1
|
Rauen KA, Tidyman WE. RASopathies - what they reveal about RAS/MAPK signaling in skeletal muscle development. Dis Model Mech 2024; 17:dmm050609. [PMID: 38847227 PMCID: PMC11179721 DOI: 10.1242/dmm.050609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024] Open
Abstract
RASopathies are rare developmental genetic syndromes caused by germline pathogenic variants in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) signal transduction pathway. Although the incidence of each RASopathy syndrome is rare, collectively, they represent one of the largest groups of multiple congenital anomaly syndromes and have severe developmental consequences. Here, we review our understanding of how RAS/MAPK dysregulation in RASopathies impacts skeletal muscle development and the importance of RAS/MAPK pathway regulation for embryonic myogenesis. We also discuss the complex interactions of this pathway with other intracellular signaling pathways in the regulation of skeletal muscle development and growth, and the opportunities that RASopathy animal models provide for exploring the use of pathway inhibitors, typically used for cancer treatment, to correct the unique skeletal myopathy caused by the dysregulation of this pathway.
Collapse
Affiliation(s)
- Katherine A Rauen
- Department of Pediatrics, Division of Genomic Medicine, University of California Davis, Sacramento, CA, 95817, USA
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| | - William E Tidyman
- University of California Davis MIND Institute, Sacramento, CA 95817, USA
| |
Collapse
|
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
|
Tidyman WE, Goodwin AF, Maeda Y, Klein OD, Rauen KA. MEK-inhibitor-mediated rescue of skeletal myopathy caused by activating Hras mutation in a Costello syndrome mouse model. Dis Model Mech 2022; 15:272258. [PMID: 34553752 PMCID: PMC8617311 DOI: 10.1242/dmm.049166] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 09/13/2021] [Indexed: 11/20/2022] Open
Abstract
Costello syndrome (CS) is a congenital disorder caused by heterozygous activating germline HRAS mutations in the canonical Ras/mitogen-activated protein kinase (Ras/MAPK) pathway. CS is one of the RASopathies, a large group of syndromes caused by mutations within various components of the Ras/MAPK pathway. An important part of the phenotype that greatly impacts quality of life is hypotonia. To gain a better understanding of the mechanisms underlying hypotonia in CS, a mouse model with an activating HrasG12V allele was utilized. We identified a skeletal myopathy that was due, in part, to inhibition of embryonic myogenesis and myofiber formation, resulting in a reduction in myofiber size and number that led to reduced muscle mass and strength. In addition to hyperactivation of the Ras/MAPK and PI3K/AKT pathways, there was a significant reduction in p38 signaling, as well as global transcriptional alterations consistent with the myopathic phenotype. Inhibition of Ras/MAPK pathway signaling using a MEK inhibitor rescued the HrasG12V myopathy phenotype both in vitro and in vivo, demonstrating that increased MAPK signaling is the main cause of the muscle phenotype in CS. Summary: A Costello syndrome (CS) mouse model carrying a heterozygous Hras p.G12V mutation was utilized to investigate Ras pathway dysregulation, revealing that increased MAPK signaling is the main cause of the muscle phenotype in CS.
Collapse
Affiliation(s)
- William E Tidyman
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA.,UC Davis MIND Institute, Sacramento, CA 95817, USA
| | - Alice F Goodwin
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA 94143, USA
| | - Yoshiko Maeda
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA.,UC Davis MIND Institute, Sacramento, CA 95817, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA 94143, USA.,Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA 94143, USA
| | - Katherine A Rauen
- Department of Pediatrics, University of California Davis, Sacramento, CA 95817, USA.,UC Davis MIND Institute, Sacramento, CA 95817, USA
| |
Collapse
|
4
|
Yohe ME, Gryder BE, Shern JF, Song YK, Chou HC, Sindiri S, Mendoza A, Patidar R, Zhang X, Guha R, Butcher D, Isanogle KA, Robinson CM, Luo X, Chen JQ, Walton A, Awasthi P, Edmondson EF, Difilippantonio S, Wei JS, Zhao K, Ferrer M, Thomas CJ, Khan J. MEK inhibition induces MYOG and remodels super-enhancers in RAS-driven rhabdomyosarcoma. Sci Transl Med 2018; 10:eaan4470. [PMID: 29973406 PMCID: PMC8054766 DOI: 10.1126/scitranslmed.aan4470] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 06/06/2018] [Indexed: 12/22/2022]
Abstract
The RAS isoforms are frequently mutated in many types of human cancers, including PAX3/PAX7 fusion-negative rhabdomyosarcoma. Pediatric RMS arises from skeletal muscle progenitor cells that have failed to differentiate normally. The role of mutant RAS in this differentiation blockade is incompletely understood. We demonstrate that oncogenic RAS, acting through the RAF-MEK [mitogen-activated protein kinase (MAPK) kinase]-ERK (extracellular signal-regulated kinase) MAPK effector pathway, inhibits myogenic differentiation in rhabdomyosarcoma by repressing the expression of the prodifferentiation myogenic transcription factor, MYOG. This repression is mediated by ERK2-dependent promoter-proximal stalling of RNA polymerase II at the MYOG locus. Small-molecule screening with a library of mechanistically defined inhibitors showed that RAS-driven RMS is vulnerable to MEK inhibition. MEK inhibition with trametinib leads to the loss of ERK2 at the MYOG promoter and releases the transcriptional stalling of MYOG expression. MYOG subsequently opens chromatin and establishes super-enhancers at genes required for late myogenic differentiation. Furthermore, trametinib, in combination with an inhibitor of IGF1R, potently decreases rhabdomyosarcoma cell viability and slows tumor growth in xenograft models. Therefore, this combination represents a potential therapeutic for RAS-mutated rhabdomyosarcoma.
Collapse
Affiliation(s)
- Marielle E Yohe
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA.
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Berkley E Gryder
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jack F Shern
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Young K Song
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Hsien-Chao Chou
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Sivasish Sindiri
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Arnulfo Mendoza
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Rajesh Patidar
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Rajarashi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Donna Butcher
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21702, USA
| | - Kristine A Isanogle
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Christina M Robinson
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Xiaoling Luo
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jin-Qiu Chen
- Collaborative Protein Technology Resource, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Ashley Walton
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Parirokh Awasthi
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Elijah F Edmondson
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21702, USA
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701, USA
| | - Jun S Wei
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Keji Zhao
- Systems Biology Center, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Bethesda, MD 20892, USA
| | - Javed Khan
- Oncogenomics Section, Genetics Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
5
|
Abstract
The developmental mechanisms that control head muscle formation are distinct from those that operate in the trunk. Head and neck muscles derive from various mesoderm populations in the embryo and are regulated by distinct transcription factors and signaling molecules. Throughout the last decade, developmental, and lineage studies in vertebrates and invertebrates have revealed the peculiar nature of the pharyngeal mesoderm that forms certain head muscles and parts of the heart. Studies in chordates, the ancestors of vertebrates, revealed an evolutionarily conserved cardiopharyngeal field that progressively facilitates the development of both heart and craniofacial structures during vertebrate evolution. This ancient regulatory circuitry preceded and facilitated the emergence of myogenic cell types and hierarchies that exist in vertebrates. This chapter summarizes studies related to the origins, signaling circuits, genetics, and evolution of the head musculature, highlighting its heterogeneous characteristics in all these aspects, with a special focus on the FGF-ERK pathway. Additionally, we address the processes of head muscle regeneration, and the development of stem cell-based therapies for treatment of muscle disorders.
Collapse
Affiliation(s)
- Inbal Michailovici
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Eigler
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Tzahor
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
6
|
Ibrahim DM, Hansen P, Rödelsperger C, Stiege AC, Doelken SC, Horn D, Jäger M, Janetzki C, Krawitz P, Leschik G, Wagner F, Scheuer T, Schmidt-von Kegler M, Seemann P, Timmermann B, Robinson PN, Mundlos S, Hecht J. Distinct global shifts in genomic binding profiles of limb malformation-associated HOXD13 mutations. Genome Res 2013; 23:2091-102. [PMID: 23995701 PMCID: PMC3847778 DOI: 10.1101/gr.157610.113] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Gene regulation by transcription factors (TFs) determines developmental programs and cell identity. Consequently, mutations in TFs can lead to dramatic phenotypes in humans by disrupting gene regulation. To date, the molecular mechanisms that actually cause these phenotypes have been difficult to address experimentally. ChIP-seq, which couples chromatin immunoprecipitation with high-throughput sequencing, allows TF function to be investigated on a genome-wide scale, enabling new approaches for the investigation of gene regulation. Here, we present the application of ChIP-seq to explore the effect of missense mutations in TFs on their genome-wide binding profile. Using a retroviral expression system in chicken mesenchymal stem cells, we elucidated the mechanism underlying a novel missense mutation in HOXD13 (Q317K) associated with a complex hand and foot malformation phenotype. The mutated glutamine (Q) is conserved in most homeodomains, a notable exception being bicoid-type homeodomains that have lysine (K) at this position. Our results show that the mutation results in a shift in the binding profile of the mutant toward a bicoid/PITX1 motif. Gene expression analysis and functional assays using in vivo overexpression studies confirm that the mutation results in a partial conversion of HOXD13 into a TF with bicoid/PITX1 properties. A similar shift was not observed with another mutation, Q317R, which is associated with brachysyndactyly, suggesting that the bicoid/PITX1-shift observed for Q317K might be related to the severe clinical phenotype. The methodology described can be used to investigate a wide spectrum of TFs and mutations that have not previously been amenable to ChIP-seq experiments.
Collapse
Affiliation(s)
- Daniel M Ibrahim
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, 13353 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Abstract
The objective of the present study was to investigate the optimal dietary n-6:n-3 PUFA ratios that regulate lipid metabolism and inflammation in pigs. A total of ninety-six cross-bred (Large White × Landrace) growing-finishing pigs (73·8 (SEM 1·6) kg) were chosen and fed one of the four isoenergetic diets with n-6:n-3 PUFA ratios of 1:1, 2·5:1, 5:1 and 10:1. The growth performance of pigs fed the diet with an n-6:n-3 PUFA ratio of 5:1 was the best, but the group fed the diet with an n-6:n-3 PUFA ratio of 1:1 had the highest muscle mass and the lowest adipose tissue mass (P< 0·05). The concentrations of IL-6 and IL-1β of pigs fed the diet with an n-6:n-3 PUFA ratio of 1:1 were decreased compared with those of the other groups (P< 0·05). The concentration of adiponectin of pigs fed the diet with an n-6:n-3 PUFA ratio of 1:1 was also markedly decreased, but the concentration of leptin was increased compared with that of the groups fed the diets with n-6:n-3 PUFA ratios of 5:1 and 10:1 (P< 0·05). Additionally, the optimal dietary ratios of n-6:n-3 PUFA of 1:1 and 5:1 markedly suppressed the expression levels of lipid metabolism-related genes and proteins such as phosphoinositide-3-kinase-α, fatty acid transport protein-1 and PPARγ. They also significantly suppressed the expression levels of the inflammatory cytokines IL-1β, TNF-α and IL-6. The results indicated that the optimal n-6:n-3 PUFA ratios of 1:1 and 5:1 exerted beneficial effects on lipid metabolism and inflammatory system, leading to the availability of more energy and nutrients for high performance and homeostatic pathways.
Collapse
|
8
|
Leroy MC, Perroud J, Darbellay B, Bernheim L, Konig S. Epidermal growth factor receptor down-regulation triggers human myoblast differentiation. PLoS One 2013; 8:e71770. [PMID: 23967242 PMCID: PMC3744467 DOI: 10.1371/journal.pone.0071770] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 07/09/2013] [Indexed: 12/02/2022] Open
Abstract
Initiation of human myoblast differentiation requires a negative shift (hyperpolarization) of the resting potential of myoblasts that depends on the activation of Kir2.1 potassium channels. These channels are regulated by a tyrosine phosphorylation. Using human primary myoblast culture, we investigated a possible role of various receptor tyrosine kinases in the induction of the differentiation process. We found that Epidermal Growth Factor Receptor (EGFR) is a key regulator of myoblast differentiation. EGFR activity is down-regulated during early human myoblast differentiation, and this event is required for normal differentiation to take place. Furthermore, EGFR silencing in proliferation conditions was able to trigger the differentiation program. This occurs through an increase of Kir2.1 channel activity that, via a rise of store-operated Ca2+ entry, leads to the expression of myogenic transcription factors and muscle specific proteins (Myogenin, Myocyte Enhancer Factor 2 (MEF2), Myosin Heavy Chain (MyHC)). Finally, blocking myoblast cell cycle in proliferation conditions using a cdk4 inhibitor greatly decreased myoblast proliferation but was not able, on its own, to promote myoblast differentiation. Taken together, these results show that EGFR down-regulation is an early event that is required for the induction of myoblast differentiation.
Collapse
Affiliation(s)
- Marina C. Leroy
- Department of Basic Neurosciences, University Medical Center, Geneva, Switzerland
| | - Julie Perroud
- Department of Basic Neurosciences, University Medical Center, Geneva, Switzerland
| | - Basile Darbellay
- Department of Clinical Neurosciences, University Hospital, Geneva, Switzerland
| | - Laurent Bernheim
- Department of Basic Neurosciences, University Medical Center, Geneva, Switzerland
| | - Stephane Konig
- Department of Basic Neurosciences, University Medical Center, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
9
|
Myostatin regulates preadipocyte differentiation and lipid metabolism of adipocyte via ERK1/2. Cell Biol Int 2012; 35:1141-6. [PMID: 21510842 DOI: 10.1042/cbi20110112] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Myostatin is known as an inhibitor of muscle development, but its role in adipogenesis and lipid metabolism is still unclear, especially the underlying mechanisms. Here, we demonstrated that myostatin inhibited 3T3-L1 preadipocyte differentiation into adipocyte by suppressing C/EBPα (CCAAT/enhancer-binding protein α) and PPARγ (peroxisome-proliferator-activated receptor γ), also activated ERK1/2 (extracellular-signal-regulated kinase 1/2). Furthermore, myostatin enhanced the phosphorylation of HSL (hormone-sensitive lipase) and ACC (acetyl-CoA carboxylase) in fully differentiated adipocytes, as well as ERK1/2. Besides, we noted that myostatin markedly raised the levels of leptin and adiponectin release and mRNA expression during preadipocyte differentiation, but the levels were inhibited by myostatin treatments in fully differentiated adipocytes. These results suggested that myostatin suppressed 3T3-L1 preadipocyte differentiation and regulated lipid metabolism of mature adipocyte, in part, via activation of ERK1/2 signalling pathway.
Collapse
|
10
|
Murray J, Huss JM. Estrogen-related receptor α regulates skeletal myocyte differentiation via modulation of the ERK MAP kinase pathway. Am J Physiol Cell Physiol 2011; 301:C630-45. [PMID: 21562305 PMCID: PMC3174569 DOI: 10.1152/ajpcell.00033.2011] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 05/06/2011] [Indexed: 02/06/2023]
Abstract
Myocyte differentiation involves complex interactions between signal transduction pathways and transcription factors. The estrogen-related receptors (ERRs) regulate energy substrate uptake, mitochondrial respiration, and biogenesis and may target structural gene programs in striated muscle. However, ERRα's role in regulating myocyte differentiation is not known. ERRα and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) are coordinately upregulated with metabolic and skeletal muscle-specific genes early in myogenesis. We analyzed effects of ERRα overexpression and loss of function in myogenic models. In C2C12 myocytes ERRα overexpression accelerated differentiation, whereas XCT790 treatment delayed myogenesis and resulted in myotubes with fewer mitochondria and disorganized sarcomeres. ERRα-/- primary myocytes showed delayed myogenesis, resulting in structurally immature myotubes with reduced sarcomeric assembly and mitochondrial function. However, sarcomeric and metabolic gene expression was unaffected or upregulated in ERRα-/- cells. Instead, ERRα-/- myocytes exhibited aberrant ERK activation early in myogenesis, consistent with delayed myotube formation. XCT790 treatment also increased ERK phosphorylation in C2C12, whereas ERRα overexpression decreased early ERK activation, consistent with the opposing effects of these treatments on differentiation. The transient induction of MAP kinase phosphatase-1 (MKP-1), which mediates ERK dephosphorylation at the onset of myogenesis, was lost in ERRα-/- myocytes and in XCT790-treated C2C12. The ERRα-PGC-1α complex activates the Dusp1 gene, which encodes MKP-1, and ERRα occupies the proximal 5' regulatory region during early differentiation in C2C12 myocytes. Finally, treatment of ERRα-/- myocytes with MEK inhibitors rescued normal ERK signaling and myogenesis. Collectively, these data demonstrate that ERRα is required for normal skeletal myocyte differentiation via modulation of MAP kinase signaling.
Collapse
MESH Headings
- Animals
- Butadienes/pharmacology
- Cell Differentiation/drug effects
- Cell Differentiation/physiology
- Creatine Kinase, Mitochondrial Form/genetics
- Dual Specificity Phosphatase 1/genetics
- Dual Specificity Phosphatase 1/metabolism
- Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Flavonoids/pharmacology
- Gene Expression/drug effects
- Gene Expression/genetics
- Kinetics
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria/metabolism
- Mitochondria/pathology
- Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Muscle Development/drug effects
- Muscle Development/physiology
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/enzymology
- Muscle Fibers, Skeletal/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Myoblasts, Skeletal/cytology
- Myoblasts, Skeletal/drug effects
- Myoblasts, Skeletal/enzymology
- Myoblasts, Skeletal/metabolism
- Myogenin/genetics
- Myosin Heavy Chains/genetics
- Nitriles/pharmacology
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
- Phosphorylation/drug effects
- Protein Binding/genetics
- Protein Serine-Threonine Kinases/genetics
- Pyruvate Dehydrogenase Acetyl-Transferring Kinase
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/deficiency
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Sarcomeres/pathology
- Thiazoles/pharmacology
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors
- Transduction, Genetic
- Troponin I/genetics
- Troponin I/metabolism
- Up-Regulation/drug effects
- Up-Regulation/genetics
- p38 Mitogen-Activated Protein Kinases/metabolism
- ERRalpha Estrogen-Related Receptor
Collapse
Affiliation(s)
- Jennifer Murray
- Division of Gene Regulation and Drug Discovery, Department of Diabetes and Metabolic Diseases Research, Beckman Research Institute, City of Hope, Duarte, California, USA
| | | |
Collapse
|
11
|
Tidyman WE, Lee HS, Rauen KA. Skeletal muscle pathology in Costello and cardio-facio-cutaneous syndromes: developmental consequences of germline Ras/MAPK activation on myogenesis. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2011; 157C:104-14. [PMID: 21495178 DOI: 10.1002/ajmg.c.30298] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cardio-facio-cutaneous syndrome (CFC) and Costello syndrome (CS) are two of the more rare RASopathies caused by altered signal transduction of the Ras/mitogen-activated protein kinase (MAPK) pathway. All of the RASopathies exhibit some degree of hypotonia, but CS and CFC are more severe. To determine if individuals with CS and CFC have an underlying skeletal myopathy, we systematically evaluated skeletal muscle pathology in both conditions. We reviewed pathology reports from six individuals who had undergone a skeletal muscle biopsy, and we reviewed histology slides on two cases with CS and one case with CFC. All patients in the cohort had histopathologic findings, and two consistent abnormalities were identified. The first was the presence of abnormal muscle fiber size and variability, and the second was the presence of type 2 fiber predominance. Given the degree of hypotonia typically present in these patients, the overall architecture of the muscle was relatively normal, without showing indications of severe structural histopathology or metabolic abnormalities. Because the Ras/MAPK pathway is vital for skeletal myogenesis, we evaluated the effects of CS and CFC mutations on myogenesis using C2C12 myoblasts. All CS/CFC mutations inhibited myoblast differentiation as indicated by fewer myosin heavy chain expressing cells and a decrease in the number of myotubes as compared to controls. These findings indicate that CS and CFC may have a true myopathy related to an inherent dysregulation of skeletal myogenesis, which further expands our understanding of the consequences of germline Ras/MAPK mutations.
Collapse
Affiliation(s)
- William E Tidyman
- Department of Orofacial Science at the University of California at San Francisco, USA
| | | | | |
Collapse
|
12
|
Jo C, Cho SJ, Jo SA. Mitogen-activated protein kinase kinase 1 (MEK1) stabilizes MyoD through direct phosphorylation at tyrosine 156 during myogenic differentiation. J Biol Chem 2011; 286:18903-13. [PMID: 21454680 DOI: 10.1074/jbc.m111.225128] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previously, we reported that mitogen-activated protein kinase kinase 1 (MEK1) activated in the mid-stage of skeletal muscle differentiation promotes myogenic differentiation. To elucidate the molecular mechanism, we investigated an activity of MEK1 for MyoD. Activated MEK1 associates with MyoD in the nucleus of differentiating myoblasts. In vitro kinase assay using active MEK1, a (32)P-labeled protein band corresponding to GST-MyoD was observed but not to mutant GST-MyoD-Y156F. Tyrosine phosphorylation of endogenous MyoD was detected with a specific anti-pMyoD-Y156 antibody; however, this response was blocked by PD184352, a MEK-specific inhibitor. These results indicate that activated MEK1 phosphorylates the MyoD-Y156 residue directly. Interestingly, the protein level of mutant MyoD-Y156F decreased compared with that of wild type but was recovered in the presence of lactacystin, a proteasome inhibitor. The protein level of MyoD-Y156E, which mimics phosphorylation at Tyr-156, was above that of wild type, indicating that the phosphorylation protects MyoD from the ubiquitin proteasome-mediated degradation. In addition, the low protein level of MyoD-Y156F was recovered over that of wild type by an additional mutation at Leu-164, a critical binding residue of MAFbx/AT-1, a Skp, Cullin, F-box (SCF) E3-ubiquitin ligase. The amount of MyoD co-precipitated with MAFbx/AT-1 also was reduced in the presence of active MEK1. Thus, these results suggested that the phosphorylation probably interrupts the binding of MAFbx/AT-1 to MyoD and thereby increases its stability. Collectively, our results suggest that MEK1 activated in differentiating myoblasts stimulates muscle differentiation by phosphorylating MyoD-Y156, which results in MyoD stabilization.
Collapse
Affiliation(s)
- Chulman Jo
- Division of Brain Disease, Center for Biomedical Science, National Institutes of Health, Korea Center for Disease Control and Prevention, 187 Osongsaengmyeong2-ro, Gangoe-myeon, Cheongwon-gun, Chungcheongbuk-do 363-951, South Korea
| | | | | |
Collapse
|
13
|
Beffy P, Del Carratore R, Masini M, Furling D, Puymirat J, Masiello P, Simili M. Altered signal transduction pathways and induction of autophagy in human myotonic dystrophy type 1 myoblasts. Int J Biochem Cell Biol 2010; 42:1973-83. [PMID: 20797447 DOI: 10.1016/j.biocel.2010.08.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 07/28/2010] [Accepted: 08/12/2010] [Indexed: 11/18/2022]
Abstract
Congenital myotonic dystrophy type 1 (CDM1) affects patients from birth and is associated with mental retardation and impaired muscle development. CDM1 patients carry 1000-3000 CTG repeats in the DMPK gene and display defective skeletal muscles differentiation, resulting in reduced size of myotubes and decreased number of satellite cells. In this study, human myoblasts in culture deriving from control and DM1 embryos (3200 CTG repeats) were analyzed using both a biochemical and electron microscopic approach, in order to provide new insights into the molecular mechanisms underlying such alteration. Interestingly, electron microscopy analysis showed not only ultrastructural features of abnormal differentiation but also revealed the presence of autophagic vacuoles in DM1 myoblasts not undergoing differentiation. In accordance with the electron microscopic findings, the autophagic markers LC3 and ATG5, but not apoptotic markers, were significantly up regulated in DM1 myoblasts after differentiating medium addition. The induction of autophagic processes in DM1 myoblasts was concomitant to p53 over-expression and inhibition of the mTOR-S6K1 pathway, causatively involved in autophagy. Moreover biochemical alterations of the two main signal transduction pathways involved in differentiation were observed in DM1 myoblasts, in particular decreased activation of p38MAPK and persistent activation of the MEK-ERK pathway. This work, while demonstrating that major signaling pathways regulating myoblasts differentiation are profoundly deranged in DM1 myoblasts, for the first time provides evidence of autophagy induction, possibly mediated by p53 activation in response to metabolic stress which might contribute to the dystrophic alterations observed in the muscles of congenital DM1 patients.
Collapse
|
14
|
MEK1 plays contrary stage-specific roles in skeletal myogenic differentiation. Cell Signal 2009; 21:1910-7. [DOI: 10.1016/j.cellsig.2009.08.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Revised: 08/14/2009] [Accepted: 08/21/2009] [Indexed: 12/11/2022]
|
15
|
Modulation of proliferation and differentiation of C2C12 skeletal muscle cells by fatty acids. Life Sci 2009; 84:415-20. [DOI: 10.1016/j.lfs.2009.01.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 01/05/2009] [Accepted: 01/12/2009] [Indexed: 11/23/2022]
|
16
|
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
|
17
|
Lim MJ, Seo YH, Choi KJ, Cho CH, Kim BS, Kim YH, Lee J, Lee H, Jung CY, Ha J, Kang I, Kim SS. Suppression of c-Src activity stimulates muscle differentiation via p38 MAPK activation. Arch Biochem Biophys 2007; 465:197-208. [PMID: 17612500 DOI: 10.1016/j.abb.2007.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2007] [Revised: 05/25/2007] [Accepted: 06/03/2007] [Indexed: 11/28/2022]
Abstract
Role of c-Src in muscle differentiation has been controversial. Here, we investigated if c-Src positively or negatively regulates muscle differentiation, using H9c2 and C2C12 cell lines. Inhibition of c-Src by treatment with PP1 and SU6656, pharmacologic inhibitors of Src family kinases, or by expression of a dominant negative c-Src, all induced muscle differentiation in proliferation medium (PM). In differentiating cells in differentiation medium (DM), c-Src activity gradually decreased and reached basal level 3 days after induction of differentiation. Inhibition of c-Src suppressed Raf/MEK/ERK pathway but activated p38 MAPK. Inhibition of p38 MAPK did not affect c-Src activity in PM. However, it reactivated Raf/MEK/ERK pathway in c-Src-inhibited cells regardless of PM or DM. Concomitant inhibition of c-Src and p38 MAPK activities blocked muscle differentiation in both media. In conclusion, suppression of c-Src activity stimulates muscle differentiation by activating p38 MAPK uni-directionally.
Collapse
Affiliation(s)
- Min Jin Lim
- Department of Biochemistry and Molecular Biology (BK21 project), Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Puente LG, Voisin S, Lee REC, Megeney LA. Reconstructing the regulatory kinase pathways of myogenesis from phosphopeptide data. Mol Cell Proteomics 2006; 5:2244-51. [PMID: 16971385 DOI: 10.1074/mcp.m600134-mcp200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Multiple kinase activities are required for skeletal muscle differentiation. However, the mechanisms by which these kinase pathways converge to coordinate the myogenic process are unknown. Using multiple phosphoprotein and phosphopeptide enrichment techniques we obtained phosphopeptides from growing and differentiating C2C12 muscle cells and determined specific peptide sequences using LC-MS/MS. To place these phosphopeptides into a rational context, a bioinformatics approach was used. Phosphorylation sites were matched to known site-specific and to site non-specific kinase-substrate interactions, and then other substrates and upstream regulators of the implicated kinases were incorporated into a model network of protein-protein interactions. The model network implicated several kinases of known relevance to myogenesis including AKT, GSK3, CDK5, p38, DYRK, and MAPKAPK2 kinases. This combination of proteomics and bioinformatics technologies should offer great utility as the volume of protein-protein and kinase-substrate information continues to increase.
Collapse
Affiliation(s)
- Lawrence G Puente
- Ottawa Health Research Institute, Molecular Medicine Program, Ottawa Hospital, and Department of Cellular and Molecular Medicine, Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ontario K1H 8L6, Canada
| | | | | | | |
Collapse
|
19
|
Li J, Johnson SE. ERK2 is required for efficient terminal differentiation of skeletal myoblasts. Biochem Biophys Res Commun 2006; 345:1425-33. [PMID: 16729973 DOI: 10.1016/j.bbrc.2006.05.051] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 05/07/2006] [Indexed: 11/24/2022]
Abstract
Terminal differentiation of skeletal myoblasts involves alignment of the mononucleated cells, fusion into multinucleated syncitia, and transcription of muscle-specific genes. Myogenesis in vivo is regulated partially by IGF-I initiated signaling that results in activation of an intracellular phosphatidylinositol 3 kinase (PI3K) signaling cascade. Downstream signaling through the Raf/MEK/ERK axis, a pathway initiated by IGF-I, also is implicated in the regulation of muscle formation. The involvement of ERK1 and ERK2 during myogenesis was examined in C2C12 myoblasts. C2C12 myoblasts stably expressing a small interfering RNA (siRNA) directed against ERK1 or ERK2 were created. Both of the kinases were reduced to trace levels as measured by Western for total ERK and retained the capacity to become phosphorylated. C2C12siERK2 knockdown myoblasts failed to fuse into multinucleated myofibers. By contrast, cells expressing a scrambled siRNA or ERK1 siRNA fused into large multinucleated structures. The block to muscle formation did not involve continued cell cycle progression or apoptosis. C2C12siERK1 myoblasts expressed an increased amount of ERK2 protein and formed larger myofibers in response to IGF-I treatment. Interestingly, IGF-I treatment of C2C12 ERK2 knockdown myoblasts did not reinstate the myogenic program arguing that ERK2 is required for differentiation. These results provide evidence for ERK2 as a positive regulator of myogenesis and suggest that ERK1 is dispensable for myoblast proliferation and differentiation.
Collapse
Affiliation(s)
- Ju Li
- Department of Animal Sciences, University of Florida, Gainesville, 32611, USA
| | | |
Collapse
|
20
|
Park SH, Lee H, Park KK, Kim HW, Park T. Taurine-Responsive Genes Related to Signal Transduction as Identified by cDNA Microarray Analyses of HepG2 Cells. J Med Food 2006; 9:33-41. [PMID: 16579726 DOI: 10.1089/jmf.2006.9.33] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Taurine-induced changes in the expression profiles of HepG2 cells were assessed using a cDNA microarray technology, and confirmed by real-time reverse transcription-polymerase chain reaction (RT-PCR) analyses. Of 8,298 human genes on the microarray, 128 genes (87 known genes) were up-regulated, and 349 (206 known genes) were down-regulated more than 2.0-fold by taurine. Among the 293 known genes regulated by taurine, a total of 44 genes were involved in signal transduction; 16 genes were up-regulated greater than 2.0-fold, and 28 genes were down-regulated more than 2.0-fold by taurine. The results of RT-PCR analyses for the five genes selected were consistent with our microarray data, although the fold changes in the expression level differed somewhat between the two analytical methods. Among signal transduction-related genes affected by taurine, four genes--mitogen-activated protein kinase (MAPK) kinase kinase 7, p21-activated kinase 4, sprouty homolog 2, and MAPK kinase 1--are implicated in the MAPK signaling pathway. Taurine also regulated the expression of signal transducer and activator of transcription (STAT) 3 gene involved in the Janus kinase-STAT pathway, and diacylglycerol kinase, zeta 104 kDa, the downstream mediator of the protein kinase C transmembrane signaling pathway. In conclusion, gene expression profiling of HepG2 cells treated with taurine provided us with new insights into the novel aspects of taurine as a possible regulator of MAPK signaling cascades and protein kinase C signaling pathways involved in cellular processes such as cell growth, differentiation, and apoptosis.
Collapse
Affiliation(s)
- Sung-Hee Park
- Department of Food and Nutrition, Yonsei University, Korea
| | | | | | | | | |
Collapse
|
21
|
Johnson SE, Winner DG, Wang X. Ran binding protein 9 interacts with Raf kinase but does not contribute to downstream ERK1/2 activation in skeletal myoblasts. Biochem Biophys Res Commun 2006; 340:409-16. [PMID: 16364241 DOI: 10.1016/j.bbrc.2005.12.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Accepted: 12/06/2005] [Indexed: 10/25/2022]
Abstract
Raf kinase is the upstream activator of MEK1/2 leading to phosphorylation and activation of ERK1/2. Sustained activation of Raf represses skeletal muscle-specific reporter gene transcription and formation of multinucleated myofibers. Inhibition of myogenesis by activated Raf involves downstream ERK1/2 as well as undefined mediators. To identify Raf-interacting proteins that may influence repression of muscle formation, a yeast two-hybrid screen was performed using a MEK1-binding defective Raf (RafBXB-T481A) as bait. Twenty cDNAs coding for Raf-interacting proteins were identified including Ran binding protein 9 (RanBP9), a protein previously reported to interact with receptor tyrosine kinases. Forced expression of RanBP9 in myogenic cells did not alter myogenesis. Co-expression of RanBP9 with constitutively active RafBXB, but not RafBXB-T481A, synergistically inhibited MyoD-directed muscle reporter gene transcription. Knockdown of RanBP9 expression did not restore the differentiation program to Raf-expressing myoblasts. Thus, RanBP9 physically associates with Raf but does not substantially contribute to the inhibitory actions of the kinase.
Collapse
|
22
|
Champigny MJ, Perry R, Rudnicki M, Igdoura SA. Overexpression of MyoD-inducible lysosomal sialidase (neu1) inhibits myogenesis in C2C12 cells. Exp Cell Res 2005; 311:157-66. [PMID: 16216242 DOI: 10.1016/j.yexcr.2005.08.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2004] [Revised: 08/16/2005] [Accepted: 08/30/2005] [Indexed: 11/24/2022]
Abstract
Lysosomal sialidase, encoded by neu1, is required for the removal of terminal sialic acid residues from a variety of sialoglycoconjugates. In humans, deficiency of this enzyme results in the inborn error of metabolism sialidosis, characterized by the accumulation of sialoglycoconjugates within the nervous system and in peripheral organs. A subset of sialidosis patients present with symptoms of profound muscle dysfunction, including progressive muscular atrophy. We have previously shown that the 5' regulatory region of murine neu1 is typical of skeletal muscle-specific genes due to the presence of several E-boxes and its responsiveness to stimulation by muscle regulatory factors (MRFs) such as MyoD. Here, we report that sialidase activity is increased 6-fold during the first 24 h of differentiation of C2C12 myoblasts followed by an attenuation to pre-differentiation levels by 48 h. We demonstrate that the lysosomal sialidase promoter is highly upregulated by MyoD through a mechanism that is dependent on the MyoD chromatin remodeling domain. We also show that the sialidase promoter is repressed by activated MEK. Inappropriate overexpression of sialidase 48 h after the onset of differentiation results in downregulation of myogenin as well as myosin heavy chain expression and in a halt of the differentiation cascade. This study indicates that lysosomal sialidase is a potent regulator of the early stages of myogenesis.
Collapse
Affiliation(s)
- Marc J Champigny
- Department of Biology, McMaster University, 1280 Main St. W. LSB 335, Hamilton, Ontario, Canada
| | | | | | | |
Collapse
|
23
|
Jo C, Kim H, Jo I, Choi I, Jung SC, Kim J, Kim SS, Jo SA. Leukemia inhibitory factor blocks early differentiation of skeletal muscle cells by activating ERK. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1743:187-97. [PMID: 15843032 DOI: 10.1016/j.bbamcr.2004.11.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2003] [Revised: 10/13/2004] [Accepted: 11/05/2004] [Indexed: 01/18/2023]
Abstract
Leukemia inhibitory factor (LIF) is a multifunctional cytokine belonging to the interleukin-6 family and has been shown to stimulate regeneration of injured skeletal muscle. Although LIF has been shown to stimulate muscle cell proliferation, its precise role in differentiation is unclear. Thus, we examined the effect of LIF on the differentiation of cultured C2C12 myoblast cells. In this study, we used both non-glycosylated LIF expressed in bacteria and glycosylated LIF secreted from NIH3T3 cells infected with Ad-LIF. Both non-glycosylated and glycosylated LIF blocked differentiation of myoblasts as measured by expression of myosin heavy chain and myotube formation. Treatment of myoblasts with LIF induced phosphorylation of ERK, and the LIF-induced inhibitory effect on myogenesis was blocked by pretreatment with U0126, a specific MEK inhibitor, and transient transfection with dominant negative (DN)-MEK1. In contrast, although LIF activated STAT3, the LIF-induced repression of the MCK transcriptional activity was not reversed by pretreatment with AG490, a specific Jak kinase inhibitor or transient transfection with DN-STAT3. Additionally, LIF exhibited its inhibitory effect on myogenesis only when cells were treated at earlier than 12 h after inducing differentiation. Taken together, these results suggest that LIF strongly inhibited early myogenic differentiation though activation of the ERK signaling pathway and its effect is irrespective of glycosylation.
Collapse
Affiliation(s)
- Chulman Jo
- Department of Biomedical Sciences, National Institute of Health, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Goeman F, Thormeyer D, Abad M, Serrano M, Schmidt O, Palmero I, Baniahmad A. Growth inhibition by the tumor suppressor p33ING1 in immortalized and primary cells: involvement of two silencing domains and effect of Ras. Mol Cell Biol 2005; 25:422-31. [PMID: 15601862 PMCID: PMC538761 DOI: 10.1128/mcb.25.1.422-431.2005] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
ING1 was identified as an inhibitor of growth and has been described as a tumor suppressor. Furthermore, the expression of ING1 is induced in senescent cells and antisense ING1 extends the proliferative life span of primary human fibroblasts. Cooperation of p33ING1 with p53 has been suggested to be an important function of ING1 in cell cycle control. Intriguingly, it has been shown that p33ING1 is associated with histone acetylation as well as with histone deacetylation function. Here we show that p33ING1 is a potent transcriptional silencer in various cell types. However, the silencing function is independent of the presence of p53. By use of deletion mutants two potent autonomous and transferable silencing domains were identified, but no evidence of an activation domain was found. The amino (N)-terminal silencing domain is sensitive to the histone deacetylase inhibitor trichostatin A (TSA) whereas the carboxy-terminal silencing function is resistant to TSA, suggesting that p33ING1 confers gene silencing through both HDAC-dependent and -independent mechanisms. Interestingly, the presence of oncogenic Ras, which is able to induce premature senescence, increases the p33ING1-mediated silencing function. Moreover, ING1-mediated silencing was reduced by coexpressing dominant-negative Ras or by treatment with the mitogen-activated protein kinase inhibitor PD98059 but not by treatment with SB203580, an inhibitor of the p38 pathway. In addition, we show that both silencing domains of ING1 are involved in cell cycle control, as measured by inhibition of colony formation of immortalized cells and by thymidine incorporation of primary human diploid fibroblasts (HDF). Interestingly, p33ING1 expression induces features of cellular senescence in HDFs.
Collapse
Affiliation(s)
- Frauke Goeman
- Genetic Institute, Justus-Liebig-University, Heinrich-Buff-Ring 58-62, 35392 Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Tiffin N, Adi S, Stokoe D, Wu NY, Rosenthal SM. Akt phosphorylation is not sufficient for insulin-like growth factor-stimulated myogenin expression but must be accompanied by down-regulation of mitogen-activated protein kinase/extracellular signal-regulated kinase phosphorylation. Endocrinology 2004; 145:4991-6. [PMID: 15489316 DOI: 10.1210/en.2004-0101] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IGF-I has a unique biphasic effect on skeletal muscle differentiation. Initially, IGF-I inhibits expression of myogenin, a skeletal muscle-specific regulatory factor essential for myogenesis. Subsequently, IGF-I switches to stimulating expression of myogenin. The mechanisms that mediate this switch in IGF action are incompletely understood. Several laboratories have demonstrated that the phosphatidylinositol-3-kinase/Akt signaling pathway is essential for myogenic differentiation and have suggested that this pathway mediates IGF-I stimulation of myogenin mRNA expression, an early critical step in the differentiation process. These studies, however, did not address concurrent Akt and MAPK/ERK1/2 phosphorylation, the latter of which is also known to regulate myogenic differentiation. In the present study in rat L6E9 muscle cells, we have manipulated ERK1/2 phosphorylation with either an upstream inhibitor or activator and examined concurrent levels of Akt and ERK1/2 phosphorylation and of myogenin mRNA expression in response to treatment with IGF-I. We find that even in the presence of phosphorylated Akt, it is only when ERK1/2 phosphorylation is inhibited that IGF-I can stimulate myogenin mRNA expression. Thus, although Akt phosphorylation may be necessary, it is not sufficient for induction of myogenic differentiation by IGF-I and must be accompanied by a decrease in ERK1/2 phosphorylation.
Collapse
Affiliation(s)
- Nicki Tiffin
- Department of Pediatrics, Box 0434, University of California San Francisco, San Francisco, California 94143-0434, USA
| | | | | | | | | |
Collapse
|
26
|
Page JL, Wang X, Sordillo LM, Johnson SE. MEKK1 signaling through p38 leads to transcriptional inactivation of E47 and repression of skeletal myogenesis. J Biol Chem 2004; 279:30966-72. [PMID: 15159407 DOI: 10.1074/jbc.m402224200] [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: 12/20/2022] Open
Abstract
Activation of the Raf kinase signal transduction pathway in skeletal myoblasts causes a complete cessation of myofiber formation and muscle gene expression. The negative impacts of the signaling pathway are realized through downstream activation of mitogen and extracellular kinase (MEK) phosphorylation-dependent events and MEK-independent signal transmission. MEKK1, a kinase that can physically associate with Raf, may contribute to the MEK-independent signaling in response to elevated Raf activity. Myogenic cells overexpressing activated Raf and kinase-defective MEKK1 remain differentiation-defective, suggesting that MEKK1 does not contribute to the inhibitory actions of Raf. However, constitutive activation of MEKK1 dramatically inhibits biochemical and morphological measures of muscle formation. MEKK1 inhibits MyoD-directed transcriptional activity without altering the ability of the protein to form heterodimers with E2A proteins or bind DNA. By contrast, the transcriptional activity of E47, the preferred dimer partner of the myogenic regulatory factors, is severely compromised by MEKK1-initiated signaling. Inhibition of MEK1/2 and JNK1/2 function did not reinstate E47-directed transcription, indicating that these two downstream kinases likely are not involved in the MEKK1-controlled transcriptional block. Inhibition of p38 signaling overcame the negative effects exerted by MEKK1 on the amino terminus of E47. Closer examination indicates that E47 is phosphorylated in vitro by p38, and deletion analysis predicts that the critical amino acid(s) phosphorylated by p38 lie outside of the minimal transcriptional activation domains. Thus, modification of E47 by p38 likely disrupts higher order protein complex formation that is necessary for muscle gene transcription.
Collapse
Affiliation(s)
- Jeanine L Page
- Department of Poultry Science, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | | | |
Collapse
|
27
|
Wang X, Thomson SR, Starkey JD, Page JL, Ealy AD, Johnson SE. Transforming growth factor beta1 is up-regulated by activated Raf in skeletal myoblasts but does not contribute to the differentiation-defective phenotype. J Biol Chem 2003; 279:2528-34. [PMID: 14594948 DOI: 10.1074/jbc.m306489200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Raf/MEK/MAPK signaling module elicits a strong negative impact on skeletal myogenesis that is reflected by a complete loss of muscle gene transcription and differentiation in multinucleated myocytes. Recent evidence indicates that Raf signaling also may contribute to myoblast cell cycle exit and cytoprotection. To further define the mechanisms by which Raf participates in cellular responses, a stable line of myoblasts expressing an estrogen receptor-Raf chimeric protein was created. The cells (23A2RafER(DD)) demonstrate a strict concentration-dependent increase in chimeric Raf protein synthesis and downstream phosphoMAPK activation. Initiation of low-level Raf activity in these cells augments contractile protein expression and myocyte fusion. By contrast, induction of high level Raf activity in 23A2RafER(DD) myoblasts inhibits the formation of myocytes and muscle reporter gene expression. Interestingly, treatment of myoblasts with conditioned medium isolated from Raf-repressive cells inhibits all of the aspects of myogenesis. Closer examination indicates that the transforming growth factor-beta(1) (TGF-beta(1)) gene is up-regulated in Raf-repressive myoblasts. The cells also direct elevated levels of Smad transcriptional activity, suggesting the existence of a TGF-beta(1) autocrine loop. However, extinguishing the biological activity of TGF-beta(1) does not restore the myogenic program. Our results provide evidence for the involvement of Raf signal transmission during myocyte formation as well as during inhibition of myogenesis.
Collapse
Affiliation(s)
- Xu Wang
- Department of Poultry Science, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | | | | | | | |
Collapse
|
28
|
Dee K, DeChant A, Weyman CM. Differential signaling through NFkappaB does not ameliorate skeletal myoblast apoptosis during differentiation. FEBS Lett 2003; 545:246-52. [PMID: 12804784 DOI: 10.1016/s0014-5793(03)00571-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
During 23A2 skeletal myoblast differentiation, roughly 30% of the population undergoes apoptosis. Further, constitutive signaling by G12V:H-Ras or Raf:CAAX abrogates this apoptosis. In this study, we demonstrate an increase in NFkappaB activity in myoblasts that have survived and are expressing muscle-specific genes. NFkappaB activity is also elevated in myoblasts expressing constitutively active G12V:H-Ras but not Raf:CAAX. Expression of a dominant negative IkappaB (IkappaB-SR) sufficient to eliminate this elevated level of NFkappaB activity, in either the 23A2 myoblasts or their G12V:H-Ras-expressing counterparts, however, does not affect survival. Furthermore, expression of a constitutively active IkappaB kinase in 23A2 myoblasts does not protect these cells from the apoptosis associated with differentiation. Since signaling by IkappaB kinase can abrogate differentiation, this result demonstrates that abrogated differentiation and abrogated apoptosis are separable phenotypes.
Collapse
Affiliation(s)
- Keith Dee
- Department of Biological, Geological, and Environmental Sciences, 2121 Euclid Avenue, Cleveland State University, Cleveland, OH 44115, USA
| | | | | |
Collapse
|
29
|
Roy S, Parinandi N, Zeigelstein R, Hu Q, Pei Y, Travers JB, Natarajan V. Hyperoxia alters phorbol ester-induced phospholipase D activation in bovine lung microvascular endothelial cells. Antioxid Redox Signal 2003; 5:217-28. [PMID: 12716481 DOI: 10.1089/152308603764816578] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We investigated the effect of hyperoxia on phospholipase D (PLD) activation in bovine lung microvascular endothelial cells (BLMVECs). Generation of intracellular reactive oxygen species in BLMVECs exposed to hyperoxia for 2 or 24 h was three-fold higher compared with normoxic cells as measured by dichlorodihydrofluorescein di(acetoxymethyl ester) fluorescence. Exposure of BLMVECs to hyperoxia for 2 or 24 h attenuated 12-O-tetradecanoylphorbol 13-acetate (TPA)-mediated PLD activation compared with normoxic cells, however, hyperoxia did not alter basal PLD activity. Antioxidants, such as propyl gallate and pyrrolidine dithiocarbamate, reversed the effect of hyperoxia on TPA-induced PLD activity. Furthermore, the TPA-induced PLD activation was inhibited not only by the protein kinase C inhibitor, Go6976, but also by the tyrosine kinase inhibitor, genistein, and by the Src kinase specific inhibitor, PP-2, suggesting the involvement of protein kinase C and also tyrosine kinases in TPA-induced PLD activation. Western blot analysis of cell lysates from the hyperoxic (2 or 24 h) BLMVECs stimulated with TPA with anti-phosphotyrosine antibody showed an attenuation in overall tyrosine phosphorylation of proteins. In conclusion, we have demonstrated that hyperoxia enhanced the generation of reactive oxygen species in lung microvascular endothelial cells and attenuated TPA-induced protein tyrosine phosphorylation and PLD activation. As protein tyrosine phosphorylation and PLD play important roles in inflammatory responses, this could provide a mechanism for the regulation of endothelial barrier function during hyperoxic lung injury.
Collapse
Affiliation(s)
- Shukla Roy
- Department of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Lee J, Hong F, Kwon S, Kim SS, Kim DO, Kang HS, Lee SJ, Ha J, Kim SS. Activation of p38 MAPK induces cell cycle arrest via inhibition of Raf/ERK pathway during muscle differentiation. Biochem Biophys Res Commun 2002; 298:765-71. [PMID: 12419320 DOI: 10.1016/s0006-291x(02)02562-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell cycle arrest is essential for initiation of muscle differentiation in myoblasts. Given the previously described essential role for p38 MAPK in myogenesis, we undertook the present study to investigate the role of p38 MAPK in the cell cycle arrest that initiates muscle differentiation. p38 MAPK activity increased during, and was required for, muscle differentiation. Inhibition of p38 MAPK stimulated Raf and ERK activities, and induced cell proliferation in differentiation medium. The concomitant inhibition of p38 MAPK and ERK, however, failed to induce differentiation or proliferation. In conclusion, inhibition of the Raf/ERK pathway and the consequent cell cycle arrest is one of the major functions of p38 MAPK during muscle differentiation.
Collapse
Affiliation(s)
- Jinhwa Lee
- Department of Biotechnology, Dongseo University, Pusan, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Johnson SE, Dorman CM, Bolanowski SA. Inhibition of myogenin expression by activated Raf is not responsible for the block to avian myogenesis. J Biol Chem 2002; 277:28742-8. [PMID: 12042315 DOI: 10.1074/jbc.m203680200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activated Raf is a potent inhibitor of skeletal muscle gene transcription and myocyte formation through stimulation of downstream MAPK. However, the molecular targets of elevated MAPK with regard to myogenic repression remain elusive. We examined the effects of activated Raf on myogenin gene expression in avian myoblasts. Overexpression of activated Raf in embryonic chick myoblasts prevented myogenin gene transcription and myocyte differentiation. Treatment with PD98059, an inhibitor of MAPK kinase (MEK), restored myogenin expression but did not reinstate the myogenic program. Using a panel of myogenin promoter deletion mutants, we were unable to identify a region within the proximal 829-bp promoter that confers responsiveness to MEK. Interestingly, our experiments identified MEF2A as a target of Raf-mediated inhibition in mouse myoblasts but not in avian myogenic cells. Embryonic myoblasts overexpressing activated Raf were unable to drive transcription from a minimal myogenin promoter reporter, containing a single E-box and MEF2 site, to levels comparable with controls. Unlike mouse myoblasts, forced expression of MEF2A did not synergistically enhance transcription from the myogenin promoter in chick myoblasts, indicating that additional molecular determinants of the block to myogenesis exist. Results of these experiments further exemplify specie differences in the mode of Raf-mediated inhibition of muscle differentiation.
Collapse
Affiliation(s)
- Sally E Johnson
- Department of Poultry Science, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | |
Collapse
|
32
|
DeChant AK, Dee K, Weyman CM. Raf-induced effects on the differentiation and apoptosis of skeletal myoblasts are determined by the level of Raf signaling: abrogation of apoptosis by Raf is downstream of caspase 3 activation. Oncogene 2002; 21:5268-79. [PMID: 12149648 DOI: 10.1038/sj.onc.1205648] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2001] [Revised: 05/03/2002] [Accepted: 05/07/2002] [Indexed: 01/22/2023]
Abstract
We examined the effect of a constitutively active Raf protein (Raf-CAAX) on the differentiation and the coincident apoptosis of skeletal myoblasts. We found that a low level of Raf signaling leads to accelerated differentiation when compared to parental myoblasts, while a higher level of Raf signaling induces a transformed morphology and abrogates both differentiation and the coincident apoptosis. Raf signaling abrogates apoptosis without blocking the activation of caspase 3 and the subsequent cleavage of caspase 3 substrates. Eliminating the signal from Raf through MEK does not restore the ability to differentiate or to undergo apoptosis in the myoblasts with a high level of Raf signal, nor does it abrogate the accelerated differentiation observed in myoblasts with lower levels of Raf signal. Constitutive signaling through MEK is required, however, to maintain a transformed morphology. These results indicate that the effect of Raf on the differentiation and apoptosis of skeletal myoblasts is dictated by the level of Raf signaling, and that Raf signaling sufficient to abrogate the apoptosis coincident with differentiation does so downstream of caspase 3 signaling.
Collapse
Affiliation(s)
- Anne K DeChant
- Department of Biological, Geological and Environmental Sciences, Cleveland State University, Cleveland, Ohio, OH 44115, USA
| | | | | |
Collapse
|
33
|
Szewczyk NJ, Peterson BK, Jacobson LA. Activation of Ras and the mitogen-activated protein kinase pathway promotes protein degradation in muscle cells of Caenorhabditis elegans. Mol Cell Biol 2002; 22:4181-8. [PMID: 12024031 PMCID: PMC133852 DOI: 10.1128/mcb.22.12.4181-4188.2002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To discover and study intracellular signals that regulate proteolysis in muscle, we have employed transgenic strains of Caenorhabditis elegans that produce a soluble LacZ reporter protein limited to body-wall and vulval muscles. This reporter protein is stable in well-fed wild-type animals, but its degradation is triggered upon a shift to 25 degrees C in a strain carrying a temperature-sensitive activating mutation in the Ras oncogene homologue let-60. These mutants are not physiologically starved, inasmuch as growth rates are normal at 25 degrees C. Ras-induced degradation is not prevented by the presence of cycloheximide added at or before the temperature shift and thus uses preexisting proteolytic systems and signaling components. Furthermore, degradation is triggered when adult animals are shifted to conditions of 25 degrees C, confirming that Ras acutely promotes protein degradation in muscles whose developmental history is normal. Reduction-of-function mutations in the downstream protein kinase Raf (lin-45), MEK (mek-2), or mitogen-activated protein kinase (MAPK) (mpk-1) prevent Ras-induced protein degradation, whereas activated MPK-1 is sufficient to trigger degradation, indicating that this kinase cascade is the principal route by which Ras signaling triggers protein degradation in muscle. This pathway is activated in hypodermal cells by the LET-23 epidermal growth factor receptor homologue, but an activating mutation in let-23 does not promote proteolysis in muscle. Starvation-induced LacZ reporter degradation is unaffected by reduction-of-function mutations in Ras, Raf, MEK, or MAPK, implying that Ras activation and starvation trigger proteolysis by mechanisms that are at least partially independent. This is the first evidence that Ras-Raf-MEK-MAPK signaling activates protein degradation in differentiated muscle.
Collapse
Affiliation(s)
- Nathaniel J Szewczyk
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, USA
| | | | | |
Collapse
|
34
|
Conejo R, de Alvaro C, Benito M, Cuadrado A, Lorenzo M. Insulin restores differentiation of Ras-transformed C2C12 myoblasts by inducing NF-kappaB through an AKT/P70S6K/p38-MAPK pathway. Oncogene 2002; 21:3739-53. [PMID: 12032842 DOI: 10.1038/sj.onc.1205469] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2001] [Revised: 03/07/2002] [Accepted: 03/11/2002] [Indexed: 11/08/2022]
Abstract
v-H-ras transformed C2C12 (C2Ras) myoblasts, overexpressing p21-Ras protein in the Ras-GTP active form, showed a differentiation-defective phenotype when cultured in low serum as compared with C2C12 myoblasts. Accordingly, the purpose of the present study was to delineate the signaling pathways that restore C2Ras myoblasts differentiation. Inhibition of p42/p44-MAPK with the chemical inhibitor PD98059, and activation of AKT/P70S6K and p38-MAPK with insulin, produced growth arrest (precluding the expression of PCNA, cyclin-D1 and retinoblastoma at the hyperphosphorylated state and inducing the expression of the cell cycle inhibitor p21(Cip)) and myogenesis (multinucleated myotubes formation and induction of creatine kinase, caveolin-3 and alpha-actin). Both events were accompanied by down-regulation of AP-1 and up-regulation of NF-kappaB transcriptional activities. Furthermore, inhibition of NF-kappaB transcriptional activity by the use of the proteasome inhibitor MG132 totally precluded differentiation by insulin+PD98059, demonstrating a direct role for NF-kappaB on C2Ras myogenesis. C2Ras myoblasts failed to restore differentiation when rapamycin or PD169316 were added in the presence of insulin+PD98059, indicating that the activation of both P70S6K and p38-MAPK was necessary to reach a fully differentiated phenotype. Finally, transient transfection of a constitutively active Myr-EGFP-AKT-HA construct (in the presence of PD98059) restored C2Ras myogenesis by its ability to activate P70S6K and p38-MAPK. A crosstalk between P70S6K and p38-MAPK was observed under rapamycin treatment in both insulin or active AKT induced myogenesis. Our results are delineating an AKT/P70S6K/p38-MAPK pathway involved in skeletal muscle differentiation.
Collapse
Affiliation(s)
- Ruben Conejo
- Departamento de Bioquimica y Biologia Molecular, Facultad de Farmacia, Universidad Complutense, 28040-Madrid, Spain
| | | | | | | | | |
Collapse
|
35
|
Thomson SR, Johnson SE. Isolation and characterization of chicken TaxREB107, a putative DNA binding protein abundantly expressed in muscle. Gene 2001; 278:81-8. [PMID: 11707324 DOI: 10.1016/s0378-1119(01)00732-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Myogenic regulatory factors (MRFs) are vital transcription factors that act at multiple points during development to establish the skeletal muscle phenotype. This class of muscle-restricted, basic helix-loop-helix (bHLH) proteins acts in concert with additional transcriptional modulators to precisely control muscle gene expression. We have isolated the chicken homologue of Tax responsive element binding protein 107 (TaxREB107). The cDNA is 83% homologous at the amino acid level to human and mouse TaxREB107 and contains a centrally located leucine zipper motif. Northern analysis demonstrated that the gene is expressed in multiple tissues including skeletal muscle. Immunofluorescent staining revealed that the cTaxREB107 protein is located in both the nuclear and cytoplasmic compartments. Distinct localization to the nucleoli supports the evidence that TaxREB107 is a ribosomal protein. Because TaxREB proteins also are implicated in transcriptional regulation, we overexpressed cTaxREB107 in embryonic myoblasts. cTaxREB107 increased troponin I reporter gene activity as well as MRF-directed transcription from a multimerized skeletal muscle E-box reporter gene (4Rtk-luc). However, cotransfection of expression plasmids coding for MyoD and cTaxREB107 did not produce an increase in 4Rtk-luc suggesting that cTaxREB107 enhances myogenic gene transcription through a means independent of a physical association with MyoD. In conclusion, our results define a role for cTaxREB107 during avian myogenesis as a positive modulator of skeletal muscle gene expression.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Blotting, Northern
- Cell Nucleus/metabolism
- Chickens/genetics
- Cytoplasm/metabolism
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- DNA, Complementary/isolation & purification
- DNA-Binding Proteins/genetics
- Gene Expression
- Gene Expression Regulation, Developmental
- Humans
- Luciferases/genetics
- Luciferases/metabolism
- Mice
- Molecular Sequence Data
- Muscles/cytology
- Muscles/embryology
- Muscles/metabolism
- MyoD Protein/genetics
- MyoD Protein/physiology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Tissue Distribution
- Transcription, Genetic
Collapse
Affiliation(s)
- S R Thomson
- Department of Poultry Science, The Pennsylvania State University, 206 Henning, University Park, PA 16802, USA
| | | |
Collapse
|
36
|
Perry RL, Parker MH, Rudnicki MA. Activated MEK1 binds the nuclear MyoD transcriptional complex to repress transactivation. Mol Cell 2001; 8:291-301. [PMID: 11545732 DOI: 10.1016/s1097-2765(01)00302-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
To elucidate the mechanism through which MAPK signaling regulates the MyoD family of transcription factors, we investigated the role of the signaling intermediate MEK1 in myogenesis. Transfection of activated MEK1 strongly repressed gene activation and myogenic conversion by the MyoD family. This repression was not mediated by direct phosphorylation of MyoD or by changes in MyoD stability or subcellular distribution. Deletion mapping revealed that MEK1-mediated repression required the MyoD amino-terminal transactivation domain. Moreover, activated MEK1 was nuclearly localized and bound a complex containing MyoD in a manner that is dependent on the presence of the MyoD amino terminus. Together, these data demonstrate that MEK1 signaling has a strong negative effect on MyoD activity via a novel mechanism involving binding of MEK1 to the nuclear MyoD transcriptional complex.
Collapse
Affiliation(s)
- R L Perry
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
37
|
Becker JR, Dorman CM, McClafferty TM, Johnson SE. Characterization of a dominant inhibitory E47 protein that suppresses C2C12 myogenesis. Exp Cell Res 2001; 267:135-43. [PMID: 11412046 DOI: 10.1006/excr.2001.5249] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Skeletal muscle formation is controlled through the coordinated actions of the muscle regulatory factors (MRFs). The activities of these basic helix-loop-helix proteins is mediated in part through heterodimer formation with a family of ubiquitous bHLH proteins, referred to as E-proteins. The primary E-protein in skeletal muscle is the E2A splice variant, E47. To further address the role of E47 during skeletal myogenesis, we created a chimeric E47 repressor protein by replacing the transcriptional activation domain with the Drosophila Engrailed transcriptional repressor domain. The dominant inhibitory E-protein (EnDeltaE47) formed homodimers capable of binding DNA and abolished E47-directed gene transcription. Stable expression of EnDeltaE47 in mouse C2C12 myoblasts effectively blocked the cells' ability to differentiate into mature myofibers. Closer examination of the molecular basis for the inhibition of myogenesis revealed that EnDeltaE47 preferentially forms heterodimers with myogenin. Interestingly, the chimeric repressor did not form DNA-binding heterodimers with MyoD in C2C12 myocytes. The failure to detect MyoD:EnDeltaE47 heterodimers in myoblasts was not due to protein conformational defects as both wild-type E47 and EnDeltaE47 readily formed DNA binding complexes with MyoD in vitro. These results indicate that E47 plays a crucial role in C2C12 myogenesis by serving as the preferred heterodimer partner of the myogenin protein.
Collapse
Affiliation(s)
- J R Becker
- Department of Poultry Science, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | | | | | | |
Collapse
|
38
|
Mitin N, Kudla AJ, Konieczny SF, Taparowsky EJ. Differential effects of Ras signaling through NFkappaB on skeletal myogenesis. Oncogene 2001; 20:1276-86. [PMID: 11313872 DOI: 10.1038/sj.onc.1204223] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2000] [Revised: 12/21/2000] [Accepted: 01/04/2001] [Indexed: 01/12/2023]
Abstract
Oncogenic Ras (H-Ras G12V) inhibits skeletal myogenesis through multiple signaling pathways. Previously, we demonstrated that the major downstream effectors of Ras (i.e., MEK/MAPK, RalGDS and Rac/Rho) play a minor, if any, role in the differentiation-defective phenotype of Ras myoblasts. Recently, NFkappaB, another Ras signaling target, has been shown to inhibit myogenesis presumably by stimulating cyclin D1 accumulation and cell cycle progression. In this study, we address the involvement of NFkappaB activation in the Ras-induced inhibition of myogenesis. Using H-Ras G12V and three G12V effector-loop variants, we detect high levels of NFkappaB transcriptional activity in C3H10T1/2-MyoD cells treated with differentiation medium. Myogenesis is blocked by all Ras proteins tested, yet only in the case of H-Ras G12V are cyclin D1 levels increased and cell cycle progression maintained. Expression of IkappaBalpha SR, an inhibitor of NFkappaB, does not reverse the differentiation-defective phenotype of Ras expressing cultures, but does induce differentiation in cultures treated with tumor necrosis factor (TNFalpha) or in cultures expressing the RelA/p65 subunit of NFkappaB. These data confirm that NFkappaB is a target of Ras and suggest that the cellular actions of NFkappaB require additional signals that are discriminated by the Ras effector-loop variants. Results with IkappaBalpha SR convincingly demonstrate that H-Ras G12V does not rely on NFkappaB activity to block myogenesis, an observation that continues to implicate another unidentified signaling pathway(s) in the inhibition of skeletal myogenesis by Ras.
Collapse
Affiliation(s)
- N Mitin
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, IN 47907-1392, USA
| | | | | | | |
Collapse
|
39
|
Pizon V, Baldacci G. Rap1A protein interferes with various MAP kinase activating pathways in skeletal myogenic cells. Oncogene 2000; 19:6074-81. [PMID: 11146560 DOI: 10.1038/sj.onc.1203984] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Constitutive expression of the activated Rap1A protein inhibits differentiation of myogenic C2 cells whereas the inactivated Rap1A protein favours cell differentiation and induces late endocytic compartments clustering. Although the role of Rap1A in MAPK activation has been analysed in various cell types, the signalling pathways activated by Rap1A have not been explored in myogenic cells. In this study, we investigated MAP kinase activity in control C2 myoblasts and in stable C2 cell lines expressing mutated Rap1A proteins. We provide evidence that Rap1A mutants promote ERK activation and that the active protein induces a more sustained activation than the inactive protein. In addition, we established that various pathways mediate transient ERK activation in control cells and in cells expressing the inactivated Rap1A protein. In these cells, ERK are activated by a Raf/MEK-dependent pathway, a P13K/Raf-independent pathway and a third undetermined pathway. In cells expressing the activated Rap1A protein, a PI3K/Raf/MEK-dependent pathway mediates transient ERK activation. However, MAPK activation appears more complex since, according to the state of the myoblasts or the duration of MAPK stimulation, we observed that Rap1A protein could interfere or not with ERK activation.
Collapse
Affiliation(s)
- V Pizon
- Institut André Lwoff, UPR 1983, Villejuif, France
| | | |
Collapse
|
40
|
Winter B, Arnold HH. Activated raf kinase inhibits muscle cell differentiation through a MEF2-dependent mechanism. J Cell Sci 2000; 113 Pt 23:4211-20. [PMID: 11069766 DOI: 10.1242/jcs.113.23.4211] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Muscle cell development is dependent on the activity of cell type-specific basic-helix-loop-helix transcription factors, MyoD, Myf-5, myogenin, and MRF4 which collaborate with myocyte enhancer factor 2 proteins to activate muscle-specific gene expression. Growth factors and activated Ras prevent differentiation of myoblasts in culture but the downstream signalling pathways are not well understood. Here, we demonstrate that active Raf kinase (Raf-BxB) completely inhibits myogenic conversion of 10T1/2 cells mediated by Myf-5 and differentiation of L6 myoblasts as indicated by the absence of myotubes, lack of myogenin expression, and markedly reduced expression of myosin heavy chain. However, activated Raf inhibits transcriptional activation by Myf-5 only partially suggesting that other potential targets of Ras/Raf signalling may be involved. Significantly, we observed that elevated Raf kinase activity in L6 muscle cells suppresses the accumulation of MEF2 protein in nuclei, while MEF2 transcription appears unaffected. Moreover, forced expression of MEF2A in 10T1/2 cells rescues MyoD dependent myogenic conversion in the presence of constitutively active Raf kinase and partially restores transactivation of a myogenin promoter-dependent reporter gene in L6 muscle cells containing activated Raf kinase. From these observations we conclude that persistent activation of Raf signalling affects nuclear MEF2 functions which may explain why myogenin expression and myoblast differentiation are inhibited.
Collapse
Affiliation(s)
- B Winter
- Department of Cell and Molecular Biology, University of Braunschweig, Spielmannstr. 7, D-38106 Braunschweig, Germany.
| | | |
Collapse
|
41
|
Dorman CM, Johnson SE. Activated Raf Inhibits Myogenesis through a Mechanism Independent of Activator Protein 1-mediated Myoblast Transformation. J Biol Chem 2000. [DOI: 10.1016/s0021-9258(19)61533-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|