemia inhibitory factor (LIF) might be the upstream factor which activated JAK2/STAT3 pathway to stimulate muscle cell differentiation. Taken together, these results provide a molecular basis for further understanding of the muscle regeneration mechanism.
Collapse
Affiliation(s)
- Yanping Yang
- Department of Hematology and Oncology, The First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Barop J, Sauer H, Steger K, Wimmer M. Differentiation-dependent PTPIP51 expression in human skeletal muscle cell culture. J Histochem Cytochem 2009; 57:425-35. [PMID: 19124842 DOI: 10.1369/jhc.2008.952846] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Protein tyrosine phosphatase-interacting protein 51 (PTPIP51) expression was analyzed in proliferating and differentiating human myogenic cells cultured in vitro. Satellite cell cultures derived from four different individuals were used in this study. To analyze the expression of PTPIP51, myoblasts were cultured under conditions promoting either proliferation or differentiation. In addition, further differentiation of already-differentiated myobtubes was inhibited by resubmitting the cells to conditions promoting proliferation. PTPIP51 protein and mRNA were investigated in samples taken at defined time intervals by immunostaining, immunoblotting, in situ hybridization, and PCR. Image analyses of fluorescence immunostainings were used to quantify PTPIP51 in cultured myoblasts and myotubes. Myoblasts grown in the presence of epidermal and fibroblast growth factors (EGF and FGF), both promoting proliferation, expressed PTPIP51 on a basic level. Differentiation to multinuclear myotubes displayed a linear increase in PTPIP51 expression. The rise in PTPIP51 protein was paralleled by an augmented expression of muscle-specific proteins, namely, sarcoplasmic reticulum Ca(2+) ATPase and myosin heavy-chain protein, both linked to a progressive state of myotubal differentiation. This differentiation-induced increase in PTPIP51 was partly reversible by resubmission of differentiated myotubes to conditions boosting proliferation. The results clearly point toward a strong association between PTPIP51 expression and differentiation in human muscle cells.
Collapse
Affiliation(s)
- Justus Barop
- Institute of Anatomy and Cell Biology, Justus-Liebig University, 35385 Giessen, Germany.
| | | | | | | |
Collapse
|
47
|
Stenzinger A, Schreiner D, Koch P, Hofer HW, Wimmer M. Cell and molecular biology of the novel protein tyrosine-phosphatase-interacting protein 51. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 275:183-246. [PMID: 19491056 DOI: 10.1016/s1937-6448(09)75006-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This chapter examines the current state of knowledge about the expression profile, as well as biochemical properties and biological functions of the evolutionarily conserved protein PTPIP51. PTPIP51 is apparently expressed in splice variants and shows a particularly high expression in epithelia, skeletal muscle, placenta, and germ cells, as well as during mammalian development and in cancer. PTPIP51 is an in vitro substrate of Src- and protein kinase A, the PTP1B/TCPTP protein tyrosine phosphatases and interacts with 14-3-3 proteins, the Nuf2 kinetochore protein, the ninein-interacting CGI-99 protein, diacylglycerol kinase alpha, and also with itself forming dimers and trimers. Although the precise cellular function remains to be elucidated, the current data implicate PTPIP51 in signaling cascades mediating proliferation, differentiation, apoptosis, and motility.
Collapse
Affiliation(s)
- Albrecht Stenzinger
- Institute of Anatomy and Cell Biology, Justus-Liebig-University, Giessen, Germany
| | | | | | | | | |
Collapse
|
48
|
Kim HJ, Archer E, Escobedo N, Tapscott SJ, Unguez GA. Inhibition of mammalian muscle differentiation by regeneration blastema extract of Sternopygus macrurus. Dev Dyn 2008; 237:2830-43. [PMID: 18816861 DOI: 10.1002/dvdy.21702] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Tissue regeneration through stem cell activation and/or cell dedifferentiation is widely distributed across the animal kingdom. By comparison, regeneration in mammals is poor and this may reflect a limited dedifferentiation potential of mature cells. Because mammalian myotubes can dedifferentiate in the presence of newt blastema extract, the present study tested the dedifferentiation induction capability of the blastema from the teleost Sternopygus macrurus (SmBE). Our in vitro data showed that SmBE did not induce cell cycle reentry of myonuclei in myotubes. Instead, SmBE caused myotubes to detach and time-lapse imaging analyses characterized the cellular events before their detachment. Furthermore, SmBE enhanced myoblast proliferation and reversibly inhibited their differentiation. These data suggest the presence of protein factors in SmBE that regulate mammalian muscle physiology and differentiation, but do not support the conservation of a dedifferentiation induction capability by the blastema of S. macrurus.
Collapse
Affiliation(s)
- Hyun-Jung Kim
- Biology Department, New Mexico State University, Las Cruces, New Mexico, USA
| | | | | | | | | |
Collapse
|
49
|
Rishniw M, Fisher PJ, Doran RM, Bliss SP, Kotlikoff MI. Striated myogenesis and peristalsis in the fetal murine esophagus occur without cell migration or interstitial cells of Cajal. Cells Tissues Organs 2008; 189:410-9. [PMID: 18784410 DOI: 10.1159/000155225] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2008] [Indexed: 11/19/2022] Open
Abstract
Esophageal striated myogenesis progresses differently from appendicular myogenesis, but the mechanism underlying this process is incompletely understood. Early theories of transdifferentiation of smooth muscle into striated muscle are not supported by transgenic fate-mapping experiments; however, the origin of esophageal striated muscle remains unknown. To better define the process of striated myogenesis, we examined myogenesis in murine fetal cultured esophageal whole-organ explants. Embryonic day 14.5 (E14.5) esophagi maintained a functional contractile phenotype for up to 7 days in culture. Striated myogenesis, as evidenced by myogenin expression, proceeded in a craniocaudal direction along the length of the esophagus. Esophageal length did not change during this process. Complete, but not partial, mechanical disruption of the rostral esophagus inhibited myogenesis distally. Addition of fibroblast growth factor-2 (FGF-2) to the culture media failed to inhibit striated myogenesis, but attenuated smooth muscle actin expression and reduced peristaltic activity. Inhibition of c-kit failed to inhibit peristalsis. These results suggest that striated myogenic precursors are resident along the entire length of the esophagus by day 14.5 and do not migrate along the esophagus after E14.5. Induction of myogenesis craniocaudally appears to require physical continuity of the esophagus and is not inhibited by FGF-2. Finally, peristalsis in E14.5 esophagi appears not to be regulated by interstitial cells of Cajal.
Collapse
Affiliation(s)
- M Rishniw
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853-6401, USA
| | | | | | | | | |
Collapse
|
50
|
Koyama T, Nakaoka Y, Fujio Y, Hirota H, Nishida K, Sugiyama S, Okamoto K, Yamauchi-Takihara K, Yoshimura M, Mochizuki S, Hori M, Hirano T, Mochizuki N. Interaction of scaffolding adaptor protein Gab1 with tyrosine phosphatase SHP2 negatively regulates IGF-I-dependent myogenic differentiation via the ERK1/2 signaling pathway. J Biol Chem 2008; 283:24234-44. [PMID: 18577518 DOI: 10.1074/jbc.m803907200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Grb2-associated binder 1 (Gab1) coordinates various receptor tyrosine kinase signaling pathways. Although skeletal muscle differentiation is regulated by some growth factors, it remains elusive whether Gab1 coordinates myogenic signals. Here, we examined the molecular mechanism of insulin-like growth factor-I (IGF-I)-mediated myogenic differentiation, focusing on Gab1 and its downstream signaling. Gab1 underwent tyrosine phosphorylation and subsequent complex formation with protein-tyrosine phosphatase SHP2 upon IGF-I stimulation in C2C12 myoblasts. On the other hand, Gab1 constitutively associated with phosphatidylinositol 3-kinase regulatory subunit p85. To delineate the role of Gab1 in IGF-I-dependent signaling, we examined the effect of adenovirus-mediated forced expression of wild-type Gab1 (Gab1(WT)), mutated Gab1 that is unable to bind SHP2 (Gab1(DeltaSHP2)), or mutated Gab1 that is unable to bind p85 (Gab1(Deltap85)), on the differentiation of C2C12 myoblasts. IGF-I-induced myogenic differentiation was enhanced in myoblasts overexpressing Gab1(DeltaSHP2), but inhibited in those overexpressing either Gab1(WT) or Gab1(Deltap85). Conversely, IGF-I-induced extracellular signal-regulated kinase 1/2 (ERK1/2) activation was significantly repressed in myoblasts overexpressing Gab1(DeltaSHP2) but enhanced in those overexpressing either Gab1(WT) or Gab1(Deltap85). Furthermore, small interference RNA-mediated Gab1 knockdown enhanced myogenic differentiation. Overexpression of catalytic-inactive SHP2 modulated IGF-I-induced myogenic differentiation and ERK1/2 activation similarly to that of Gab1(DeltaSHP2), suggesting that Gab1-SHP2 complex inhibits IGF-I-dependent myogenesis through ERK1/2. Consistently, the blockade of ERK1/2 pathway reversed the inhibitory effect of Gab1(WT) overexpression on myogenic differentiation, and constitutive activation of the ERK1/2 pathway suppressed the enhanced myogenic differentiation by overexpression of Gab1(DeltaSHP2). Collectively, these data suggest that the Gab1-SHP2-ERK1/2 signaling pathway comprises an inhibitory axis for IGF-I-dependent myogenic differentiation.
Collapse
Affiliation(s)
- Tatsuya Koyama
- Department of Structural Analysis, National Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|