1
|
Pundlik SS, Barik A, Venkateshvaran A, Sahoo SS, Jaysingh MA, Math RGH, Lal H, Hashmi MA, Ramanathan A. Senescent cells inhibit mouse myoblast differentiation via the SASP-lipid 15d-PGJ 2 mediated modification and control of HRas. eLife 2024; 13:RP95229. [PMID: 39196610 PMCID: PMC11357351 DOI: 10.7554/elife.95229] [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: 08/29/2024] Open
Abstract
Senescent cells are characterized by multiple features such as increased expression of senescence-associated β-galactosidase activity (SA β-gal) and cell cycle inhibitors such as p21 or p16. They accumulate with tissue damage and dysregulate tissue homeostasis. In the context of skeletal muscle, it is known that agents used for chemotherapy such as Doxorubicin (Doxo) cause buildup of senescent cells, leading to the inhibition of tissue regeneration. Senescent cells influence the neighboring cells via numerous secreted factors which form the senescence-associated secreted phenotype (SASP). Lipids are emerging as a key component of SASP that can control tissue homeostasis. Arachidonic acid-derived lipids have been shown to accumulate within senescent cells, specifically 15d-PGJ2, which is an electrophilic lipid produced by the non-enzymatic dehydration of the prostaglandin PGD2. This study shows that 15d-PGJ2 is also released by Doxo-induced senescent cells as an SASP factor. Treatment of skeletal muscle myoblasts with the conditioned medium from these senescent cells inhibits myoblast fusion during differentiation. Inhibition of L-PTGDS, the enzyme that synthesizes PGD2, diminishes the release of 15d-PGJ2 by senescent cells and restores muscle differentiation. We further show that this lipid post-translationally modifies Cys184 of HRas in C2C12 mouse skeletal myoblasts, causing a reduction in the localization of HRas to the Golgi, increased HRas binding to Ras Binding Domain (RBD) of RAF Kinase (RAF-RBD), and activation of cellular Mitogen Activated Protein (MAP) kinase-Extracellular Signal Regulated Kinase (Erk) signaling (but not the Akt signaling). Mutating C184 of HRas prevents the ability of 15d-PGJ2 to inhibit the differentiation of muscle cells and control the activity of HRas. This work shows that 15d-PGJ2 released from senescent cells could be targeted to restore muscle homeostasis after chemotherapy.
Collapse
Affiliation(s)
- Swarang Sachin Pundlik
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- Manipal Academy of Higher Education (MAHE)ManipalIndia
| | - Alok Barik
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
| | - Ashwin Venkateshvaran
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
| | - Snehasudha Subhadarshini Sahoo
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- University of North Carolina at Chapel HillChapel HillUnited States
| | - Mahapatra Anshuman Jaysingh
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K)MohanpurIndia
- Division of Biology and Biomedical Sciences, Washington University in St LouisSt LouisUnited States
| | | | - Heera Lal
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- Manipal Academy of Higher Education (MAHE)ManipalIndia
| | - Maroof Athar Hashmi
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- Manipal Academy of Higher Education (MAHE)ManipalIndia
| | - Arvind Ramanathan
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
| |
Collapse
|
2
|
Chippalkatti R, Parisi B, Kouzi F, Laurini C, Ben Fredj N, Abankwa DK. RAS isoform specific activities are disrupted by disease associated mutations during cell differentiation. Eur J Cell Biol 2024; 103:151425. [PMID: 38795504 DOI: 10.1016/j.ejcb.2024.151425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/02/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024] Open
Abstract
The RAS-MAPK-pathway is aberrantly regulated in cancer and developmental diseases called RASopathies. While typically the impact of Ras on the proliferation of various cancer cell lines is assessed, it is poorly established how Ras affects cellular differentiation. Here we implement the C2C12 myoblast cell line to systematically study the effect of Ras mutants and Ras-pathway drugs on differentiation. We first provide evidence that a minor pool of Pax7+ progenitors replenishes a major pool of transit amplifying cells that are ready to differentiate. Our data indicate that Ras isoforms have distinct roles in the differentiating culture, where K-Ras depletion increases and H-Ras depletion decreases terminal differentiation. This assay could therefore provide significant new insights into Ras biology and Ras-driven diseases. In line with this, we found that all oncogenic Ras mutants block terminal differentiation of transit amplifying cells. By contrast, RASopathy associated K-Ras variants were less able to block differentiation. Profiling of eight targeted Ras-pathway drugs on seven oncogenic Ras mutants revealed their allele-specific activities and distinct abilities to restore normal differentiation as compared to triggering cell death. In particular, the MEK-inhibitor trametinib could broadly restore differentiation, while the mTOR-inhibitor rapamycin broadly suppressed differentiation. We expect that this quantitative assessment of the impact of Ras-pathway mutants and drugs on cellular differentiation has great potential to complement cancer cell proliferation data.
Collapse
Affiliation(s)
- Rohan Chippalkatti
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette 4362, Luxembourg
| | - Bianca Parisi
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette 4362, Luxembourg
| | - Farah Kouzi
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette 4362, Luxembourg
| | - Christina Laurini
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette 4362, Luxembourg
| | - Nesrine Ben Fredj
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette 4362, Luxembourg
| | - Daniel Kwaku Abankwa
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette 4362, Luxembourg.
| |
Collapse
|
3
|
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
|
4
|
Zhang M, Guo Y, Su R, Corazzin M, Hou R, Xie J, Zhang Y, Zhao L, Su L, Jin Y. Transcriptome analysis reveals the molecular regulatory network of muscle development and meat quality in Sunit lamb supplemented with dietary probiotic. Meat Sci 2022; 194:108996. [PMID: 36195032 DOI: 10.1016/j.meatsci.2022.108996] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 11/27/2022]
Abstract
Supplementing animal feed with probiotic additives can promote muscle production and improve meat quality. The study aimed to explore the effects of dietary probiotics supplementation on the performance, meat quality and muscle transcriptome profile in Sunit lamb. Overall, feeding probiotics significantly increased the body length, LT area, pH24h and intramuscular fat (IMF) content, but decreased cooking loss and meat shear force compared to the control group (P < .05). A total of 651 differentially expressed genes (DEGs) were found in probiotic supplemented lambs. Pathway analysis revealed that DEGs were involved in multiple pathways related to muscle development and fat deposition, such as the ECM-receptor interactions, the MAPK signaling pathway and the FoxO signaling pathway. Therefore, dietary probiotic supplementation can improve muscle development and final meat quality in Sunit lambs by altering gene expression profiles associated with key pathways, providing unique insights into the molecular mechanisms by which dietary probiotics regulate muscle development in the lamb industry.
Collapse
Affiliation(s)
- Min Zhang
- College of Food Science and Engineering, Inner Mongolia Agriculture University, China
| | - Yueying Guo
- College of Food Science and Engineering, Inner Mongolia Agriculture University, China
| | - Rina Su
- Inner Mongolia Vocational College of Chemical Engineering, China
| | - Mirco Corazzin
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Italy
| | - Ran Hou
- College of Food Science and Engineering, Inner Mongolia Agriculture University, China
| | - Jingyu Xie
- College of Food Science and Engineering, Inner Mongolia Agriculture University, China
| | - Yue Zhang
- College of Food Science and Engineering, Inner Mongolia Agriculture University, China
| | - Lihua Zhao
- College of Food Science and Engineering, Inner Mongolia Agriculture University, China
| | - Lin Su
- College of Food Science and Engineering, Inner Mongolia Agriculture University, China
| | - Ye Jin
- College of Food Science and Engineering, Inner Mongolia Agriculture University, China.
| |
Collapse
|
5
|
Barutcu AR, Elizalde G, Gonzalez AE, Soni K, Rinn JL, Wagers AJ, Almada AE. Prolonged FOS activity disrupts a global myogenic transcriptional program by altering 3D chromatin architecture in primary muscle progenitor cells. Skelet Muscle 2022; 12:20. [PMID: 35971133 PMCID: PMC9377060 DOI: 10.1186/s13395-022-00303-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/04/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The AP-1 transcription factor, FBJ osteosarcoma oncogene (FOS), is induced in adult muscle satellite cells (SCs) within hours following muscle damage and is required for effective stem cell activation and muscle repair. However, why FOS is rapidly downregulated before SCs enter cell cycle as progenitor cells (i.e., transiently expressed) remains unclear. Further, whether boosting FOS levels in the proliferating progeny of SCs can enhance their myogenic properties needs further evaluation. METHODS We established an inducible, FOS expression system to evaluate the impact of persistent FOS activity in muscle progenitor cells ex vivo. We performed various assays to measure cellular proliferation and differentiation, as well as uncover changes in RNA levels and three-dimensional (3D) chromatin interactions. RESULTS Persistent FOS activity in primary muscle progenitor cells severely antagonizes their ability to differentiate and form myotubes within the first 2 weeks in culture. RNA-seq analysis revealed that ectopic FOS activity in muscle progenitor cells suppressed a global pro-myogenic transcriptional program, while activating a stress-induced, mitogen-activated protein kinase (MAPK) transcriptional signature. Additionally, we observed various FOS-dependent, chromosomal re-organization events in A/B compartments, topologically associated domains (TADs), and genomic loops near FOS-regulated genes. CONCLUSIONS Our results suggest that elevated FOS activity in recently activated muscle progenitor cells perturbs cellular differentiation by altering the 3D chromosome organization near critical pro-myogenic genes. This work highlights the crucial importance of tightly controlling FOS expression in the muscle lineage and suggests that in states of chronic stress or disease, persistent FOS activity in muscle precursor cells may disrupt the muscle-forming process.
Collapse
Affiliation(s)
- A Rasim Barutcu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Present address: Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Gabriel Elizalde
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alfredo E Gonzalez
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Kartik Soni
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - John L Rinn
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Present address: BioFrontiers and Department of Biochemistry, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Albert E Almada
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, USA.
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Schroer AB, Mohamed JS, Willard MD, Setola V, Oestreich E, Siderovski DP. A role for Regulator of G protein Signaling-12 (RGS12) in the balance between myoblast proliferation and differentiation. PLoS One 2019; 14:e0216167. [PMID: 31408461 PMCID: PMC6691989 DOI: 10.1371/journal.pone.0216167] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/15/2019] [Indexed: 12/28/2022] Open
Abstract
Regulators of G Protein Signaling (RGS proteins) inhibit G protein-coupled receptor (GPCR) signaling by accelerating the GTP hydrolysis rate of activated Gα subunits. Some RGS proteins exert additional signal modulatory functions, and RGS12 is one such protein, with five additional, functional domains: a PDZ domain, a phosphotyrosine-binding domain, two Ras-binding domains, and a Gα·GDP-binding GoLoco motif. RGS12 expression is temporospatially regulated in developing mouse embryos, with notable expression in somites and developing skeletal muscle. We therefore examined whether RGS12 is involved in the skeletal muscle myogenic program. In the adult mouse, RGS12 is expressed in the tibialis anterior (TA) muscle, and its expression is increased early after cardiotoxin-induced injury, suggesting a role in muscle regeneration. Consistent with a potential role in coordinating myogenic signals, RGS12 is also expressed in primary myoblasts; as these cells undergo differentiation and fusion into myotubes, RGS12 protein abundance is reduced. Myoblasts isolated from mice lacking Rgs12 expression have an impaired ability to differentiate into myotubes ex vivo, suggesting that RGS12 may play a role as a modulator/switch for differentiation. We also assessed the muscle regenerative capacity of mice conditionally deficient in skeletal muscle Rgs12 expression (via Pax7-driven Cre recombinase expression), following cardiotoxin-induced damage to the TA muscle. Eight days post-damage, mice lacking RGS12 in skeletal muscle had attenuated repair of muscle fibers. However, when mice lacking skeletal muscle expression of Rgs12 were cross-bred with mdx mice (a model of human Duchenne muscular dystrophy), no increase in muscle degeneration was observed over time. These data support the hypothesis that RGS12 plays a role in coordinating signals during the myogenic program in select circumstances, but loss of the protein may be compensated for within model syndromes of prolonged bouts of muscle damage and repair.
Collapse
Affiliation(s)
- Adam B. Schroer
- Department of Physiology & Pharmacology, WVU School of Medicine, West Virginia University, Morgantown, WV, United States of America
| | - Junaith S. Mohamed
- Division of Exercise Physiology, West Virginia University, Morgantown, WV, United States of America
| | - Melinda D. Willard
- Department of Pharmacology, The University of North Carolina, Chapel Hill, NC, United States of America
| | - Vincent Setola
- Department of Neuroscience, West Virginia University, Morgantown, WV, United States of America
| | - Emily Oestreich
- Department of Pharmacology, The University of North Carolina, Chapel Hill, NC, United States of America
- * E-mail: (EO); (DPS)
| | - David P. Siderovski
- Department of Physiology & Pharmacology, WVU School of Medicine, West Virginia University, Morgantown, WV, United States of America
- * E-mail: (EO); (DPS)
| |
Collapse
|
9
|
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]
|
10
|
The Possible Role of Complete Loss of Myostatin in Limiting Excessive Proliferation of Muscle Cells (C2C12) via Activation of MicroRNAs. Int J Mol Sci 2019; 20:ijms20030643. [PMID: 30717351 PMCID: PMC6386905 DOI: 10.3390/ijms20030643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Myostatin (MSTN) is a member of the TGF-β superfamily that negatively regulates skeletal muscle growth and differentiation. However, the mechanism by which complete MSTN deletion limits excessive proliferation of muscle cells remains unclear. In this study, we knocked out MSTN in mouse myoblast lines using a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9) system and sequenced the mRNA and miRNA transcriptomes. The results show that complete loss of MSTN upregulates seven miRNAs targeting an interaction network composed of 28 downregulated genes, including TGFB1, FOS and RB1. These genes are closely associated with tumorigenesis and cell proliferation. Our study suggests that complete loss of MSTN may limit excessive cell proliferation via activation of miRNAs. These data will contribute to the treatment of rhabdomyosarcoma (RMS).
Collapse
|
11
|
Inhibition of the JNK/MAPK signaling pathway by myogenesis-associated miRNAs is required for skeletal muscle development. Cell Death Differ 2018; 25:1581-1597. [PMID: 29449644 PMCID: PMC6143622 DOI: 10.1038/s41418-018-0063-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 12/24/2017] [Accepted: 01/04/2018] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle differentiation is controlled by multiple cell signaling pathways, however, the JNK/MAPK signaling pathway dominating this process has not been fully elucidated. Here, we report that the JNK/MAPK pathway was significantly downregulated in the late stages of myogenesis, and in contrast to P38/MAPK pathway, it negatively regulated skeletal muscle differentiation. Based on the PAR-CLIP-seq analysis, we identified six elevated miRNAs (miR-1a-3p, miR-133a-3p, miR-133b-3p, miR-206-3p, miR-128-3p, miR-351-5p), namely myogenesis-associated miRNAs (mamiRs), negatively controlled the JNK/MAPK pathway by repressing multiple factors for the phosphorylation of the JNK/MAPK pathway, including MEKK1, MEKK2, MKK7, and c-Jun but not JNK protein itself, and as a result, expression of transcriptional factor MyoD and mamiRs were further promoted. Our study revealed a novel double-negative feedback regulatory pattern of cell-specific miRNAs by targeting phosphorylation kinase signaling cascade responsible for skeletal muscle development.
Collapse
|
12
|
Choi JY, Hwang CY, Lee B, Lee SM, Bahn YJ, Lee KP, Kang M, Kim YS, Woo SH, Lim JY, Kim E, Kwon KS. Age-associated repression of type 1 inositol 1, 4, 5-triphosphate receptor impairs muscle regeneration. Aging (Albany NY) 2017; 8:2062-2080. [PMID: 27658230 PMCID: PMC5076452 DOI: 10.18632/aging.101039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022]
Abstract
Skeletal muscle mass and power decrease with age, leading to impairment of mobility and metabolism in the elderly. Ca2+ signaling is crucial for myoblast differentiation as well as muscle contraction through activation of transcription factors and Ca2+-dependent kinases and phosphatases. Ca2+ channels, such as dihydropyridine receptor (DHPR), two-pore channel (TPC) and inositol 1,4,5-triphosphate receptor (ITPR), function to maintain Ca2+ homeostasis in myoblasts. Here, we observed a significant decrease in expression of type 1 IP3 receptor (ITPR1), but not types 2 and 3, in aged mice skeletal muscle and isolated myoblasts, compared with those of young mice. ITPR1 knockdown using shRNA-expressing viruses in C2C12 myoblasts and tibialis anterior muscle of mice inhibited myotube formation and muscle regeneration after injury, respectively, a typical phenotype of aged muscle. This aging phenotype was associated with repression of muscle-specific genes and activation of the epidermal growth factor receptor (EGFR)-Ras-extracellular signal-regulated kinase (ERK) pathway. ERK inhibition by U0126 not only induced recovery of myotube formation in old myoblasts but also facilitated muscle regeneration after injury in aged muscle. The conserved decline in ITPR1 expression in aged human skeletal muscle suggests utility as a potential therapeutic target for sarcopenia, which can be treated using ERK inhibition strategies.
Collapse
Affiliation(s)
- Jeong Yi Choi
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea.,College of Biological Science and Biotechnology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Chae Young Hwang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea.,Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Bora Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seung-Min Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Young Jae Bahn
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Kwang-Pyo Lee
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Moonkyung Kang
- Graduate School of New Drug Discovery & Development, Chungnam National University, Daejeon 34143, Republic of Korea
| | - Yeon-Soo Kim
- Graduate School of New Drug Discovery & Development, Chungnam National University, Daejeon 34143, Republic of Korea
| | - Sun-Hee Woo
- College of Pharmacy, Chungnam National University, Daejeon 34143, Republic of Korea
| | - Jae-Young Lim
- Department of Rehabilitation Medicine, Seoul National University Bundang Hospital, Gyeonggi-do 13620, Republic of Korea
| | - Eunhee Kim
- College of Biological Science and Biotechnology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Ki-Sun Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
13
|
Cao Y. Tumorigenesis as a process of gradual loss of original cell identity and gain of properties of neural precursor/progenitor cells. Cell Biosci 2017; 7:61. [PMID: 29177029 PMCID: PMC5693707 DOI: 10.1186/s13578-017-0188-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/27/2017] [Indexed: 02/07/2023] Open
Abstract
Cancer is a complex disease without a unified explanation for its cause so far. Our recent work demonstrates that cancer cells share similar regulatory networks and characteristics with embryonic neural cells. Based on the study, I will address the relationship between tumor and neural cells in more details. I collected the evidence from various aspects of cancer development in many other studies, and integrated the information from studies on cancer cell properties, cell fate specification during embryonic development and evolution. Synthesis of the information strongly supports that cancer cells share much more similarities with neural progenitor/stem cells than with mesenchymal-type cells and that tumorigenesis represents a process of gradual loss of cell or lineage identity and gain of characteristics of neural cells. I also discuss cancer EMT, a concept having been under intense debate, and possibly the true meaning of EMT in cancer initiation and development. This synthesis provides fresh insights into a unified explanation for and a previously unrecognized nature of tumorigenesis, which might not be revealed by studies on individual molecular events. The review will also present some brief suggestions for cancer research based on the proposed model of tumorigenesis.
Collapse
Affiliation(s)
- Ying Cao
- Model Animal Research Center and MOE Key Laboratory of Model Animals for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-Tech Zone, Nanjing, 210061 China
| |
Collapse
|
14
|
Gyoja F. Basic helix-loop-helix transcription factors in evolution: Roles in development of mesoderm and neural tissues. Genesis 2017; 55. [PMID: 28804953 DOI: 10.1002/dvg.23051] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/10/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022]
Abstract
Basic helix-loop-helix (bHLH) transcription factors have attracted the attention of developmental and evolutionary biologists for decades because of their conserved functions in mesodermal and neural tissue formation in both vertebrates and fruit flies. Their evolutionary history is of special interest because it will likely provide insights into developmental processes and refinement of metazoan-specific traits. This review briefly considers advances in developmental biological studies on bHLHs/HLHs. I also discuss recent genome-wide surveys and molecular phylogenetic analyses of these factors in a wide range of metazoans. I hypothesize that interactions between metazoan-specific Group A, D, and E bHLH/HLH factors enabled a sophisticated transition system from cell proliferation to differentiation in multicellular development. This control mechanism probably emerged initially to organize a multicellular animal body and was subsequently recruited to form evolutionarily novel tissues, which differentiated during a later ontogenetic phase.
Collapse
Affiliation(s)
- Fuki Gyoja
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Okinawa, 904-0495, Japan.,Department of Biology, Faculty of Science and Engineering, Konan University, 8-9-1 Okamoto, Higashinada, Kobe, 658-8501, Japan
| |
Collapse
|
15
|
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
|
16
|
Truong MTH, Huynh OT, Pham LH, Van Pham P. Direct reprogramming of fibroblasts into endothelial progenitor cells by defined factors. BIOMEDICAL RESEARCH AND THERAPY 2016. [DOI: 10.7603/s40730-016-0038-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
17
|
Kemler D, Dahley O, Roßwag S, Litfin M, Kassel O. The LIM domain protein nTRIP6 acts as a co-repressor for the transcription factor MEF2C in myoblasts. Sci Rep 2016; 6:27746. [PMID: 27292777 PMCID: PMC4904203 DOI: 10.1038/srep27746] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/23/2016] [Indexed: 12/17/2022] Open
Abstract
The transcription factor Myocyte enhancer factor 2C (MEF2C) plays a key role in the late differentiation of skeletal muscle progenitor cells, the so-called myoblasts. During myoblast differentiation, both MEF2C expression and transcriptional activity are regulated. We have reported that nTRIP6, the nuclear isoform of the focal adhesion LIM domain protein TRIP6, acts as an adaptor transcriptional co-activator for several transcription factors. It interacts with the promoter-bound transcription factors and consequently mediates the recruitment of other co-activators. Based on a described interaction between MEF2C and TRIP6 in a yeast-two-hybrid screen, we hypothesised a co-regulatory function of nTRIP6 for MEF2C. In proliferating myoblasts, nTRIP6 interacted with MEF2C and was recruited together with MEF2C to the MEF2-binding regions of the MEF2C target genes Myom2, Mb, Tnni2 and Des. Silencing nTRIP6 or preventing its interaction with MEF2C increased MEF2C transcriptional activity and increased the expression of these MEF2C target genes. Thus, nTRIP6 acts as a co-repressor for MEF2C. Mechanistically, nTRIP6 mediated the recruitment of the class IIa histone deacetylase HDAC5 to the MEF2C-bound promoters. In conclusion, our results unravel a transcriptional co-repressor function for nTRIP6. This adaptor co-regulator can thus exert either co-activator or co-repressor functions, depending on the transcription factor it interacts with.
Collapse
Affiliation(s)
- Denise Kemler
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - Oliver Dahley
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - Sven Roßwag
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - Margarethe Litfin
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| | - Olivier Kassel
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics, Karlsruhe, Germany
| |
Collapse
|
18
|
Hou X, Yang Y, Zhu S, Hua C, Zhou R, Mu Y, Tang Z, Li K. Comparison of skeletal muscle miRNA and mRNA profiles among three pig breeds. Mol Genet Genomics 2015; 291:559-73. [PMID: 26458558 DOI: 10.1007/s00438-015-1126-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/28/2015] [Indexed: 11/30/2022]
Abstract
The pig is an important source of animal protein, and is also an ideal model for human disease. There are significant differences in growth rate, muscle mass, and meat quality between different breeds. To understand the molecular mechanisms underlying porcine skeletal muscle phenotypes, we performed mRNA and miRNA profiling of muscle from three different breeds of pig, Landrace (lean-type), Tongcheng (obese-type), and Wuzhishan (mini-type) by Solexa sequencing. Forty-three genes and 106 miRNAs were differentially expressed between Landrace and Tongcheng pigs, 92 genes and 151 miRNAs were differentially expressed between Tongcheng and Wuzhishan pigs, and 145 genes and 156 miRNAs were differential expressed between Landrace and Wuzhishan pigs. Gene ontology analysis suggested that genes differentially expressed between Landrace and Tongcheng pigs were mainly involved in the biological processes of oxidative stress and muscle organ development. Meanwhile, for Tongcheng vs Wuzhishan and Landrace vs Wuzhishan pigs, the differentially expressed genes were involved in fatty acid metabolism, oxidative stress, muscle contraction, and muscle organ development, processes that are closely related to meat quality. To investigate the molecular mechanisms underlying meat quality diversity based on differentially expressed genes and miRNAs, interaction networks were constructed, according to target prediction results and integration analysis of up-regulated genes with down-regulated miRNAs or down-regulated genes with up-regulated miRNAs. Our findings identify candidate genes and miRNAs associated with muscle development and indicate their potential roles in muscle phenotype variance between different pig breeds. These results serve as a foundation for further studies on muscle development and molecular breeding.
Collapse
Affiliation(s)
- Xinhua Hou
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Yalan Yang
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China.,Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, People's Republic of China
| | - Shiyun Zhu
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Chaoju Hua
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Rong Zhou
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Yulian Mu
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| | - Zhonglin Tang
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China. .,Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, People's Republic of China.
| | - Kui Li
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China
| |
Collapse
|
19
|
|
20
|
Development of genetically flexible mouse models of sarcoma using RCAS-TVA mediated gene delivery. PLoS One 2014; 9:e94817. [PMID: 24733554 PMCID: PMC3986235 DOI: 10.1371/journal.pone.0094817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 03/19/2014] [Indexed: 11/19/2022] Open
Abstract
Sarcomas are a heterogeneous group of mesenchymal malignancies and unfortunately there are limited functional genomics platforms to assess the molecular pathways contributing to sarcomagenesis. Thus, novel model systems are needed to validate which genes should be targeted for therapeutic intervention. We hypothesized that delivery of oncogenes into mouse skeletal muscle using a retroviral (RCAS-TVA) system would result in sarcomagenesis. We also sought to determine if the cell type transformed (mesenchymal progenitors vs. terminally differentiated tissues) would influence sarcoma biology. Cells transduced with RCAS vectors directing the expression of oncoproteins KrasG12D, c-Myc and/or Igf2 were injected into the hindlimbs of mice that expressed the retroviral TVA receptor in neural/mesenchymal progenitors, skeletal/cardiac muscle or ubiquitously (N-tva, AKE and BKE strains respectively). Disrupting the G1 checkpoint CDKN2 (p16/p19-/-) resulted in sarcoma in 30% of p16/p19-/- xN-tva mice with a median latency of 23 weeks (range 8-40 weeks). A similar incidence occurred in p16/p19-/- xBKE mice (32%), however, a shorter median latency (10.4 weeks) was observed. p16/p19-/- xAKE mice also developed sarcomas (24% incidence; median 9 weeks) yet 31% of mice also developed lung sarcomas. Gene-anchored PCR demonstrated retroviral DNA integration in 86% of N-tva, 93% of BKE and 88% of AKE tumors. KrasG12D was the most frequent oncogene isolated. Oncogene delivery by the RCAS-TVA system can generate sarcomas in mice with a defective cell cycle checkpoint. Sarcoma biology differed between the different RCAS models we created, likely due to the cell population being transformed. This genetically flexible system will be a valuable tool for sarcoma research.
Collapse
|
21
|
Abstract
The human Mediator complex is a central integrator for transcription and represents a primary interface that allows DNA-binding transcription factors to communicate their regulatory signals to the RNA polymerase II enzyme. Because Mediator is dynamic both in terms of subunit composition and structure, it presents challenges as a target for small molecule probes. Moreover, little high-resolution structural information exists for Mediator. Its global requirement for transcription, as well as its distinct, transcription factor specific interaction surfaces, however, suggest that development of probes that bind specific Mediator subunits might enable gene- and pathway-specific modulation of transcription. Here we provide a brief overview of the Mediator complex, highlighting biological and structural features that make it an attractive target for molecular probes. We then outline several chemical strategies that might be effective for targeting the complex.
Collapse
Affiliation(s)
| | - Dylan J Taatjes
- Dept. of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80303 USA
| |
Collapse
|
22
|
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
|
23
|
Abstract
Rhabdomyosarcoma (RMS), an aggressive malignant neoplasm that shows features of skeletal muscle, is the most common soft tissue tumor of childhood. In children, the major subtypes are embryonal and alveolar. Although localized disease responds to a multimodal treatment, the prognosis for patients with high-risk features and metastasis remains dismal. Several in vivo models of RMS have been developed in mouse, human xenografts, zebrafish, and Drosophila to better understand the underlying mechanisms governing malignancy. The findings so far have indicated the potential role of skeletal muscle precursor cells in malignant transformation. To better understand histogenesis and different aspects of tumorigenesis in RMS, we have previously developed a robust zebrafish model of kRAS-induced RMS, which shares morphologic and immunophenotypic features with the human counterpart. Cross-species mircroarray comparisons confirm that conserved genetic pathways drive RMS growth. The ease for ex vivo manipulation allows the development of different transgenic and co-injection strategies to induce tumor formation in zebrafish. In contrast to other vertebrate model systems, the tumor onset in zebrafish is short, allowing for efficient study of different tumor processes including tumor growth, self-renewal, and maintenance.
Collapse
|
24
|
Singh S, Vinson C, Gurley CM, Nolen GT, Beggs ML, Nagarajan R, Wagner EF, Parham DM, Peterson CA. Impaired Wnt signaling in embryonal rhabdomyosarcoma cells from p53/c-fos double mutant mice. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2055-66. [PMID: 20829439 DOI: 10.2353/ajpath.2010.091195] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Rhabdomyosarcoma is a primitive neoplasm with a poorly understood etiology that exhibits features of fetal skeletal muscle. It represents the most frequent malignant soft tissue sarcoma affecting the pediatric population and is often treated very aggressively. Embryonal rhabdomyosarcoma (ERMS) and alveolar rhabdomyosarcoma constitute the two major subtypes and exhibit different molecular features. We investigated one potential molecular basis for ERMS by using cells derived from tumors produced in p53(-/-)/c-fos(-/-) mice. This model closely recapitulates the timing, location, molecular markers, and histology seen in human ERMS. A combined chromatin immunoprecipitation/promoter microarray approach was used to identify promoters bound by the c-Jun-containing AP-1 complex in the tumor-derived cells that lacked c-Fos. Identification of the Wnt2 gene and its overexpression in ERMS cells was confirmed in human rhabdomyosarcoma cell lines and prompted further analysis of the Wnt signaling pathway. Contrary to our expectations, the canonical Wnt/β-catenin signaling pathway was down-regulated in ERMS cells compared with normal myoblasts, and activating this pathway promoted myogenic differentiation. Furthermore, the identification of both survivin and sfrp2 through promoter and expression analyses suggested that increased resistance to apoptosis was associated with the inhibition of the Wnt signaling pathway. These results suggest that altered AP-1 activity that leads to the down-regulation of the Wnt pathway may contribute to the inhibition of myogenic differentiation and resistance to apoptosis in ERMS cases.
Collapse
Affiliation(s)
- Shalini Singh
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Proto-oncogenic H-Ras, K-Ras, and N-Ras are involved in muscle differentiation via phosphatidylinositol 3-kinase. Cell Res 2010; 20:919-34. [DOI: 10.1038/cr.2010.92] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
26
|
Bakkar N, Guttridge DC. NF-kappaB signaling: a tale of two pathways in skeletal myogenesis. Physiol Rev 2010; 90:495-511. [PMID: 20393192 DOI: 10.1152/physrev.00040.2009] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
NF-kappaB is a ubiquitiously expressed transcription factor that plays vital roles in innate immunity and other processes involving cellular survival, proliferation, and differentiation. Activation of NF-kappaB is controlled by an IkappaB kinase (IKK) complex that can direct either canonical (classical) NF-kappaB signaling by degrading the IkappaB inhibitor and releasing p65/p50 dimers to the nucleus, or causes p100 processing and nuclear translocation of RelB/p52 via a noncanonical (alternative) pathway. Under physiological conditions, NF-kappaB activity is transiently regulated, whereas constitutive activation of this transcription factor typically in the classical pathway is associated with a multitude of disease conditions, including those related to skeletal muscle. How NF-kappaB functions in muscle diseases is currently under intense investigation. Insight into this role of NF-kappaB may be gained by understanding at a more basic level how this transcription factor contributes to skeletal muscle cell differentiation. Recent data from knockout mice support that the classical NF-kappaB pathway functions as an inhibitor of skeletal myogenesis and muscle regeneration acting through multiple mechanisms. In contrast, alternative NF-kappaB signaling does not appear to be required for myofiber conversion, but instead functions in myotube homeostasis by regulating mitochondrial biogenesis. Additional knowledge of these signaling pathways in skeletal myogenesis should aid in the development of specific inhibitors that may be useful in treatments of muscle disorders.
Collapse
Affiliation(s)
- Nadine Bakkar
- Department of Molecular Virology, Immunology, and Medical Genetics, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | | |
Collapse
|
27
|
Martinelli S, McDowell HP, Vigne SD, Kokai G, Uccini S, Tartaglia M, Dominici C. RAS signaling dysregulation in human embryonal Rhabdomyosarcoma. Genes Chromosomes Cancer 2009; 48:975-82. [DOI: 10.1002/gcc.20702] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
28
|
Pedraza-Alva G, Zingg J, Donda A, Pérez-Martínez L. Estrogen receptor regulates MyoD gene expression by preventing AP-1-mediated repression. Biochem Biophys Res Commun 2009; 389:360-5. [DOI: 10.1016/j.bbrc.2009.08.153] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 08/27/2009] [Indexed: 01/22/2023]
|
29
|
Cardinali B, Castellani L, Fasanaro P, Basso A, Alemà S, Martelli F, Falcone G. Microrna-221 and microrna-222 modulate differentiation and maturation of skeletal muscle cells. PLoS One 2009; 4:e7607. [PMID: 19859555 PMCID: PMC2762614 DOI: 10.1371/journal.pone.0007607] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Accepted: 10/07/2009] [Indexed: 12/16/2022] Open
Abstract
Background MicroRNAs (miRNAs) are a class of small non-coding RNAs that have recently emerged as important regulators of gene expression. They negatively regulate gene expression post-transcriptionally by translational repression and target mRNA degradation. miRNAs have been shown to play crucial roles in muscle development and in regulation of muscle cell proliferation and differentiation. Methodology/Principal Findings By comparing miRNA expression profiling of proliferating myoblasts versus differentiated myotubes, a number of modulated miRNAs, not previously implicated in regulation of myogenic differentiation, were identified. Among these, miR-221 and miR-222 were strongly down-regulated upon differentiation of both primary and established myogenic cells. Conversely, miR-221 and miR-222 expression was restored in post-mitotic, terminally differentiated myotubes subjected to Src tyrosine kinase activation. By the use of specific inhibitors we provide evidence that expression of miR-221 and miR-222 is under the control of the Ras-MAPK pathway. Both in myoblasts and in myotubes, levels of the cell cycle inhibitor p27 inversely correlated with miR-221 and miR-222 expression, and indeed we show that p27 mRNA is a direct target of these miRNAs in myogenic cells. Ectopic expression of miR-221 and miR-222 in myoblasts undergoing differentiation induced a delay in withdrawal from the cell cycle and in myogenin expression, followed by inhibition of sarcomeric protein accumulation. When miR-221 and miR-222 were expressed in myotubes undergoing maturation, a profound alteration of myofibrillar organization was observed. Conclusions/Significance miR-221 and miR-222 have been found to be modulated during myogenesis and to play a role both in the progression from myoblasts to myocytes and in the achievement of the fully differentiated phenotype. Identification of miRNAs modulating muscle gene expression is crucial for the understanding of the circuits controlling skeletal muscle differentiation and maintenance.
Collapse
Affiliation(s)
- Beatrice Cardinali
- Istituto di Biologia Cellulare, Consiglio Nazionale delle Ricerche, Monterotondo, Italy
| | - Loriana Castellani
- Istituto di Biologia Cellulare, Consiglio Nazionale delle Ricerche, Monterotondo, Italy
- Dipartimento di Scienze Motorie e della Salute, Università degli Studi di Cassino, Cassino, Italy
| | | | - Annalisa Basso
- Istituto di Biologia Cellulare, Consiglio Nazionale delle Ricerche, Monterotondo, Italy
| | - Stefano Alemà
- Istituto di Biologia Cellulare, Consiglio Nazionale delle Ricerche, Monterotondo, Italy
| | - Fabio Martelli
- Laboratorio di Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IRCCS, Rome, Italy
| | - Germana Falcone
- Istituto di Biologia Cellulare, Consiglio Nazionale delle Ricerche, Monterotondo, Italy
- * E-mail:
| |
Collapse
|
30
|
Wäsch R, Robbins JA, Cross FR. The emerging role of APC/CCdh1 in controlling differentiation, genomic stability and tumor suppression. Oncogene 2009; 29:1-10. [PMID: 19826416 DOI: 10.1038/onc.2009.325] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Deregulation of the G1/G0 phase of the cell cycle can lead to cancer. During G1, most cells commit alternatively to DNA replication and division, or to cell-cycle exit and differentiation. The anaphase-promoting complex or cyclosome (APC/C) activated by Cdh1 coordinately eliminates positive cell-cycle regulators as well as inhibitors of differentiation, thereby coupling cell-cycle exit and differentiation. Misregulation of Cdh1 thus has the potential to promote both cell-cycle re-entry and either perturbed differentiation or dedifferentiation. In addition, APC/C(Cdh1) is required to maintain genomic stability. As a result, loss of Cdh1 can contribute to tumorigenesis in the form of proliferation of poorly differentiated and genetically unstable cells.
Collapse
Affiliation(s)
- R Wäsch
- Department of Hematology and Oncology, University Medical Center, Freiburg, Germany.
| | | | | |
Collapse
|
31
|
Scholz ME, Meissner JD, Scheibe RJ, Umeda PK, Chang KC, Gros G, Kubis HP. Different roles of H-ras for regulation of myosin heavy chain promoters in satellite cell-derived muscle cell culture during proliferation and differentiation. Am J Physiol Cell Physiol 2009; 297:C1012-8. [PMID: 19625607 DOI: 10.1152/ajpcell.00567.2008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effect of constitutively activated proto-oncogene H-ras (H-rasQ61L) on the regulation of myosin heavy chain (MHC) promoter activities was investigated in rabbit satellite cell-derived muscle cell culture during the proliferation stage and early and later stages of differentiation, respectively. During proliferation, overexpression of H-rasQ61L did not affect basal level of activity of the slow MHCI/beta or the fast MHCIId/x promoter luciferase reporter gene construct in transient transfection assays. By contrast, H-rasQ61L affected both MHC promoter activities during differentiation, and this effect changes from inactivation after 2 days to activation after 4 days of differentiation. The activating effect of H-rasQ61L on both MHC promoters after 4 days of differentiation was significantly reduced by LY-294002, a specific inhibitor of the phosphoinositol-3-kinase (PI3K), a downstream target of Ras. Furthermore, the protein kinase Akt (protein kinase B), a downstream target of PI3k, was activated 4 days after initiation of differentiation in myotubes overexpressing H-rasQ61L. By contrast, inhibition of another Ras downstream pathway, mitogen-activated protein kinase kinase 1/2-extracellular signal-regulated protein kinase 1/2 (MKK1/2-ERK1/2-MAPK), increased activities of both MHC promoters, indicating a suppressive role of this pathway. Moreover, the Ras-PI3K-Akt signaling pathway is involved in the activation of MHCI/beta and IId/x promoters in a later stage of differentiation of muscle cells, presumably by a known inhibiting effect of activated Akt on the MKK1/2-ERK1/2-MAPK pathway. The experiments demonstrate that during differentiation of muscle cells activated H-ras is an important regulator of MHC isoform promoter function with opposite effects during early and later stages.
Collapse
Affiliation(s)
- Michael E Scholz
- Department of Physiology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
32
|
Larsson LG, Anton R, Ivhed I, Öberg F, Pettersson U, Nilsson K. C-Junis Induced to High Continuous Expression During Differentiation of Hematopoietic Cells and is Regulated Independently from C-Fos. Leuk Lymphoma 2009; 4:193-204. [DOI: 10.3109/10428199109068065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
33
|
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
|
34
|
Falcone G, Gauzzi MC, Tatò F, Alemà S. Differential control of muscle-specific gene expression specified by src and myc oncogenes in myogenic cells. CIBA FOUNDATION SYMPOSIUM 2007; 150:250-8; discussion 258-61. [PMID: 2115425 DOI: 10.1002/9780470513927.ch15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Myogenic cells can be transformed in vitro by the introduction of several exogenous viral oncogenes. Transformed myoblasts are prevented from terminal differentiation into myotubes by the continuous expression of oncogenes such as myc and src, chosen as prototypes of nuclear and cytoplasmic oncogenes. A comparative analysis of the relationship between transformation and differentiation in myoblasts and cells belonging to other lineages has led to the proposal that terminal differentiation of myc-transformed quail myoblasts is indirectly prevented by the loss of growth control and that myc-bearing cells remain susceptible to growth regulation by interaction with adjacent normal cells. On the contrary, the src oncogene appears to affect expression of the myogenic programme via a direct mechanism, independent from abnormal growth control. There is increasing evidence for the existence of master regulatory genes that govern and influence muscle development in vivo and myogenic differentiation in vitro. Expression of cytoplasmic oncogenes such as src, ras and polyoma middle T in the mouse myogenic cell line, C2, results in inhibition of biochemical differentiation and a marked down-regulation of the MyoD1 and myogenin genes.
Collapse
Affiliation(s)
- G Falcone
- Istituto di Biologia Cellulare, C.N.R., Roma, Italy
| | | | | | | |
Collapse
|
35
|
Parker TG, Chow KL, Schwartz RJ, Schneider MD. TGF-beta 1 and fibroblast growth factors selectively up-regulate tissue-specific fetal genes in cardiac muscle cells. CIBA FOUNDATION SYMPOSIUM 2007; 157:152-60; discussion 161-4. [PMID: 1712696 DOI: 10.1002/9780470514061.ch10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
TGF-beta 1, like basic and acidic fibroblast growth factor (FGF), inhibits differentiated gene expression in skeletal myoblasts. It potentiates FGF-beta 1 down-regulated expression of the alpha-myosin heavy chain gene and the sarcoplasmic reticulum calcium ATPase gene, yet up-regulated expression of the genes for beta-myosin heavy chain, atrial natriuretic factor, and both skeletal and smooth muscle alpha-actin-four transcripts associated with the embryonic heart. TGF-beta 1 did not affect cardiac alpha-actin gene expression. These responses resemble the generalized 'fetal' phenotype seen during hypertrophy triggered by a haemodynamic load. Chick skeletal and cardiac alpha-actin promoter-driven reported genes were transfected into neonatal rat cardiac myocytes. TGF-beta 1 stimulated skeletal alpha-actin transcription, but not transcription from the cardiac alpha-actin promoter. Basic FGF produced the same results as TGF-beta 1, but acidic FGF suppressed expression of both alpha-actin genes; these results were true for purified and recombinant FGFs. Modulation of alpha-actin transcription by growth factors corresponded accurately to control of the endogenous genes. Three positive cis-acting elements were critical for skeletal alpha-actin transcription in cardiac, as well as skeletal, myocytes, particularly the downstream CCAAT box-associated repeat. Thus, TGF-beta 1 and FGFs selectively induce an ensemble of 'fetal' genes and differentially regulate alpha-actin transcription in cardiac muscle cells.
Collapse
Affiliation(s)
- T G Parker
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030-3498
| | | | | | | |
Collapse
|
36
|
Ciuffini L, Castellani L, Salvati E, Galletti S, Falcone G, Alemà S. Delineating v-Src downstream effector pathways in transformed myoblasts. Oncogene 2007; 27:528-39. [PMID: 17637741 DOI: 10.1038/sj.onc.1210665] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In this study, we delineate the intracellular signalling pathways modulated by a conditional v-Src tyrosine kinase that lead to unrestrained proliferation and block of differentiation of primary avian myoblasts. By inhibiting Ras-MAPK kinase and phosphatidylinositol 3-kinase with different means, we find that both pathways play crucial roles in controlling v-Src-sustained growth factor and anchorage independence for proliferation. The Ras-MAPK kinase pathway also contributes to block of differentiation independently of cell proliferation since inhibition of this pathway both in proliferating and growth-arrested v-Src-transformed myoblasts induces expression of muscle-specific genes, fusion into multinucleated myotubes and assembly of specialized contractile structures. Importantly, we find that the p38 MAPK pathway is inhibited by v-Src in myoblasts and its forced activation results in growth inhibition and expression of differentiation, indicating p38 MAPK as a critical target of v-Src in growth transformation and myogenic differentiation. Furthermore, we show that downregulation of p38 MAPK activation may occur via Ras-MAPK kinase, thus highlighting a cross-regulation between the two pathways. Finally, we report that the simultaneous inhibition of MAPK kinase and calpain, combined to activation of p38 MAPK, are sufficient to reconstitute largely the differentiation potential of v-Src-transformed myoblasts.
Collapse
Affiliation(s)
- L Ciuffini
- Istituto di Biologia Cellulare, Consiglio Nazionale delle Ricerche, Monterotondo Scalo (RM), Italy
| | | | | | | | | | | |
Collapse
|
37
|
Yokoyama T, Takano K, Yoshida A, Katada F, Sun P, Takenawa T, Andoh T, Endo T. DA-Raf1, a competent intrinsic dominant-negative antagonist of the Ras-ERK pathway, is required for myogenic differentiation. ACTA ACUST UNITED AC 2007; 177:781-93. [PMID: 17535970 PMCID: PMC2064279 DOI: 10.1083/jcb.200703195] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ras activates Raf, leading to the extracellular-regulated kinase (ERK)–mitogen-activated protein kinase pathway, which is involved in a variety of cellular, physiological, and pathological responses. Thus, regulators of this Ras–Raf interaction play crucial roles in these responses. In this study, we report a novel regulator of the Ras–Raf interaction named DA-Raf1. DA-Raf1 is a splicing isoform of A-Raf with a wider tissue distribution than A-Raf. It contains the Ras-binding domain but lacks the kinase domain, which is responsible for activation of the ERK pathway. As inferred from its structure, DA-Raf1 bound to activated Ras as well as M-Ras and interfered with the ERK pathway. The Ras–ERK pathway is essential for the negative regulation of myogenic differentiation induced by growth factors. DA-Raf1 served as a positive regulator of myogenic differentiation by inducing cell cycle arrest, the expression of myogenin and other muscle-specific proteins, and myotube formation. These results imply that DA-Raf1 is the first identified competent, intrinsic, dominant-negative antagonist of the Ras–ERK pathway.
Collapse
Affiliation(s)
- Takashi Yokoyama
- Department of Biology, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
van der Burgt I, Kupsky W, Stassou S, Nadroo A, Barroso C, Diem A, Kratz CP, Dvorsky R, Ahmadian MR, Zenker M. Myopathy caused by HRAS germline mutations: implications for disturbed myogenic differentiation in the presence of constitutive HRas activation. J Med Genet 2007; 44:459-62. [PMID: 17412879 PMCID: PMC2598013 DOI: 10.1136/jmg.2007.049270] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Rare reports on patients with congenital myopathy with excess of muscle spindles (CMEMS), hypertrophic cardiomyopathy and variable features resembling Noonan syndrome have been published, but the genetic basis of this condition is so far unknown. METHODS AND RESULTS We analysed PTPN11 and RAS genes in five unrelated patients with this phenotype, and found HRAS mutations in four of them. Two disease-associated mutations, G12V and G12S, have previously been observed in patients with Costello syndrome (CS), and two other mutations, E63K and Q22K, are novel. All four mutations are predicted to enhance downstream HRas signalling, suggesting that CMEMS is a developmental consequence of sustained HRas activation in skeletal muscle. CONCLUSION This type of myopathy may represent a previously unrecognized manifestation of CS. However, some patients carrying HRAS mutations may exhibit prominent congenital muscular dysfunction, although features of CS may be less obvious, suggesting that germline HRAS mutations may underlie some cases of otherwise unclassified neonatal neuromuscular disorders.
Collapse
|
39
|
Tsumura H, Yoshida T, Saito H, Imanaka-Yoshida K, Suzuki N. Cooperation of oncogenic K-ras and p53 deficiency in pleomorphic rhabdomyosarcoma development in adult mice. Oncogene 2006; 25:7673-9. [PMID: 16785989 DOI: 10.1038/sj.onc.1209749] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human rhabdomyosarcomas (RMSs) frequently demonstrate genetic alterations in ras and p53. To investigate their possible involvement in the tumorigenesis, we generated a knock-in mouse line with oncogenic K-ras, conditionally expressed by Cre/LoxP system on a background of p53 alteration. Electroporation of Cre expression vector in skeletal muscle tissues resulted in the generation of tumor in adults with tumor incidences of 100% at 10 weeks and 40% at 15 weeks, in p53(-/-) and p53(-/+) backgrounds, respectively. The tumor histology was pleomorphic RMS with characteristic bizarre giant cells, positive for desmin and alpha-sarcomeric actin and exhibiting remarkable increase in total and phosphorylated extracellular signal-regulated protein kinase (ERK)1 and ERK2. Loss of the wild-type p53 was detected in K-rasG12V-expressed tumors of p53(-/+) mice. Early lesions 3 weeks after electroporation consisted of proliferating populations of myogenic progenitors, including stem cells positive for ScaI antigen, immature cells positive for desmin and neural cell adhesion molecule-positive myotubes. Thus, cooperation of oncogenic K-ras and p53 deficiency resulted in the development of pleomorphic RMS in adult mice, providing a useful mouse model for further detailed studies.
Collapse
Affiliation(s)
- H Tsumura
- Department of Animal Genomics, Functional Genomics Institute, Mie University Life Science Research Center, Tsu, Mie, Japan
| | | | | | | | | |
Collapse
|
40
|
Dogra C, Changotra H, Mohan S, Kumar A. Tumor Necrosis Factor-like Weak Inducer of Apoptosis Inhibits Skeletal Myogenesis through Sustained Activation of Nuclear Factor-κB and Degradation of MyoD Protein. J Biol Chem 2006; 281:10327-36. [PMID: 16461349 DOI: 10.1074/jbc.m511131200] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
In this study we have investigated the effect and the mechanisms by which tumor necrosis factor-like weak inducer of apoptosis (TWEAK) modulates myogenic differentiation. Treatment of C2C12 myoblasts with TWEAK inhibited their differentiation evident by a decrease in the expression of creatine kinase, myosin heavy chain-fast twitch, myogenin, and the formation of multinucleated myotubes. TWEAK also inhibited the differentiation of mouse primary myoblasts. Conversely, the proliferation of C2C12 myoblasts and the expression of a cell-cycle regulator cyclin D1 were increased in response to TWEAK treatment. Inhibition of cellular proliferation using hydroxyurea only partially reversed the inhibitory effect of TWEAK on myogenic differentiation. Treatment of C2C12 myoblasts with TWEAK resulted in the activation of nuclear factor-kappaB (NF-kappaB), the (IkappaB) IkappaB kinase (IKK) complex, and the phosphorylation and degradation of IkappaBalpha protein. Inhibition of NF-kappaB activity by overexpression of a dominant negative mutant of IkappaBalpha (IkappaBalphaDeltaN) significantly increased the myogenic differentiation in TWEAK-treated C2C12 cultures. Furthermore, overexpression of a dominant negative mutant of IKKbeta (IKKbetaK44A) but not IKKalpha (IKKalphaK44M) reversed the inhibitory effect of TWEAK on myogenesis. TWEAK inhibited the expression of myogenic regulatory factors MyoD and myogenin and also induced the degradation of MyoD protein. Finally, inhibition of NF-kappaB activation through overexpression of IKKbetaK44A prevented the degradation of MyoD protein. Overall, our data suggest that TWEAK inhibits myogenesis through the activation of NF-kappaB signaling pathway and degradation of MyoD protein.
Collapse
Affiliation(s)
- Charu Dogra
- Molecular Genetics Division, Musculoskeletal Disease Center, Jerry L. Pettis Veterans Administration Medical Center, 11201 Benton Street (151), Loma Linda, CA 92357, USA
| | | | | | | |
Collapse
|
41
|
Kitzmann M, Bonnieu A, Duret C, Vernus B, Barro M, Laoudj-Chenivesse D, Verdi JM, Carnac G. Inhibition of Notch signaling induces myotube hypertrophy by recruiting a subpopulation of reserve cells. J Cell Physiol 2006; 208:538-48. [PMID: 16741964 DOI: 10.1002/jcp.20688] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
During muscle differentiation, a population of quiescent undifferentiated myoblasts (reserve cells) emerges among mature muscle cells. However, the molecular mechanisms underlying such cell segregation and the characterization of this subpopulation of myoblasts remain to be determined. Notch is known to control the behavior and fate of murine muscle stem cells. In this study, we examined the role of Notch in myoblast segregation. We showed that inhibition of Notch activity by either overexpressing Numb or by using a pharmacological gamma-secretase inhibitor (DAPT) enhanced differentiation of murine and human myoblasts. This effect was not restricted to in vitro culture systems since DAPT-treated zebrafish embryos also showed increased differentiation. Using C2.7 myoblasts as a model, we showed that inhibition of Notch induced myotube hypertrophy by recruiting reserve cells that do not normally fuse. We further showed that endogenous Notch-signaling components were differentially expressed and activated in reserve cells with respect to Notch 1 and CD34 expression. We identified CD34 negative reserve cells as the subpopulation of myoblasts recruited to fuse into myotubes during differentiation in response to Notch inhibition. Therefore, we showed here that the activation of Notch 1 is important to maintain a subpopulation of CD34 negative reserve cells in an undifferentiated state.
Collapse
Affiliation(s)
- Magali Kitzmann
- Adult stem cells and facioscapulohumeral dystrophy," CNRS FRE2593, 1919 route de Mende, 34293 Montpellier 5, France
| | | | | | | | | | | | | | | |
Collapse
|
42
|
McIntyre GI. Cell hydration as the primary factor in carcinogenesis: A unifying concept. Med Hypotheses 2005; 66:518-26. [PMID: 16271440 DOI: 10.1016/j.mehy.2005.09.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2005] [Revised: 09/09/2005] [Accepted: 09/12/2005] [Indexed: 11/18/2022]
Abstract
The paper discusses the unifying concept that cell hydration is the primary factor in the mechanism of carcinogenesis. The concept includes the following hypotheses: (1) Increased cell hydration causes cancer not only by promoting cell division and oncogene expression, but also by inactivating genes inducing cell differentiation, and by preventing apoptosis. Conversely, factors that reduce cell hydration prevent cancer by inhibiting cell division and oncogene expression, while activating genes inducing cell differentiation, and by promoting apoptosis. The unique ability of cell hydration to have these opposite effects on cell behavior and gene expression can account for its postulated role as the primary factor in both the promotion and prevention of cancer. (2) A progressive increase in cell hydration, induced by successive mutations and/or epigenetic changes, is the basic mechanism of multi-step carcinogenesis, the degree of malignancy increasing with the degree of cell hydration. (3) The increased hydration of cancer cells accelerates their respiration rate, thereby enhancing their ability to compete for nutrients with their normal counterparts. This effect may play a major role in promoting tumor growth and in the postulated mechanism of multi-step carcinogenesis. (4) Increased cell hydration is also proposed as an alternative or additional explanation of the carcinogenetic effect of inflammatory agents and of hormones. A survey of the literature provides evidence consistent with these hypotheses, but suggestions are included for further investigations to test their validity and their implications. From a clinical perspective, the abnormally high water content of cancer cells permits the use of microwave technology for tumor detection and treatment. Also of considerable therapeutic significance is the increased sensitivity if cancer cells to desiccation, postulated to result from genetic changes induced by increased hydration. This may well be the achilles heel of cancer, and recent investigations indicate that it may be exploited very effectively in the treatment of the disease. In conclusion, I suggest that the need for studies on the molecular biology of cancer to be supplemented by more information on environmental effects on gene expression and on the biochemical and physiological factors that mediate genetic effects at the cellular level. This approach might also be used to assess the validity of the postulated role of cell hydration as a factor of particular significance.
Collapse
|
43
|
Jethanandani P, Kramer RH. Alpha7 integrin expression is negatively regulated by deltaEF1 during skeletal myogenesis. J Biol Chem 2005; 280:36037-46. [PMID: 16129691 DOI: 10.1074/jbc.m508698200] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Alpha7 integrin levels increase dramatically as myoblasts differentiate to myotubes. A negative regulatory element with putative sites for deltaEF1 is present in the alpha7 proximal promoter region. To define the role of deltaEF1 in regulating alpha7 integrin expression, we overexpressed deltaEF1 in C2C12 myoblasts. This resulted in a major down-regulation of alpha7 protein expression. Promoter assays revealed that C2C12 myoblasts transfected with deltaEF1 showed a decrease in activity of the 2.8-kb alpha7 promoter fragment, indicating regulation of alpha7 integrin at the transcriptional level. We have identified two E-box-like sites for deltaEF1 in the negative regulatory region. Mutation of these sites enhanced alpha7 promoter activity, indicating that these sites function in repression. MYOD, an activator of alpha7 integrin transcription, can compete with deltaEF1 for binding at these sites in gel shift assay. By using chromatin immunoprecipitation, we demonstrated a reciprocal binding of deltaEF1 and MYOD to this regulatory element depending on the stage of differentiation: deltaEF1 is preferentially bound in myoblasts to this region, whereas MYOD is bound in myotubes. The N-terminal region of deltaEF1 is necessary for alpha7 repression, and this region also binds the co-activator p300/CBP. Importantly, we found that the p300/CBP co-activator can overcome repression by deltaEF1, suggesting that deltaEF1 can titrate limiting amounts of this co-activator. These findings suggest that deltaEF1 has a role in suppressing integrin expression in myoblasts by displacing MYOD and competing for p300/CBP co-activator.
Collapse
Affiliation(s)
- Poonam Jethanandani
- Department of Cell and Tissue Biology, University of California, San Francisco, California 94143-0640, USA
| | | |
Collapse
|
44
|
Cole F, Zhang W, Geyra A, Kang JS, Krauss RS. Positive regulation of myogenic bHLH factors and skeletal muscle development by the cell surface receptor CDO. Dev Cell 2005; 7:843-54. [PMID: 15572127 DOI: 10.1016/j.devcel.2004.10.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2003] [Revised: 06/26/2004] [Accepted: 09/22/2004] [Indexed: 12/21/2022]
Abstract
Skeletal myogenesis is controlled by bHLH transcription factors of the MyoD family that, along with MEF-2 factors, comprise a positive feedback network that maintains the myogenic transcriptional program. Cell-cell contact between muscle precursors promotes myogenesis, but little is known of the underlying mechanisms. CDO, an Ig superfamily member, is a component of a cell surface receptor complex found at sites of cell-cell contact that positively regulates myogenesis in vitro. We report here that mice lacking CDO display delayed skeletal muscle development. Additionally, satellite cells from these mice differentiate defectively in vitro. CDO functions to activate myogenic bHLH factors via enhanced heterodimer formation, most likely by inducing hyperphosphorylation of E proteins. The Cdo gene is, in turn, a target of MyoD. The promyogenic effect of cell-cell contact is therefore linked to the activity of myogenic bHLH factors. Furthermore, the myogenic positive feedback network extends from the cell surface to the nucleus.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Blotting, Western
- Cell Adhesion Molecules/metabolism
- Cell Adhesion Molecules/physiology
- Cell Communication
- Cell Line
- Cell Membrane/metabolism
- Cell Nucleus/metabolism
- Cells, Cultured
- Dimerization
- Genes, Reporter
- Immunohistochemistry
- Immunoprecipitation
- In Situ Hybridization
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred C3H
- Models, Biological
- Muscle, Skeletal/cytology
- Muscle, Skeletal/metabolism
- MyoD Protein/metabolism
- Phosphoric Monoester Hydrolases/metabolism
- Phosphorylation
- Protein Processing, Post-Translational
- Protein Structure, Tertiary
- RNA/metabolism
- Time Factors
- Transfection
- Transgenes
- Tumor Suppressor Proteins/metabolism
- Tumor Suppressor Proteins/physiology
Collapse
Affiliation(s)
- Francesca Cole
- Brookdale Department of Molecular, Cell, and Developmental Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | |
Collapse
|
45
|
|
46
|
Azmi S, Ozog A, Taneja R. Sharp-1/DEC2 inhibits skeletal muscle differentiation through repression of myogenic transcription factors. J Biol Chem 2004; 279:52643-52. [PMID: 15448136 DOI: 10.1074/jbc.m409188200] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Skeletal muscle differentiation is regulated by the basic-helix-loop-helix (bHLH) family of transcription factors. The myogenic bHLH factors form heterodimers with the ubiquitously expressed bHLH E-proteins and bind E-box (CANNTG) sites present in the promoters of several muscle-specific genes. Our previous studies have shown that the bHLH factor Sharp-1 is expressed in skeletal muscle and interacts with MyoD and E-proteins. However, its role in regulation of myogenic differentiation remains unknown. We report here that endogenous Sharp-1 is expressed in proliferating C2C12 myoblasts and is down-regulated during myogenic differentiation. Constitutive expression of Sharp-1 in C2C12 myoblasts promotes cell cycle exit causing a decrease in cyclin D1 expression but blocks terminal differentiation. Although MyoD expression is not inhibited, the induction of differentiation-specific genes such as myogenin, MEF2C, and myosin heavy chain is impaired by Sharp-1 overexpression. We demonstrate that the interaction of Sharp-1 with MyoD and E-proteins results in reduced DNA binding and transactivation from MyoD-dependent E-box sites. Re-expression of MyoD approximately E47 rescues the differentiation defect imposed by Sharp-1, suggesting that myogenic bHLH factors function downstream of Sharp-1. Our data suggest that protein-protein interactions between Sharp-1, MyoD, and E47 resulting in interference with MyoD function underlies Sharp-1-mediated repression of myogenic differentiation.
Collapse
Affiliation(s)
- Sameena Azmi
- Brookdale Department of Molecular, Cell, and Developmental Biology, Mount Sinai School of Medicine, New York, New York 10029-6574, USA
| | | | | |
Collapse
|
47
|
Nishiyama T, Kii I, Kudo A. Inactivation of Rho/ROCK signaling is crucial for the nuclear accumulation of FKHR and myoblast fusion. J Biol Chem 2004; 279:47311-9. [PMID: 15322110 DOI: 10.1074/jbc.m403546200] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Myoblast fusion is a critical process for the terminal differentiation of skeletal muscle. To elucidate the intracellular mechanisms regulating myoblast fusion, we studied the roles of signaling through the small GTPase Rho and its effector, the Rho-associated kinase ROCK, in myoblast fusion of mouse C2C12 cells. We found that Rho activity, which was high in proliferating myoblasts, decreased during myogenesis. Expression of a constitutively active form of Rho blocked myoblast fusion, but not the earlier steps of differentiation. Consistently, ROCK activity was also decreased in differentiating C2C12 cells, and an active ROCK mutant prevented their fusion. Furthermore, inactivation of ROCK by the specific inhibitor Y-27632 enhanced myoblast fusion, even in cells expressing the active Rho mutant. Thus, the down-regulation of Rho/ROCK signaling is required for myoblast fusion. We also found that Rho/ROCK signaling was required for retaining FKHR, a transcription factor implicated in myoblast fusion, in the cytoplasm and that inactivation of ROCK was essential for the nuclear accumulation of FKHR that took place just before the onset of myoblast fusion. Moreover, ROCK directly phosphorylated FKHR in vitro. We conclude that the inactivation of Rho/ROCK signaling is a prerequisite for FKHR nuclear translocation and myoblast fusion in C2C12 cells, providing evidence for a novel regulatory role of Rho/ROCK signaling in myogenic differentiation.
Collapse
Affiliation(s)
- Tomoko Nishiyama
- Department of Biological Information, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| | | | | |
Collapse
|
48
|
Ait-Si-Ali S, Guasconi V, Fritsch L, Yahi H, Sekhri R, Naguibneva I, Robin P, Cabon F, Polesskaya A, Harel-Bellan A. A Suv39h-dependent mechanism for silencing S-phase genes in differentiating but not in cycling cells. EMBO J 2004; 23:605-15. [PMID: 14765126 PMCID: PMC1271807 DOI: 10.1038/sj.emboj.7600074] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2003] [Accepted: 12/17/2003] [Indexed: 12/18/2022] Open
Abstract
The Rb/E2F complex represses S-phase genes both in cycling cells and in cells that have permanently exited from the cell cycle and entered a terminal differentiation pathway. Here we show that S-phase gene repression, which involves histone-modifying enzymes, occurs through distinct mechanisms in these two situations. We used chromatin immunoprecipitation to show that methylation of histone H3 lysine 9 (H3K9) occurs at several Rb/E2F target promoters in differentiating cells but not in cycling cells. Furthermore, phenotypic knock-down experiments using siRNAs showed that the histone methyltransferase Suv39h is required for histone H3K9 methylation and subsequent repression of S-phase gene promoters in differentiating cells, but not in cycling cells. These results indicate that the E2F target gene permanent silencing mechanism that is triggered upon terminal differentiation is distinct from the transient repression mechanism in cycling cells. Finally, Suv39h-depleted myoblasts were unable to express early or late muscle differentiation markers. Thus, appropriately timed H3K9 methylation by Suv39h seems to be part of the control switch for exiting the cell cycle and entering differentiation.
Collapse
Affiliation(s)
- Slimane Ait-Si-Ali
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
| | - Valentina Guasconi
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
| | - Lauriane Fritsch
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
| | - Hakima Yahi
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
| | - Redha Sekhri
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
| | - Irina Naguibneva
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
| | - Philippe Robin
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
| | - Florence Cabon
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
| | - Anna Polesskaya
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
| | - Annick Harel-Bellan
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, Villejuif, France
- UPR 9079 CNRS-Labellisée Ligue Nationale Contre le Cancer, Institut André Lwoff, 7 rue Guy Moquet, 94800 Villejuif, France. Tel.: +33 149 583385; Fax: +33 149 583307; E-mail:
| |
Collapse
|
49
|
Falcone G, Ciuffini L, Gauzzi MC, Provenzano C, Strano S, Gallo R, Castellani L, Alemà S. v-Src inhibits myogenic differentiation by interfering with the regulatory network of muscle-specific transcriptional activators at multiple levels. Oncogene 2004; 22:8302-15. [PMID: 14614454 DOI: 10.1038/sj.onc.1206915] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The conversion of skeletal myoblasts to terminally differentiated myocytes is negatively controlled by several growth factors and oncoproteins. In this study, we have investigated the molecular mechanisms by which v-Src, a prototypic tyrosine kinase, perturbs myogenesis in primary avian myoblasts and in established murine C2C12 satellite cells. We determined the expression levels of the cell cycle regulators pRb, cyclin D1 and D3 and cyclin-dependent kinase inhibitors p21 and p27 in v-Src-transformed myoblasts and found that, in contrast to myogenin, they are normally modulated by differentiative cues, implying that v-Src affects myogenesis independent of cell proliferation. We then examined the levels of expression, DNA-binding ability and transcription-activation potentials of myogenic regulatory factors in transformed myoblasts and in myotubes after reactivation of a temperature-sensitive allele of v-Src. Our results reveal two distinct potential modes of repression targeted to myogenic factors. On the one hand, we show that v-Src reversibly inhibits the expression of MyoD and myogenin in C2C12 cells and of myogenin in quail myoblasts. Remarkably, these loci become resistant to activation of the kinase in the postmitotic compartment. On the other hand, we demonstrate that v-Src efficiently inhibits muscle gene expression by repressing the transcriptional activity of myogenic factors without affecting MyoD DNA-binding activity. Indeed, forced expression of MyoD and myogenin allows terminal differentiation of transformed myoblasts. Finally, we found that ectopic expression of the coactivator p300 restores transcription from extrachromosomal muscle-specific promoters.
Collapse
Affiliation(s)
- Germana Falcone
- Istituto di Biologia Cellulare, Consiglio Nazionale delle Ricerche, Monterotondo 00016, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
McBride K, Charron F, Lefebvre C, Nemer M. Interaction with GATA transcription factors provides a mechanism for cell-specific effects of c-Fos. Oncogene 2003; 22:8403-12. [PMID: 14627981 DOI: 10.1038/sj.onc.1206877] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
c-Fos is a multifunctional transcription factor that is involved in cellular proliferation, differentiation and apoptosis. c-Fos is rapidly induced by a variety of hormones, growth factors and other extracellular stimuli, resulting in cell-specific responses. One potential mechanism underlying the cell-specific effects of c-Fos may be its ability to regulate gene expression through interaction with tissue-restricted transcription factors. We report here that c-Fos interacts with the cell-specific GATA proteins to potentiate their ability to transactivate target promoters, via GATA-binding sites. c-Fos is recruited to GATA proteins through direct interaction with their N-terminal activation domain. Neither the leucine zipper nor the DNA-binding domain of c-Fos is required for physical interaction with GATA proteins. Instead, a C-terminal domain located between amino acids 235 and 296, which is conserved in FosB but not in the nontransforming Fos family members, FosB/SF or Fra-1, is essential for c-Fos-GATA interaction. These data suggest that c-Fos may act as an inducible cofactor for cell-specific transcription factors and unravel a novel mechanism for transcriptional regulation by c-Fos, independent of the well-studied AP-1 pathway. The results also raise the possibility that dysregulated interaction with cell-specific transcription factors may be an important component in cellular transformation by nuclear oncogenes.
Collapse
Affiliation(s)
- Kevin McBride
- Laboratoire de Développement et différenciation cardiaques, Institut de recherches cliniques de Montréal, 110, avenue des Pins Ouest, Montréal, QC, Canada H2W 1R7
| | | | | | | |
Collapse
|