1
|
Zhang X, Mille-Fragoso LS, Kaseniit KE, Lee AP, Zhang M, Call CC, Hu Y, Xie Y, Gao XJ. Post-transcriptional modular synthetic receptors. Nat Chem Biol 2025:10.1038/s41589-025-01872-w. [PMID: 40155716 DOI: 10.1038/s41589-025-01872-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/06/2025] [Indexed: 04/01/2025]
Abstract
Inspired by the power of transcriptional synthetic receptors and hoping to complement them to expand the toolbox for cell engineering, we establish LIDAR (Ligand-Induced Dimerization-Activating RNA editing), a modular post-transcriptional synthetic receptor platform that harnesses RNA editing by adenosine deaminases acting on RNA. LIDAR is compatible with various receptor architectures in different cellular contexts and enables the sensing of diverse ligands and the production of functional outputs. Furthermore, LIDAR can sense orthogonal signals in the same cell and produce synthetic spatial patterns, potentially enabling the programming of complex multicellular behaviors. Lastly, LIDAR is compatible with compact encoding and can be delivered as synthetic mRNA. Thus, LIDAR expands the family of synthetic receptors, holding the promise to empower basic research and therapeutic applications.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Luis S Mille-Fragoso
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Bio-X, Stanford University, Stanford, CA, USA
| | - K Eerik Kaseniit
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Stanford Bio-X, Stanford University, Stanford, CA, USA
| | - Arden P Lee
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Meng Zhang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Connor C Call
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Yixin Hu
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Yunxin Xie
- The Chinese Undergraduate Visiting Research (UGVR) Program, Stanford, CA, USA
| | - Xiaojing J Gao
- Stanford Bio-X, Stanford University, Stanford, CA, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
2
|
Zhang X, Mille-Fragoso LS, Eerik Kaseniit K, Lee AP, Zhang M, Call CC, Hu Y, Xie Y, Gao XJ. Post-Transcriptional Modular Synthetic Receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592453. [PMID: 38746461 PMCID: PMC11092781 DOI: 10.1101/2024.05.03.592453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Inspired by the power of transcriptional synthetic receptors and hoping to complement them to expand the toolbox for cell engineering, we establish LIDAR (Ligand-Induced Dimerization Activating RNA editing), a modular post-transcriptional synthetic receptor platform that harnesses RNA editing by ADAR. LIDAR is compatible with various receptor architectures in different cellular contexts, and enables the sensing of diverse ligands and the production of functional outputs. Furthermore, LIDAR can sense orthogonal signals in the same cell and produce synthetic spatial patterns, potentially enabling the programming of complex multicellular behaviors. Finally, LIDAR is compatible with compact encoding and can be delivered as synthetic mRNA. Thus, LIDAR expands the family of synthetic receptors, holding the promise to empower basic research and therapeutic applications.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of Bioengineering, Stanford University; Stanford, 94305, USA
- Sarafan ChEM-H, Stanford University; Stanford, 94305, USA
| | - Luis S. Mille-Fragoso
- Department of Bioengineering, Stanford University; Stanford, 94305, USA
- Sarafan ChEM-H, Stanford University; Stanford, 94305, USA
- Stanford Bio-X, Stanford University; Stanford, 94305, USA
| | - K. Eerik Kaseniit
- Department of Bioengineering, Stanford University; Stanford, 94305, USA
- Stanford Bio-X, Stanford University; Stanford, 94305, USA
| | - Arden P. Lee
- Sarafan ChEM-H, Stanford University; Stanford, 94305, USA
| | - Meng Zhang
- Department of Chemical Engineering, Stanford University; Stanford, 94305, USA
| | - Connor C. Call
- Department of Chemical Engineering, Stanford University; Stanford, 94305, USA
| | - Yixin Hu
- Department of Biology, Stanford University; Stanford, 94305, USA
| | - Yunxin Xie
- Department of Chemical Engineering, Stanford University; Stanford, 94305, USA
- The Chinese Undergraduate Visiting Research (UGVR) Program; Stanford, 94305, USA
| | - Xiaojing J. Gao
- Department of Chemical Engineering, Stanford University; Stanford, 94305, USA
- Sarafan ChEM-H, Stanford University; Stanford, 94305, USA
- Stanford Bio-X, Stanford University; Stanford, 94305, USA
| |
Collapse
|
3
|
Pu Y, Yang J, Pan Q, Li C, Wang L, Xie X, Chen X, Xiao F, Chen G. MGST3 regulates BACE1 protein translation and amyloidogenesis by controlling the RGS4-mediated AKT signaling pathway. J Biol Chem 2024; 300:107530. [PMID: 38971310 PMCID: PMC11332907 DOI: 10.1016/j.jbc.2024.107530] [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: 03/05/2024] [Revised: 06/03/2024] [Accepted: 06/16/2024] [Indexed: 07/08/2024] Open
Abstract
Microsomal glutathione transferase 3 (MGST3) regulates eicosanoid and glutathione metabolism. These processes are associated with oxidative stress and apoptosis, suggesting that MGST3 might play a role in the pathophysiology of Alzheimer's disease. Here, we report that knockdown (KD) of MGST3 in cell lines reduced the protein level of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) and the resulting amyloidogenesis. Interestingly, MGST3 KD did not alter intracellular reactive oxygen species level but selectively reduced the expression of apoptosis indicators which could be associated with the receptor of cysteinyl leukotrienes, the downstream metabolites of MGST3 in arachidonic acid pathway. We then showed that the effect of MGST3 on BACE1 was independent of cysteinyl leukotrienes but involved a translational mechanism. Further RNA-seq analysis identified that regulator of G-protein signaling 4 (RGS4) was a target gene of MGST3. Silencing of RGS4 inhibited BACE1 translation and prevented MGST3 KD-mediated reduction of BACE1. The potential mechanism was related to AKT activity, as the protein level of phosphorylated AKT was significantly reduced by silencing of MGST3 and RGS4, and the AKT inhibitor abolished the effect of MGST3/RGS4 on phosphorylated AKT and BACE1. Together, MGST3 regulated amyloidogenesis by controlling BACE1 protein expression, which was mediated by RGS4 and downstream AKT signaling pathway.
Collapse
Affiliation(s)
- Yalan Pu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China; Department of Neurology, Langzhong People's Hospital, Nanchong, Sichuan, China
| | - Jie Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China; Affiliated Sichuan Provincial Rehabilitation Hospital of Chengdu University of TCM, Sichuan, China
| | - Qiuling Pan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Chenlu Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Lu Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xiaoyong Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Xue Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Fei Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China.
| | - Guojun Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China.
| |
Collapse
|
4
|
Calbay O, Padia R, Akter M, Sun L, Li B, Qian N, Guo J, Fu Z, Jin L, Huang S. ASC/inflammasome-independent pyroptosis in ovarian cancer cells through translational augmentation of caspase-1. iScience 2023; 26:108408. [PMID: 38058301 PMCID: PMC10696124 DOI: 10.1016/j.isci.2023.108408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/24/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
Canonical pyroptosis is type of programmed cell death depending on active caspase-1, and the inflammasome carries out caspase-1 activation. Here, we showed that docosahexaenoic acid (DHA) induced ovarian cancer cell deaths in caspase-1-dependent manner. DHA increased caspase-1 activity and led to interleukin-1β secretion and gasdermin D cleavage while disulfiram inhibited DHA-induced cell death, suggesting that DHA triggered pyroptosis. Intriguingly, ASC, the molecule recruiting caspase-1 to inflammasome for activation, was dispensable for DHA-induced pyroptosis. Instead, we observed remarkable elevation in caspase-1 abundance concurrent with the activation of caspase-1 in DHA-treated cells. As ectopically overexpressing caspase-1 resulted in robust amount of active caspase-1, we reason that DHA activates caspase-1 and pyroptosis through the generation of excessive amount of caspase-1 protein. Mechanistically, DHA increased caspase-1 by specifically accelerating caspase-1 protein synthesis via the p38MAPK/Mnk1 signaling pathway. We have uncovered an unknown pyroptosis mechanism in which caspase-1-dependent pyroptosis can occur without the participation of ASC/inflammasome.
Collapse
Affiliation(s)
- Ozlem Calbay
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Ravi Padia
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Mahmuda Akter
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Lei Sun
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Bin Li
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Nicole Qian
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Jianhui Guo
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Zheng Fu
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| | - Lingtao Jin
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Shuang Huang
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
5
|
Hu B, Liu T, Wu Z, Phan SH. P53 regulates CCAAT/Enhancer binding protein β gene expression. Gene 2023; 884:147675. [PMID: 37541559 DOI: 10.1016/j.gene.2023.147675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/13/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND The transcription factor CCAAT/enhancer-binding protein β (C/EBPβ) is implicated in diverse processes and diseases. Its two isoforms, namely liver-enriched activator protein (LAP) and liver-enriched inhibitor protein (LIP) are translated from the same mRNA. They share the same C-terminal DNA binding domain except LAP has an extra N-terminal activation domain. Probably due to its higher affinity for its DNA cognate sequences, LIP can inhibit LAP transcriptional activity even at substoichiometric levels. However, the regulatory mechanism of C/EBPβ gene expression and the LAP: LIP ratio is unclear. METHODS In this study, the C/EBPβ promoter sequence was scanned for conserved P53 response element (P53RE), and binding of P53 to the C/EBPβ promoter was tested by Electrophoretic Mobility Shift Assay (EMSA) and chromatin immunoprecipitation assay. P53 over-expression and dominant negative P53 expression plasmids were transfected into rat lung fibroblasts and tested for C/EBPβ gene transcription and expression. Western blot analysis was used to test the regulation of C/EBPβ LAP and LIP isoforms. Constructs containing the LAP 5'untranslated region (5'UTR) or the LIP 5'UTR region were used to test the importance of 5'UTR in the control of C/EBPβ LAP and LIP translation. RESULTS The C/EBPβ promoter sequence was found to contain a conserved P53 response element (P53RE), which binds P53 as demonstrated by Electrophoresis Mobility Shift Assay and chromatin immunoprecipitation assays. P53 over-expression suppressed while dominant negative P53 stimulated C/EBPβ gene transcription and expression. Western blot analysis showed that P53 differentially regulated the translation of the C/EBPβ LAP and LIP isoforms through the regulation of eIF4E and eIF4E-BP1. Further studies with constructs containing the LAP 5'untranslated region (5'UTR) or the LIP 5'UTR region showed that the 5'UTR is important in differential control of C/EBPβ LAP and LIP translation. CONCLUSION Analysis of the effects of P53 on C/EBPβ expression revealed a novel mechanism by which P53 could antagonize the effects of C/EBPβ on its target gene expression. For the first time, P53 is shown to be a repressor of C/EBPβ gene expression at both transcriptional and translational levels, with a differential effect in the magnitude of the effect on LAP vs. LIP isoforms.
Collapse
Affiliation(s)
- Biao Hu
- Department of Internal Medicine, University of Michigan Medical School, 1600 Huron Parkway, Ann Arbor, MI 48109 USA
| | - Tianju Liu
- Department of Pathology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109 USA
| | - Zhe Wu
- Department of Pathology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109 USA
| | - Sem H Phan
- Department of Pathology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109 USA.
| |
Collapse
|
6
|
Istomine R, Al-Aubodah TA, Alvarez F, Smith JA, Wagner C, Piccirillo CA. The eIF4EBP-eIF4E axis regulates CD4 + T cell differentiation through modulation of T cell activation and metabolism. iScience 2023; 26:106683. [PMID: 37187701 PMCID: PMC10176268 DOI: 10.1016/j.isci.2023.106683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/27/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
CD4+ T cells are critical for adaptive immunity, differentiating into distinct effector and regulatory subsets. Although the transcriptional programs underlying their differentiation are known, recent research has highlighted the importance of mRNA translation in determining protein abundance. We previously conducted genome-wide analysis of translation in CD4+ T cells revealing distinct translational signatures distinguishing these subsets, identifying eIF4E as a central differentially translated transcript. As eIF4E is vital for eukaryotic translation, we examined how altered eIF4E activity affected T cell function using mice lacking eIF4E-binding proteins (BP-/-). BP-/- effector T cells showed elevated Th1 responses ex vivo and upon viral challenge with enhanced Th1 differentiation observed in vitro. This was accompanied by increased TCR activation and elevated glycolytic activity. This study highlights how regulating T cell-intrinsic eIF4E activity can influence T cell activation and differentiation, suggesting the eIF4EBP-eIF4E axis as a potential therapeutic target for controlling aberrant T cell responses.
Collapse
Affiliation(s)
- Roman Istomine
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC H4A 3J1, Canada
| | - Tho-Alfakar Al-Aubodah
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC H4A 3J1, Canada
| | - Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC H4A 3J1, Canada
| | - Jacob A. Smith
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Carston Wagner
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Centre of Excellence in Translational Immunology (CETI), Montréal, QC H4A 3J1, Canada
- Corresponding author
| |
Collapse
|
7
|
Christie M, Igreja C. eIF4E-homologous protein (4EHP): a multifarious cap-binding protein. FEBS J 2023; 290:266-285. [PMID: 34758096 DOI: 10.1111/febs.16275] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 02/05/2023]
Abstract
The cap-binding protein 4EHP/eIF4E2 has been a recent object of interest in the field of post-transcriptional gene regulation and translational control. From ribosome-associated quality control, to RNA decay and microRNA-mediated gene silencing, this member of the eIF4E protein family regulates gene expression through numerous pathways. Low in abundance but ubiquitously expressed, 4EHP interacts with different binding partners to form multiple protein complexes that regulate translation in a variety of biological contexts. Documented functions of 4EHP primarily relate to its role as a translational repressor, but recent findings indicate that it might also participate in the activation of translation in specific settings. In this review, we discuss the known functions, properties and mechanisms that involve 4EHP in the control of gene expression. We also discuss our current understanding of how 4EHP processes are regulated in eukaryotic cells, and the diseases implicated with dysregulation of 4EHP-mediated translational control.
Collapse
Affiliation(s)
- Mary Christie
- School of Life and Environmental Sciences, The University of Sydney, NSW, Australia
| | - Cátia Igreja
- Department for Integrative Evolutionary Biology, Max Planck Institute for Developmental Biology, Tübingen, Germany
| |
Collapse
|
8
|
Systems Drug Design for Muscle Invasive Bladder Cancer and Advanced Bladder Cancer by Genome-Wide Microarray Data and Deep Learning Method with Drug Design Specifications. Int J Mol Sci 2022; 23:ijms232213869. [PMID: 36430344 PMCID: PMC9692470 DOI: 10.3390/ijms232213869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Bladder cancer is the 10th most common cancer worldwide. Due to the lack of understanding of the oncogenic mechanisms between muscle-invasive bladder cancer (MIBC) and advanced bladder cancer (ABC) and the limitations of current treatments, novel therapeutic approaches are urgently needed. In this study, we utilized the systems biology method via genome-wide microarray data to explore the oncogenic mechanisms of MIBC and ABC to identify their respective drug targets for systems drug discovery. First, we constructed the candidate genome-wide genetic and epigenetic networks (GWGEN) through big data mining. Second, we applied the system identification and system order detection method to delete false positives in candidate GWGENs to obtain the real GWGENs of MIBC and ABC from their genome-wide microarray data. Third, we extracted the core GWGENs from the real GWGENs by selecting the significant proteins, genes and epigenetics via the principal network projection (PNP) method. Finally, we obtained the core signaling pathways from the corresponding core GWGEN through the annotations of the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway to investigate the carcinogenic mechanisms of MIBC and ABC. Based on the carcinogenic mechanisms, we selected the significant drug targets NFKB1, LEF1 and MYC for MIBC, and LEF1, MYC, NOTCH1 and FOXO1 for ABC. To design molecular drug combinations for MIBC and ABC, we employed a deep neural network (DNN)-based drug-target interaction (DTI) model with drug specifications. The DNN-based DTI model was trained by drug-target interaction databases to predict the candidate drugs for MIBC and ABC, respectively. Subsequently, the drug design specifications based on regulation ability, sensitivity and toxicity were employed as filter criteria for screening the potential drug combinations of Embelin and Obatoclax for MIBC, and Obatoclax, Entinostat and Imiquimod for ABC from their candidate drugs. In conclusion, we not only investigated the oncogenic mechanisms of MIBC and ABC, but also provided promising therapeutic options for MIBC and ABC, respectively.
Collapse
|
9
|
Xu W, Kannan S, Verma CS, Nacro K. Update on the Development of MNK Inhibitors as Therapeutic Agents. J Med Chem 2021; 65:983-1007. [PMID: 34533957 DOI: 10.1021/acs.jmedchem.1c00368] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mitogen-activated protein kinase-interacting kinases 1 and 2 (MNK1/2) represent a central class of enzymes that are activated by extracellular signal-regulated kinase (ERK) or p38 mitogen-activated protein (MAP) kinases. MNK1 and MNK2 coordinate cellular signaling, control production of inflammatory chemokines, and regulate cell proliferation and survival. MNK1/2 are referred to as serine/threonine kinases as they phosphorylate serine or threonine residues on their substrates. Upon activation, MNK1/2 phosphorylate eukaryotic translation initiation factor 4E (eIF4E) at Ser209, which in turn initiates ribosome assembly and protein translation. Deleterious overexpression of MNK1/2 and/or eIF4E have been reported in several diseases including cancers, neurological disorders, autism, and inflammation. Recently, there have been intense efforts toward the development of potent and selective inhibitors of MNK1/2 in both academia and industry. Herein, we review the current understanding of the structural and biological aspects of MNK1/2 and provide an update of pharmacological inhibitors of MNK1/2 including candidates in clinical trials.
Collapse
Affiliation(s)
- Weijun Xu
- Experimental Drug Development Centre (EDDC), A*STAR, 10 Biopolis Road, Chromos #05-01, 138670, Singapore
| | | | - Chandra S Verma
- Bioinformatics Institute (BII), A*STAR, 30 Biopolis Street, #07-01 Matrix, 138671, Singapore.,Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore.,School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore
| | - Kassoum Nacro
- Experimental Drug Development Centre (EDDC), A*STAR, 10 Biopolis Road, Chromos #05-01, 138670, Singapore
| |
Collapse
|
10
|
Evaluation of carboxyfluorescein-labeled 7-methylguanine nucleotides as probes for studying cap-binding proteins by fluorescence anisotropy. Sci Rep 2021; 11:7687. [PMID: 33833335 PMCID: PMC8032668 DOI: 10.1038/s41598-021-87306-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/26/2021] [Indexed: 11/17/2022] Open
Abstract
Fluorescence anisotropy (FA) is a powerful technique for the discovery of protein inhibitors in a high-throughput manner. In this study, we sought to develop new universal FA-based assays for the evaluation of compounds targeting mRNA 5′ cap-binding proteins of therapeutic interest, including eukaryotic translation initiation factor 4E and scavenger decapping enzyme. For this purpose, a library of 19 carboxyfluorescein probes based on 7-methylguanine nucleotides was evaluated as FA probes for these proteins. Optimal probe:protein systems were further investigated in competitive binding experiments and adapted for high-throughput screening. Using a small in-house library of compounds, we verified and confirmed the accuracy of the developed FA assay to study cap-binding protein binders. The applications of the most promising probes were then extended to include evaluation of allosteric inhibitors as well as RNA ligands. From this analysis, we confirmed the utility of the method to study small molecule ligands and evaluate differently 5′ capped RNAs.
Collapse
|
11
|
Khan MA, Malik A, Domashevskiy AV, San A, Khan JM. Interaction of ferritin iron responsive element (IRE) mRNA with translation initiation factor eIF4F. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 243:118776. [PMID: 32829157 DOI: 10.1016/j.saa.2020.118776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 07/26/2020] [Accepted: 07/26/2020] [Indexed: 06/11/2023]
Abstract
The interaction of ferritin iron responsive element (IRE) mRNA with eIF4F was examined by fluorescence and circular dichroism spectroscopy. Fluorescence quenching data indicated that eIF4F contains one high affinity binding site for ferritin IRE RNA. The Scatchard analysis revealed strong binding affinity (Ka = 11.1 × 107 M-1) and binding capacity (n = 1.0) between IRE RNA and eIF4F. The binding affinity of IRE RNA for eIF4F decreased (~4-fold) as temperature increased (from 5 °C to 30 °C). The van't Hoff analysis revealed that IRE RNA binding to eIF4F is enthalpy-driven (ΔH = -47.1 ± 3.4 kJ/mol) and entropy-opposed (ΔS = -30.1 ± 1.5 J/mol/K). The addition of iron increased the enthalpic, while decreasing the entropic contribution towards the eIF4F•IRE RNA complex, resulting in favorable free energy (ΔG = -49.8 ± 2.8 kJ/mol). Thermodynamic values and ionic strength data suggest that the presence of iron increases hydrogen bonding and decreases hydrophobic interactions, leading to formation of a more stable complex. The interaction of IRE RNA with eIF4F at higher concentrations produced significant changes in the secondary structure of the protein, as revealed from the far-UV CD results, clearly illustrating the structural alterations resulted from formation of the eIF4F•IRE RNA complex. A Lineweaver-Burk plot showed an uncompetitive binding behavior between IRE RNA and m7G cap for the eIF4F, indicating that there are different binding sites on the eIF4F for the IRE RNA and the cap analog; molecular docking analysis further supports this notion. Our findings suggest that the eIF4F•IRE RNA complex formation is accompanied by an elevated hydrogen bonding and weakened hydrophobic interactions, leading to an overall conformational change, favored in terms of its free energy. The conformational change in the eIF4F structure, caused by the IRE RNA binding, provides a more stable platform for effective IRE translation in iron homeostasis.
Collapse
Affiliation(s)
- Mateen A Khan
- Department of Life Sciences, College of Science & General Studies, Alfaisal University, Riyadh, Saudi Arabia.
| | - Ajamaluddin Malik
- Department of Biochemistry, Protein Research Laboratory, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Artem V Domashevskiy
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY 10019, USA
| | - Avdar San
- Department of Chemistry, Brooklyn College of the City University of New York, NY, New York, USA
| | - Javed M Khan
- Department of Food Science and Nutrition, Faculty of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
12
|
Yang X, Zhong W, Cao R. Phosphorylation of the mRNA cap-binding protein eIF4E and cancer. Cell Signal 2020; 73:109689. [PMID: 32535199 PMCID: PMC8049097 DOI: 10.1016/j.cellsig.2020.109689] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/21/2020] [Accepted: 06/02/2020] [Indexed: 12/22/2022]
Abstract
Dysregulated protein synthesis is frequently involved in oncogenesis and cancer progression. Translation initiation is thought to be the rate-limiting step in protein synthesis, and the mRNA 5' cap-binding protein eukaryotic translation initiation factor 4E (eIF4E) is a pivotal factor that initiates translation. The activities of eIF4E are regulated at multiple levels, one of which is through its phosphorylation at Serine 209 by the mitogen-activated protein kinase-interacting kinases (MNKs, including MNK1 and MNK2). Benefiting from novel mouse genetic tools and pharmacological MNK inhibitors, our understanding of a role for eIF4E phosphorylation in tumor biology and cancer therapy has greatly evolved in recent years. Importantly, recent studies have found that the level of eIF4E phosphorylation is frequently upregulated in a wide variety of human cancer types, and phosphorylation of eIF4E drives a number of important processes in cancer biology, including cell transformation, proliferation, apoptosis, metastasis and angiogenesis. The MNK-eIF4E axis is being assessed as a therapeutic target either alone or in combination with other therapies in different cancer models. As novel MNK inhibitors are being developed, experimental studies bring new hope to cure human cancers that are not responsive to traditional therapies. Herein we review recent progress on our understanding of a mechanistic role for phosphorylation of eIF4E in cancer biology and therapy.
Collapse
Affiliation(s)
- Xiaotong Yang
- School of Medicine, Tsinghua University, Beijing 100084, China; National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Ruifeng Cao
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN 55812, USA; Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
13
|
Batool A, Majeed ST, Aashaq S, Majeed R, Andrabi KI. Eukaryotic Initiation Factor 4E phosphorylation acts a switch for its binding to 4E-BP1 and mRNA cap assembly. Biochem Biophys Res Commun 2020; 527:489-495. [PMID: 32336547 DOI: 10.1016/j.bbrc.2020.04.086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 04/16/2020] [Indexed: 11/28/2022]
Abstract
Translational regulation has invited considerable interest consequent of its circumstantial dysregulation during cancer genesis. eIF4E (Eukaryotic Initiation Factor 4E) has been identified as an important factor involved in tumor progression by way of instrumenting the convergence of oncogenic signals for up-regulation of Cap-dependent translation. In the backdrop of dramatic eIF4E over-expression in a large population of human cancers, we suggest that the tumorigenic property of eIF4E is strictly attributed to its phosphorylation state. We provide evidence that while phosphorylated eIF4E fails to be sequestered by 4E-BP1, its dephosphorylated form shows overwhelming binding with 4E-BP1 without any consideration to the state of 4E-BP1 phosphorylation to suggest that eIF4E-4EBP1 binding is governed by eIF4E phosphorylation instead of 4E-BP1. We also show that eIF4E engages in Cap-assembly formation preferably in a phosphorylation-dependent manner to suggest that eIF4E phosphorylation rather than 4E-BP1 regulates its availability for Cap-assembly.
Collapse
Affiliation(s)
- Asiya Batool
- Department of Biotechnology, Science Block, University of Kashmir, Srinagar, 190006, J&K, India
| | - Sheikh Tahir Majeed
- Department of Biotechnology, Science Block, University of Kashmir, Srinagar, 190006, J&K, India
| | - Sabreena Aashaq
- Department of Biotechnology, Science Block, University of Kashmir, Srinagar, 190006, J&K, India
| | - Rabiya Majeed
- Department of Biotechnology, Science Block, University of Kashmir, Srinagar, 190006, J&K, India
| | - Khurshid Iqbal Andrabi
- Department of Biotechnology, Science Block, University of Kashmir, Srinagar, 190006, J&K, India.
| |
Collapse
|
14
|
Ala-Poikela M, Rajamäki ML, Valkonen JP. A Novel Interaction Network Used by Potyviruses in Virus-Host Interactions at the Protein Level. Viruses 2019; 11:E1158. [PMID: 31847316 PMCID: PMC6950583 DOI: 10.3390/v11121158] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/30/2022] Open
Abstract
Host proteins that are central to infection of potyviruses (genus Potyvirus; family Potyviridae) include the eukaryotic translation initiation factors eIF4E and eIF(iso)4E. The potyviral genome-linked protein (VPg) and the helper component proteinase (HCpro) interact with each other and with eIF4E and eIF(iso)4E and proteins are involved in the same functions during viral infection. VPg interacts with eIF4E/eIF(iso)4E via the 7-methylguanosine cap-binding region, whereas HCpro interacts with eIF4E/eIF(iso)4E via the 4E-binding motif YXXXXLΦ, similar to the motif in eIF4G. In this study, HCpro and VPg were found to interact in the nucleus, nucleolus, and cytoplasm in cells infected with the potyvirus potato virus A (PVA). In the cytoplasm, interactions between HCpro and VPg occurred in punctate bodies not associated with viral replication vesicles. In addition to HCpro, the 4E-binding motif was recognized in VPg of PVA. Mutations in the 4E-binding motif of VPg from PVA weakened interactions with eIF4E and heavily reduced PVA virulence. Furthermore, mutations in the 4G-binding domain of eIF4E reduced interactions with VPg and abolished interactions with HCpro. Thus, HCpro and VPg can both interact with eIF4E using the 4E-binding motif. Our results suggest a novel interaction network used by potyviruses to interact with host plants via translation initiation factors.
Collapse
Affiliation(s)
| | - Minna-Liisa Rajamäki
- Department of Agricultural Sciences, University of Helsinki, P.O. Box 27, FI-00014 Helsinki, Finland;
| | - Jari P.T. Valkonen
- Department of Agricultural Sciences, University of Helsinki, P.O. Box 27, FI-00014 Helsinki, Finland;
| |
Collapse
|
15
|
Batool A, Majeed ST, Aashaq S, Majeed R, Bhat NN, Andrabi KI. Eukaryotic initiation factor 4E is a novel effector of mTORC1 signaling pathway in cross talk with Mnk1. Mol Cell Biochem 2019; 465:13-26. [DOI: 10.1007/s11010-019-03663-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
|
16
|
Kats IR, Klann E. Translating from cancer to the brain: regulation of protein synthesis by eIF4F. ACTA ACUST UNITED AC 2019; 26:332-342. [PMID: 31416906 PMCID: PMC6699409 DOI: 10.1101/lm.050047.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/03/2019] [Indexed: 12/27/2022]
Abstract
Formation of eukaryotic initiation factor 4F (eIF4F) is widely considered to be the rate-limiting step in cap-dependent translation initiation. Components of eIF4F are often up-regulated in various cancers, and much work has been done to elucidate the role of each of the translation initiation factors in cancer cell growth and survival. In fact, many of the basic mechanisms describing how eIF4F is assembled and how it functions to regulate translation initiation were first investigated in cancer cell lines. These same eIF4F translational control pathways also are relevant for neuronal signaling that underlies long-lasting synaptic plasticity and memory, and in neurological diseases where eIF4F and its upstream regulators are dysregulated. Although eIF4F is important in cancer and for brain function, there is not always a clear path to use the results of studies performed in cancer models to inform one of the roles that the same translation factors have in neuronal signaling. Issues arise when extrapolating from cell lines to tissue, and differences are likely to exist in how eIF4F and its upstream regulatory pathways are expressed in the diverse neuronal subtypes found in the brain. This review focuses on summarizing the role of eIF4F and its accessory proteins in cancer, and how this information has been utilized to investigate neuronal signaling, synaptic function, and animal behavior. Certain aspects of eIF4F regulation are consistent across cancer and neuroscience, whereas some results are more complicated to interpret, likely due to differences in the complexity of the brain, its billions of neurons and synapses, and its diverse cell types.
Collapse
Affiliation(s)
- Ilona R Kats
- Sackler Graduate Program, New York University School of Medicine, New York, New York 10016, USA.,Center for Neural Science, New York University, New York, New York 10003, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, New York 10003, USA.,NYU Neuroscience Institute, New York University School of Medicine, New York, New York 10016, USA
| |
Collapse
|
17
|
Batool A, Aashaq S, Andrabi KI. Eukaryotic initiation factor 4E (eIF4E): A recap of the cap-binding protein. J Cell Biochem 2019; 120:14201-14212. [PMID: 31074051 DOI: 10.1002/jcb.28851] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/29/2022]
Abstract
Eukaryotic initiation factor 4E (eIF4E), a fundamental effector and rate limiting element of protein synthesis, binds the 7-methylguanosine cap at the 5' end of eukaryotic messenger RNA (mRNA) specifically as a constituent of eIF4F translation initiation complex thus facilitating the recruitment of mRNA to the ribosomes. This review focusses on the engagement of signals contributing to growth factor originated maxim and their role in the activation of eIF4E to achieve a collective influence on cellular growth, with a key focus on conjuring vital processes like protein synthesis. The review invites considerable interest in elevating the appeal of eIF4E beyond its role in regulating translation viz a viz cancer genesis, attributed to its phosphorylation state that improves the prospect for the growth of the cancerous cell. This review highlights the latest studies that have envisioned to target these pathways and ultimately the translational machinery for therapeutic intervention. The review also brings forward the prospect of eIF4E to act as a converging juncture for signaling pathways like mTOR/PI3K and Mnk/MAPK to promote tumorigenesis.
Collapse
Affiliation(s)
- Asiya Batool
- Department of Biotechnology and Bioinformatics, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Sabreena Aashaq
- Department of Biotechnology and Bioinformatics, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Khurshid I Andrabi
- Department of Biotechnology and Bioinformatics, University of Kashmir, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
18
|
Zhang-James Y, Vaudel M, Mjaavatten O, Berven FS, Haavik J, Faraone SV. Effect of disease-associated SLC9A9 mutations on protein-protein interaction networks: implications for molecular mechanisms for ADHD and autism. ACTA ACUST UNITED AC 2019; 11:91-105. [PMID: 30927234 DOI: 10.1007/s12402-018-0281-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022]
Abstract
Na+/H+ Exchanger 9 (NHE9) is an endosomal membrane protein encoded by the Solute Carrier 9A, member 9 gene (SLC9A9). SLC9A9 has been implicated in attention deficit hyperactivity disorder (ADHD), autism spectrum disorders (ASDs), epilepsy, multiple sclerosis and cancers. To better understand the function of NHE9 and the effects of disease-associated variants on protein-protein interactions, we conducted a quantitative analysis of the NHE9 interactome using co-immunoprecipitation and isobaric labeling-based quantitative mass spectrometry. We identified 100 proteins that interact with NHE9. These proteins were enriched in known functional pathways for NHE9: the endocytosis, protein ubiquitination and phagosome pathways, as well as some novel pathways including oxidative stress, mitochondrial dysfunction, mTOR signaling, cell death and RNA processing pathways. An ADHD-associated mutation (A409P) significantly altered NHE9's interactions with a subset of proteins involved in caveolae-mediated endocytosis and MAP2K2-mediated downstream signaling. An ASD nonsense mutation in SLC9A9, R423X, produced no-detectable amount of NHE9, suggesting the overall loss of NHE9 functional networks. In addition, seven of the NHE9 interactors are products of known autism candidate genes (Simons Foundation Autism Research Initiative, SFARI Gene) and 90% of the NHE9 interactome overlap with SFARI protein interaction network PIN (p < 0.0001), supporting the role of NHE9 interactome in ASDs molecular mechanisms. Our results provide a detailed understanding of the functions of protein NHE9 and its disrupted interactions, possibly underlying ADHD and ASDs. Furthermore, our methodological framework proved useful for functional characterization of disease-associated genetic variants and suggestion of druggable targets.
Collapse
Affiliation(s)
- Yanli Zhang-James
- Departments of Psychiatry, SUNY Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Marc Vaudel
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Olav Mjaavatten
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Frode S Berven
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway.,K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway.,Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Stephen V Faraone
- Departments of Psychiatry, SUNY Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA. .,Neuroscience and Physiology, SUNY Upstate Medical University, 750 East Adams St., Syracuse, NY, 13210, USA.
| |
Collapse
|
19
|
Hassanzadeh G, Naing T, Graber T, Jafarnejad SM, Stojdl DF, Alain T, Holcik M. Characterizing Cellular Responses During Oncolytic Maraba Virus Infection. Int J Mol Sci 2019; 20:ijms20030580. [PMID: 30700020 PMCID: PMC6387032 DOI: 10.3390/ijms20030580] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 02/07/2023] Open
Abstract
The rising demand for powerful oncolytic virotherapy agents has led to the identification of Maraba virus, one of the most potent oncolytic viruses from Rhabdoviridae family which displays high selectivity for killing malignant cells and low cytotoxicity in normal cells. Although the virus is readied to be used for clinical trials, the interactions between the virus and the host cells is still unclear. Using a newly developed interferon-sensitive mutant Maraba virus (MG1), we have identified two key regulators of global translation (4E-BP1 and eIF2α) as being involved in the regulation of protein synthesis in the infected cells. Despite the translational arrest upon viral stress, we showed an up-regulation of anti-apoptotic Bcl-xL protein that provides a survival benefit for the host cell, yet facilitates effective viral propagation. Given the fact that eIF5B canonically regulates 60S ribosome subunit end joining and is able to replace the role of eIF2 in delivering initiator tRNA to the 40S ribosome subunit upon the phosphorylation of eIF2α we have tested whether eIF5B mediates the translation of target mRNAs during MG1 infection. Our results show that the inhibition of eIF5B significantly down-regulates the level of Bcl-xL steady-state mRNA, thus indirectly attenuates viral propagation.
Collapse
Affiliation(s)
- Golnoush Hassanzadeh
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada.
| | - Thet Naing
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada.
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada.
| | - Tyson Graber
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada.
| | - Seyed Mehdi Jafarnejad
- Centre for Cancer Research and Cell Biology (CCRCB), Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, UK.
| | - David F Stojdl
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada.
| | - Tommy Alain
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada.
| | - Martin Holcik
- Molecular Biomedicine Program, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada.
- Department of Health Sciences, Carleton University, Ottawa, ON K1S 5B6, Canada.
| |
Collapse
|
20
|
The effects of kinase modulation on in vitro maturation according to different cumulus-oocyte complex morphologies. PLoS One 2018; 13:e0205495. [PMID: 30308003 PMCID: PMC6181369 DOI: 10.1371/journal.pone.0205495] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 09/26/2018] [Indexed: 01/21/2023] Open
Abstract
Successful production of transgenic pigs requires oocytes with a high developmental competence. However, cumulus-oocyte complexes (COCs) obtained from antral follicles have a heterogeneous morphology. COCs can be classified into one of two classes: class I, with five or more layers of cumulus cells; and class II, with one or two layers of cumulus cells. Activator [e.g., epidermal growth factor (EGF)] or inhibitors (e.g., wortmannin and U0126) are added to modulate kinases in oocytes during meiosis. In the present study, we investigated the effects of kinase modulation on nuclear and cytoplasmic maturation in COCs. Class I COCs showed a significantly higher developmental competence than class II COCs. Moreover, the expression of two kinases, AKT and ERK, differed between class I and class II COCs during in vitro maturation (IVM). Initially, inhibition of the PI3K/AKT signaling pathway in class I COCs during early IVM (0-22 h) decreased developmental parameters, such as blastocyst formation rate, blastomere number, and cell survival. Conversely, EGF-mediated AKT activation in class II COCs enhanced developmental capacity. Regarding the MAPK signaling pathway, inhibition of ERK by U0126 in class II COCs during early IVM impaired developmental competence. However, transient treatment with U0126 in class II COCs increased oocyte maturation and AKT activity, improving embryonic development. Additionally, western blotting showed that inhibition of ERK activity negatively regulated the AKT signaling pathway, indicative of a relationship between AKT and MAPK signaling in the process underlying meiotic progression in pigs. These findings may help increase the developmental competence and utilization rate of pig COCs with regard to the production of transgenic pigs and improve our understanding of kinase-associated meiosis events.
Collapse
|
21
|
Freire ER, Moura DMN, Bezerra MJR, Xavier CC, Morais-Sobral MC, Vashisht AA, Rezende AM, Wohlschlegel JA, Sturm NR, de Melo Neto OP, Campbell DA. Trypanosoma brucei EIF4E2 cap-binding protein binds a homolog of the histone-mRNA stem-loop-binding protein. Curr Genet 2017; 64:821-839. [DOI: 10.1007/s00294-017-0795-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/01/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022]
|
22
|
BRD7 regulates the insulin-signaling pathway by increasing phosphorylation of GSK3β. Cell Mol Life Sci 2017; 75:1857-1869. [PMID: 29127434 DOI: 10.1007/s00018-017-2711-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 10/18/2022]
Abstract
Reduced hepatic expression levels of bromodomain-containing protein 7 (BRD7) have been suggested to play a role in the development of glucose intolerance in obesity. However, the molecular mechanism by which BRD7 regulates glucose metabolism has remained unclear. Here, we show that BRD7 increases phosphorylation of glycogen synthase kinase 3β (GSK3β) in response to activation of the insulin receptor-signaling pathway shortly after insulin stimulation and the nutrient-sensing pathway after feeding. BRD7 mediates phosphorylation of GSK3β at the Serine 9 residue and this effect on GSK3β occurs even in the absence of AKT activity. Using both in vitro and in vivo models, we further demonstrate that BRD7 mediates phosphorylation of ribosomal protein S6 kinase (S6K) and leads to increased phosphorylation of the eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) and, therefore, relieves its inhibition of the eukaryotic translation initiation factor 4E (eIF4E). However, the increase in phosphorylation of 4E-BP1 with BRD7 overexpression is blunted in the absence of AKT activity. In addition, using liver-specific BRD7 knockout (LBKO) mice, we show that BRD7 is required for mTORC1 activity on its downstream molecules. These findings show a novel basis for understanding the molecular dynamics of glucose metabolism and suggest the unique function of BRD7 in the regulation of glucose homeostasis.
Collapse
|
23
|
Li G, Chong T, Xiang X, Yang J, Li H. Downregulation of microRNA-15a suppresses the proliferation and invasion of renal cell carcinoma via direct targeting of eIF4E. Oncol Rep 2017; 38:1995-2002. [PMID: 28849086 PMCID: PMC5652948 DOI: 10.3892/or.2017.5901] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
The downregulation of microRNA-15a has been reported in several human tumors. However, its expression and functional importance in renal cell carcinoma (RCC) remain unknown. The aim of the present study was to investigate its expression, biological functions and underlying mechanisms in RCC tumorigenesis. The expression levels of miR-15a were examined by qRT-PCR in 40 RCC specimens and adjacent‑paired normal tissues. Cell Counting Kit-8 (CCK-8), colony formation, flow cytometry and Transwell assays were used to explore the potential influence of miR-15a transfection on RCC cell proliferation, the cell cycle, cell apoptosis, and cell invasion. Luciferase reporter assays were performed to confirm the potential target of miR-15a, in combination with qRT-PCR, western blotting and immunohistochemical assays. We found that miR-15a was significantly downregulated in most RCC specimens compared with adjacent normal tissues (P<0.01). Overexpression of miR-15a inhibited cellular growth, suppressed invasion and arrested cells at the G1/G0 phase, and induced cell apoptosis in RCC cells. Luciferase assays revealed that miR-15a directly targeted the binding site of the 3'-untranslated region (3'-UTR) of eIF4E, and inhibited its expression at both mRNA and protein levels. eIF4E expression was negatively associated with miR-15a expression in RCC tissues. eIF4E overexpression treatment partially abrogated the inhibitory effect of miR-15a on cell proliferation and invasion, as well as inactivated P13K/AKT/mTOR signaling in RCC cells. In conclusion, the present study indicated that miR-15a downregulation was associated with cell proliferation and invasion by directly targeting eIF4E during RCC progression. Thus, it may serve as a potential tumor suppressor and therapeutic target for the treatment of RCC.
Collapse
Affiliation(s)
- Gang Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Tie Chong
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xiaolong Xiang
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jie Yang
- Department of Nursing, Xi'an Beifang Chinese Medicine Skin Disease Hospital, Xi'an, Shaanxi 710002, P.R. China
| | - Hongliang Li
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
24
|
Cheng N, Alshammari F, Hughes E, Khanbabaei M, Rho JM. Dendritic overgrowth and elevated ERK signaling during neonatal development in a mouse model of autism. PLoS One 2017; 12:e0179409. [PMID: 28609458 PMCID: PMC5469475 DOI: 10.1371/journal.pone.0179409] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 05/30/2017] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (hereafter referred to as “ASD”) is a heterogeneous neurodevelopmental condition characterized by impaired social communication and interactions, and restricted, repetitive activities or interests. Alterations in network connectivity and memory function are frequently observed in autism patients, often involving the hippocampus. However, specific changes during early brain development leading to disrupted functioning remain largely unclear. Here, we investigated the development of dendritic arbor of hippocampal CA1 pyramidal neurons in the BTBR T+tf/J (BTBR) mouse model of autism. BTBR mice display the defining behavioural features of autism, and also exhibit impaired learning and memory. We found that compared to control C57BL/6J (B6) animals, the lengths of both apical and basal dendrites were significantly greater in neonatal BTBR animals. Further, basal dendrites in the BTBR mice had higher branching complexity. In contrast, cross-sectional area of the soma was unchanged. In addition, we observed a similar density of CA1 pyramidal neurons and thickness of the neuronal layer between the two strains. Thus, there was a specific, compartmentalized overgrowth of dendrites during early development in the BTBR animals. Biochemical analysis further showed that the extracellular signal-regulated kinases (ERK) pathway was up-regulated in the hippocampus of neonatal BTBR animals. Since dendritic structure is critical for information integration and relay, our data suggest that altered development of dendrites could potentially contribute to impaired hippocampal function and behavior observed in the BTBR model, and that this might be related to increased activation of the ERK pathway.
Collapse
Affiliation(s)
- Ning Cheng
- Developmental Neurosciences Research Program, Alberta Children’s Hospital Research Institute (ACHRI), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| | - Fawaz Alshammari
- O’Brien Centre for the Bachelor of Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Elizabeth Hughes
- Developmental Neurosciences Research Program, Alberta Children’s Hospital Research Institute (ACHRI), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Maryam Khanbabaei
- Developmental Neurosciences Research Program, Alberta Children’s Hospital Research Institute (ACHRI), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jong M. Rho
- Departments of Pediatrics, Clinical Neurosciences, Physiology & Pharmacology, Alberta Children’s Hospital Research Institute (ACHRI), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
25
|
Doll S, Urisman A, Oses-Prieto JA, Arnott D, Burlingame AL. Quantitative Proteomics Reveals Fundamental Regulatory Differences in Oncogenic HRAS and Isocitrate Dehydrogenase (IDH1) Driven Astrocytoma. Mol Cell Proteomics 2017; 16:39-56. [PMID: 27834733 PMCID: PMC5217781 DOI: 10.1074/mcp.m116.063883] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/04/2016] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiformes (GBMs) are high-grade astrocytomas and the most common brain malignancies. Primary GBMs are often associated with disturbed RAS signaling, and expression of oncogenic HRAS results in a malignant phenotype in glioma cell lines. Secondary GBMs arise from lower-grade astrocytomas, have slower progression than primary tumors, and contain IDH1 mutations in over 70% of cases. Despite significant amount of accumulating genomic and transcriptomic data, the fundamental mechanistic differences of gliomagenesis in these two types of high-grade astrocytoma remain poorly understood. Only a few studies have attempted to investigate the proteome, phosphorylation signaling, and epigenetic regulation in astrocytoma. In the present study, we applied quantitative phosphoproteomics to identify the main signaling differences between oncogenic HRAS and mutant IDH1-driven glioma cells as models of primary and secondary GBM, respectively. Our analysis confirms the driving roles of the MAPK and PI3K/mTOR signaling pathways in HRAS driven cells and additionally uncovers dysregulation of other signaling pathways. Although a subset of the signaling changes mediated by HRAS could be reversed by a MEK inhibitor, dual inhibition of MEK and PI3K resulted in more complete reversal of the phosphorylation patterns produced by HRAS expression. In contrast, cells expressing mutant IDH1 did not show significant activation of MAPK or PI3K/mTOR pathways. Instead, global downregulation of protein expression was observed. Targeted proteomic analysis of histone modifications identified significant histone methylation, acetylation, and butyrylation changes in the mutant IDH1 expressing cells, consistent with a global transcriptional repressive state. Our findings offer novel mechanistic insight linking mutant IDH1 associated inhibition of histone demethylases with specific histone modification changes to produce global transcriptional repression in secondary glioblastoma. Our proteomic datasets are available for download and provide a comprehensive catalogue of alterations in protein abundance, phosphorylation, and histone modifications in oncogenic HRAS and IDH1 driven astrocytoma cells beyond the transcriptomic level.
Collapse
Affiliation(s)
- Sophia Doll
- From the ‡Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158-2517 California
| | - Anatoly Urisman
- From the ‡Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158-2517 California
| | - Juan A Oses-Prieto
- From the ‡Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158-2517 California
| | - David Arnott
- §Department of Protein Chemistry, Genentech Inc, South San Francisco, 94158-2517 California
| | - Alma L Burlingame
- From the ‡Department of Pharmaceutical Chemistry, University of California, San Francisco, 94158-2517 California;
| |
Collapse
|
26
|
Fan W, Wang W, Mao X, Chu S, Feng J, Xiao D, Zhou J, Fan S. Elevated levels of p-Mnk1, p-eIF4E and p-p70S6K proteins are associated with tumor recurrence and poor prognosis in astrocytomas. J Neurooncol 2016; 131:485-493. [PMID: 27900644 DOI: 10.1007/s11060-016-2327-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 11/08/2016] [Indexed: 10/20/2022]
Abstract
Malignant astrocytomas are able to invade neighboring and distant areas of the normal brain. Signaling pathway alterations play important role in the development of astrocytomas. Deregulation of eukaryotic translation initiation factor 4E (eIF4E) by MAP kinase-interacting kinases (Mnk) on Ser-209 directly or PI3K/mTOR/S6K pathway indirectly has a critical effect on promoting cellular proliferation, malignant transformation and metastasis. We examined and analyzed the correlation between expression of p-Mnk1, p-eIF4E and p-p70S6K proteins and clinicopathological features in 103 astrocytomas and 54 non-tumorous brain tissues. The results indicated that positive percentage of overexpression of p-Mnk1 and p-eIF4E proteins in astrocytomas were significantly higher than that of in the non-tumorous brain tissues (P < 0.05). Elevated p-Mnk1 and p-eIF4E and co-overexpressed three proteins were associated with tumor recurrence (P = 0.003, P = 0.006, P = 0.007, respectively). Overexpressed p-eIF4E significantly correlated with the tumor size (P = 0.019). In addition, overexpression of p-eIF4E and three proteins common expression were related to the WHO grade of astrocytomas (P = 0.001, P = 0.044 respectively). Spearman's rank correlation test further showed that the expression of p-Mnk1 was strongly positive correlated with the expression of p-eIF4E in astrocytomas (r = 0.294, P = 0.003). Besides, overexpression of p-eIF4E and co-expression of p-Mnk1, p-eIF4E and p-p70S6K proteins were inversely correlated with overall survival rates of astrocytomas. Multivariate Cox regression analysis further identified that the elevated p-eIF4E expression, three proteins common expression were correlated with unfavorable prognosis of astrocytomas regardless of ages and WHO grades. Taken together, overexpression of p-eIF4E and co-expression of p-Mnk1, p-eIF4E and p-p70S6K proteins could be used as novel independent poor prognostic biomarkers for patients with astrocytomas.
Collapse
Affiliation(s)
- Weibing Fan
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410011, Hunan, China
| | - Weiyuan Wang
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| | - Xinfa Mao
- Department of Neurology, The Third Hospital of Changsha, Changsha, 410011, Hunan, China
| | - Shuzhou Chu
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Juan Feng
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Desheng Xiao
- Department of Pathology, Xiangya School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Jianhua Zhou
- Department of Pathology, Xiangya School of Medicine, Central South University, Changsha, 410011, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
27
|
McGlory C, Devries MC, Phillips SM. Skeletal muscle and resistance exercise training; the role of protein synthesis in recovery and remodeling. J Appl Physiol (1985) 2016; 122:541-548. [PMID: 27742803 DOI: 10.1152/japplphysiol.00613.2016] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/04/2016] [Accepted: 10/05/2016] [Indexed: 12/22/2022] Open
Abstract
Exercise results in the rapid remodeling of skeletal muscle. This process is underpinned by acute and chronic changes in both gene and protein synthesis. In this short review we provide a brief summary of our current understanding regarding how exercise influences these processes as well as the subsequent impact on muscle protein turnover and resultant shift in muscle phenotype. We explore concepts of ribosomal biogenesis and the potential role of increased translational capacity vs. translational efficiency in contributing to muscular hypertrophy. We also examine whether high-intensity sprinting-type exercise promotes changes in protein turnover that lead to hypertrophy or merely a change in mitochondrial content. Finally, we propose novel areas for future study that will fill existing knowledge gaps in the fields of translational research and exercise science.
Collapse
Affiliation(s)
- Chris McGlory
- Department of Kinesiology, McMaster University, Ontario, Canada
| | | | | |
Collapse
|
28
|
Chu J, Cargnello M, Topisirovic I, Pelletier J. Translation Initiation Factors: Reprogramming Protein Synthesis in Cancer. Trends Cell Biol 2016; 26:918-933. [PMID: 27426745 DOI: 10.1016/j.tcb.2016.06.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/03/2016] [Accepted: 06/13/2016] [Indexed: 12/11/2022]
Abstract
Control of mRNA translation plays a crucial role in the regulation of gene expression and is critical for cellular homeostasis. Dysregulation of translation initiation factors has been documented in several pathologies including cancer. Aberrant function of translation initiation factors leads to translation reprogramming that promotes proliferation, survival, angiogenesis, and metastasis. In such context, understanding how altered levels (and presumably activity) of initiation factors can contribute to tumor initiation and/or maintenance is of major interest for the development of novel therapeutic strategies. In this review we provide an overview of translation initiation mechanisms and focus on recent findings describing the role of individual initiation factors and their aberrant activity in cancer.
Collapse
Affiliation(s)
- Jennifer Chu
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Marie Cargnello
- Lady Davis Institute, SMBD JGH, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Quebec, Canada
| | - Ivan Topisirovic
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada; Lady Davis Institute, SMBD JGH, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Quebec, Canada.
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, McGill University, Quebec, Canada; The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
29
|
ur Rasool R, Rah B, Amin H, Nayak D, Chakraborty S, Rawoof A, Mintoo MJ, Yousuf K, Mukherjee D, Kumar LD, Mondhe DM, Goswami A. Dual modulation of Ras-Mnk and PI3K-AKT-mTOR pathways: A Novel c-FLIP inhibitory mechanism of 3-AWA mediated translational attenuation through dephosphorylation of eIF4E. Sci Rep 2016; 6:18800. [PMID: 26728896 PMCID: PMC4700468 DOI: 10.1038/srep18800] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/24/2015] [Indexed: 12/19/2022] Open
Abstract
The eukaryotic translation initiation factor 4E (eIF4E) is considered as a key survival protein involved in cell cycle progression, transformation and apoptosis resistance. Herein, we demonstrate that medicinal plant derivative 3-AWA (from Withaferin A) suppressed the proliferation and metastasis of CaP cells through abrogation of eIF4E activation and expression via c-FLIP dependent mechanism. This translational attenuation prevents the de novo synthesis of major players of metastatic cascades viz. c-FLIP, c-Myc and cyclin D1. Moreover, the suppression of c-FLIP due to inhibition of translation initiation complex by 3-AWA enhanced FAS trafficking, BID and caspase 8 cleavage. Further ectopically restored c-Myc and GFP-HRas mediated activation of eIF4E was reduced by 3-AWA in transformed NIH3T3 cells. Detailed underlying mechanisms revealed that 3-AWA inhibited Ras-Mnk and PI3-AKT-mTOR, two major pathways through which eIF4E converges upon eIF4F hub. In addition to in vitro studies, we confirmed that 3-AWA efficiently suppressed tumor growth and metastasis in different mouse models. Given that 3-AWA inhibits c-FLIP through abrogation of translation initiation by co-targeting mTOR and Mnk-eIF4E, it (3-AWA) can be exploited as a lead pharmacophore for promising anti-cancer therapeutic development.
Collapse
Affiliation(s)
- Reyaz ur Rasool
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Bilal Rah
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Hina Amin
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Debasis Nayak
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Souneek Chakraborty
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Abdul Rawoof
- Center for Cellular and Molecular Biology, Uppal Road, Hyderabad, AP-50007, India
| | - Mubashir Javed Mintoo
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Khalid Yousuf
- Natural Product Chemistry, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Debaraj Mukherjee
- Natural Product Chemistry, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Lekha Dinesh Kumar
- Center for Cellular and Molecular Biology, Uppal Road, Hyderabad, AP-50007, India
| | - Dilip Manikaro Mondhe
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| | - Anindya Goswami
- Academy of Scientific & Innovative Research (AcSIR), New Delhi, India
- Cancer Pharmacology Division, Indian Institute of Integrative Medicine (CSIR), Canal Road, Jammu Tawi, J&K – 180001, India
| |
Collapse
|
30
|
Erdogan CS, Hansen BW, Vang O. Are invertebrates relevant models in ageing research? Focus on the effects of rapamycin on TOR. Mech Ageing Dev 2016; 153:22-9. [PMID: 26763146 DOI: 10.1016/j.mad.2015.12.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/24/2015] [Accepted: 12/29/2015] [Indexed: 12/16/2022]
Abstract
Ageing is the organisms increased susceptibility to death, which is linked to accumulated damage in the cells and tissues. Ageing is a complex process regulated by crosstalk of various pathways in the cells. Ageing is highly regulated by the Target of Rapamycin (TOR) pathway activity. TOR is an evolutionary conserved key protein kinase in the TOR pathway that regulates growth, proliferation and cell metabolism in response to nutrients, growth factors and stress. Comparing the ageing process in invertebrate model organisms with relatively short lifespan with mammals provides valuable information about the molecular mechanisms underlying the ageing process faster than mammal systems. Inhibition of the TOR pathway activity via either genetic manipulation or rapamycin increases lifespan profoundly in most invertebrate model organisms. This contribution will review the recent findings in invertebrates concerning the TOR pathway and effects of TOR inhibition by rapamycin on lifespan. Besides some contradictory results, the majority points out that rapamycin induces longevity. This suggests that administration of rapamycin in invertebrates is a promising tool for pursuing the scientific puzzle of lifespan prolongation.
Collapse
Affiliation(s)
- Cihan Suleyman Erdogan
- Roskilde University, Department of Science and Environment, Universitetsvej 1, DK-4000, Denmark
| | - Benni Winding Hansen
- Roskilde University, Department of Science and Environment, Universitetsvej 1, DK-4000, Denmark
| | - Ole Vang
- Roskilde University, Department of Science and Environment, Universitetsvej 1, DK-4000, Denmark.
| |
Collapse
|
31
|
Purine nucleosides in neuroregeneration and neuroprotection. Neuropharmacology 2015; 104:226-42. [PMID: 26577017 DOI: 10.1016/j.neuropharm.2015.11.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 12/20/2022]
Abstract
In the present review, we stress the importance of the purine nucleosides, adenosine and guanosine, in protecting the nervous system, both centrally and peripherally, via activation of their receptors and intracellular signalling mechanisms. A most novel part of the review focus on the mechanisms of neuronal regeneration that are targeted by nucleosides, including a recently identified action of adenosine on axonal growth and microtubule dynamics. Discussion on the role of the purine nucleosides transversally with the most established neurotrophic factors, e.g. brain derived neurotrophic factor (BDNF), glial derived neurotrophic factor (GDNF), is also focused considering the intimate relationship between some adenosine receptors, as is the case of the A2A receptors, and receptors for neurotrophins. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
|
32
|
Wang Y, Arribas-Layton M, Chen Y, Lykke-Andersen J, Sen GL. DDX6 Orchestrates Mammalian Progenitor Function through the mRNA Degradation and Translation Pathways. Mol Cell 2015; 60:118-30. [PMID: 26412305 DOI: 10.1016/j.molcel.2015.08.014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 06/11/2015] [Accepted: 08/18/2015] [Indexed: 12/26/2022]
Abstract
In adult tissues, stem and progenitor cells must balance proliferation and differentiation to maintain homeostasis. How this is done is unclear. Here, we show that the DEAD box RNA helicase, DDX6 is necessary for maintaining adult progenitor cell function. DDX6 loss results in premature differentiation and decreased proliferation of epidermal progenitor cells. To maintain self-renewal, DDX6 associates with YBX1 to bind the stem loops found in the 3' UTRs of regulators of proliferation/self-renewal (CDK1, EZH2) and recruit them to EIF4E to facilitate their translation. To prevent premature differentiation of progenitor cells, DDX6 regulates the 5' UTR of differentiation inducing transcription factor, KLF4 and degrades its transcripts through association with mRNA degradation proteins. Our results demonstrate that progenitor function is maintained by DDX6 complexes through two distinct pathways that include the degradation of differentiation-inducing transcripts and by promoting the translation of self-renewal and proliferation mRNAs.
Collapse
Affiliation(s)
- Ying Wang
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093-0869, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0869, USA; UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - Marc Arribas-Layton
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - Yifang Chen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093-0869, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0869, USA; UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - Jens Lykke-Andersen
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0869, USA
| | - George L Sen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093-0869, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093-0869, USA; UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093-0869, USA.
| |
Collapse
|
33
|
Liu F, Wang X, Li J, Gu K, Lv L, Zhang S, Che D, Cao J, Jin S, Yu Y. miR-34c-3p functions as a tumour suppressor by inhibiting eIF4E expression in non-small cell lung cancer. Cell Prolif 2015; 48:582-92. [PMID: 26250586 DOI: 10.1111/cpr.12201] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/15/2015] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally regulate gene expression and mediate diverse physiological processes. In this study, we investigated functions of miRNA miR-34c-3p in non-small cell lung cancer (NSCLC). MATERIALS AND METHODS miR-34c-3p expression was evaluated by qPCR. Cell viability was examined by MTT and proliferation by cell cycle analysis. Cell migration and invasion were tested using Transwells with/without Matrigel coating. Western blot analysis was performed for eIF4E, c-Myc, Cyclin D1, survivin and Mcl-1 protein expression. RESULTS miR-34c-3p expression was significantly reduced in tissues and serum samples from NSCLC patients and in NSCLC cell lines A549, H460, H23, H157 and H1299. Overexpression of miR-34c-3p in A549 and H157 cells reduced cell proliferation, migration and invasion, whereas transfection with miR-34c-3p inhibitor (miR-34c-3p-in) produced opposite effects. Target analysis using algorithms miRanda, TargetScan and DIANA identified eIF4E as a potential target of miR-34c-3p. Luciferase assay using the eIF4E 3'-UTR reporter carrying a putative miR-34c-3p target sequence revealed eIF4E to be a specific target of miR-34c-3p. Overexpression of miR-34c-3p in NSCLS cell lines led to significant reduction in mRNA and protein levels of eIF4E, whereas inhibition of miR-34c-3p resulted in significant increase in eIf4e protein levels, confirming eIF4E to be a direct target of miR-34c-3p in NSCLS. Overexpression of eIF4E in A549 cells promoted cell proliferation, migration and invasion, which were partially reversed by miR-34c-3p. CONCLUSION miR-34c-3p directly targeted eIF4E and reduced miR-34c-3p expression in NSCLC, promoting cell cycle progression, proliferation, migration and invasion.
Collapse
Affiliation(s)
- Fang Liu
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Xuefeng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Jiebing Li
- Medical Imaging Division, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Kuo Gu
- The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Liyan Lv
- Department of Medical Oncology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Shuai Zhang
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Dehai Che
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Jingyan Cao
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Shi Jin
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| | - Yan Yu
- Department of Medical Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
34
|
Integrin β6 can be translationally regulated by eukaryotic initiation factor 4E: Contributing to colonic tumor malignancy. Tumour Biol 2015; 36:6541-50. [PMID: 25982998 DOI: 10.1007/s13277-015-3348-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 03/16/2015] [Indexed: 01/09/2023] Open
Abstract
It is well known that both eukaryotic initiation factor 4E (eIF4E) and integrin αvβ6 can contribute to malignant behavior of colon cancer. We have found that integrin αvβ6 and eIF4E were co-expressed and positively correlated in colon cancer tissues. Recently, deregulation of the protein synthesis apparatus has begun to gain attention as a major participant in cancer development and progression. However, the regulation of integrin β6 expression at translational level has never been investigated before. In present study, gene-silencing technique for eIF4E by small interfering RNA (siRNA) was used in all the subsequent experiments, in order to investigate whether eIF4E could translationally regulate expression of integrin β6 in colon cancer SW480 and HT-29 cell lines. Additionally, the subsequent effects of eIF4E knockdown on cellular malignant behavior were observed. siRNA in SW480 and HT-29 transfectants. Subsequently, protein expression of β6 was markedly suppressed, while mRNA expression of β6 showed no significant variation before and after eIF4E RNA interfering. Therefore, it could be seen that eIF4E could upregulate the expression of β6, without effect on β6 mRNA expression. More importantly, after treated with eIF4E siRNA, cellular migratory capacity on fibronectin of HT-29 and β6-transfected SW480 as well as their survival to 5-FU was decreased distinctly. Expression of integrin β6 could be translationally regulated by eIF4E, which subsequently contributed to tumor malignancy through enhancing cellular migration, survival, anti-apoptosis, and chemoresistance of colon cancer in vitro. Thus, targeting eIF4E in integrin αvβ6 expressing tumors can be a potential therapeutic strategy for patients with colon cancer.
Collapse
|
35
|
Abstract
Dysregulation of mRNA translation is a frequent feature of neoplasia. Many oncogenes and tumour suppressors affect the translation machinery, making aberrant translation a widespread characteristic of tumour cells, independent of the genetic make-up of the cancer. Therefore, therapeutic agents that target components of the protein synthesis apparatus hold promise as novel anticancer drugs that can overcome intra-tumour heterogeneity. In this Review, we discuss the role of translation in cancer, with a particular focus on the eIF4F (eukaryotic translation initiation factor 4F) complex, and provide an overview of recent efforts aiming to 'translate' these results to the clinic.
Collapse
|
36
|
eIF4E as a control target for viruses. Viruses 2015; 7:739-50. [PMID: 25690796 PMCID: PMC4353914 DOI: 10.3390/v7020739] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/06/2015] [Accepted: 02/11/2015] [Indexed: 01/04/2023] Open
Abstract
Translation is a complex process involving diverse cellular proteins, including the translation initiation factor eIF4E, which has been shown to be a protein that is a point for translational regulation. Viruses require components from the host cell to complete their replication cycles. Various studies show how eIF4E and its regulatory cellular proteins are manipulated during viral infections. Interestingly, viral action mechanisms in eIF4E are diverse and have an impact not only on viral protein synthesis, but also on other aspects that are important for the replication cycle, such as the proliferation of infected cells and stimulation of viral reactivation. This review shows how some viruses use eIF4E and its regulatory proteins for their own benefit in order to spread themselves.
Collapse
|
37
|
Santini E, Klann E. Reciprocal signaling between translational control pathways and synaptic proteins in autism spectrum disorders. Sci Signal 2014; 7:re10. [PMID: 25351249 DOI: 10.1126/scisignal.2005832] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Autism spectrum disorder (ASD) is a heterogeneous group of heritable neurodevelopmental disorders. Symptoms of ASD, which include deficits in social interaction skills, impaired communication ability, and ritualistic-like repetitive behaviors, appear in early childhood and continue throughout life. Genetic studies have revealed at least two clusters of genes frequently associated with ASD and intellectual disability: those encoding proteins involved in translational control and those encoding proteins involved in synaptic function. We hypothesize that mutations occurring in these two clusters of genes interfere with interconnected downstream signaling pathways in neuronal cells to cause ASD symptomatology. In this review, we discuss the monogenic forms of ASD caused by mutations in genes encoding for proteins that regulate translation and synaptic function. Specifically, we describe the function of these proteins, the intracellular signaling pathways that they regulate, and the current mouse models used to characterize the synaptic and behavioral features associated with their mutation. Finally, we summarize recent studies that have established a connection between mRNA translation and synaptic function in models of ASD and propose that dysregulation of one has a detrimental impact on the other.
Collapse
Affiliation(s)
- Emanuela Santini
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Eric Klann
- Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
38
|
Lim JH, Lee YM, Lee G, Choi YJ, Lim BO, Kim YJ, Choi DK, Park JW. PRMT5 is essential for the eIF4E-mediated 5'-cap dependent translation. Biochem Biophys Res Commun 2014; 452:1016-21. [PMID: 25234597 DOI: 10.1016/j.bbrc.2014.09.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 09/09/2014] [Indexed: 11/16/2022]
Abstract
It is becoming clear that PRMT5 plays essential roles in cell cycle progression, survival, and responses to external stresses. However, the precise mechanisms underlying such roles of PRMT5 have not been clearly understood. Previously, we have demonstrated that PRMT5 participates in cellular adaptation to hypoxia by ensuring 5'-cap dependent translation of HIF-1α. Given that c-Myc and cyclin D1 expressions are also tightly regulated in 5'-cap dependent manner, we here tested the possibility that PRMT5 promotes cell proliferation by increasing de novo syntheses of the oncoproteins. c-Myc and cyclin D1 were found to be noticeably downregulated by PRMT5 knock-down. A RNA immunoprecipitation analysis, which can identify RNA-protein interactions, showed that PRMT5 is required for the interaction among eIF4E and 5'-UTRs of HIF-1α, c-Myc and cyclin D1 mRNAs. In addition, PRMT5 knock-down inhibited cell proliferation by inducing cell cycle arrest at the G1 phase. More importantly, ectopic expression of eIF4E significantly rescued the cell cycle progression and cell proliferation even in PRMT5-deficeint condition. Based on these results, we propose that PRMT5 determines cell fate by regulating 5'-cap dependent translation of proteins essential for proliferation and survival.
Collapse
Affiliation(s)
- Ji-Hong Lim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 380-701, Chungbuk, Republic of Korea
| | - Yoon-Mi Lee
- Department of Food Bioscience, College of Biomedical & Health Science, Konkuk University, Chungju 380-701, Chungbuk, Republic of Korea
| | - Gibok Lee
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 380-701, Chungbuk, Republic of Korea
| | - Yong-Joon Choi
- Departments of Pharmacology and Biomedical Science, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 110-799, Republic of Korea
| | - Beong-Ou Lim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 380-701, Chungbuk, Republic of Korea
| | - Young Jun Kim
- Department of Biomedical Chemistry, College of Biomedical & Health Science, Konkuk University, Chungju 380-701, Chungbuk, Republic of Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju 380-701, Chungbuk, Republic of Korea
| | - Jong-Wan Park
- Departments of Pharmacology and Biomedical Science, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 110-799, Republic of Korea.
| |
Collapse
|
39
|
Freire ER, Vashisht AA, Malvezzi AM, Zuberek J, Langousis G, Saada EA, Nascimento JDF, Stepinski J, Darzynkiewicz E, Hill K, De Melo Neto OP, Wohlschlegel JA, Sturm NR, Campbell DA. eIF4F-like complexes formed by cap-binding homolog TbEIF4E5 with TbEIF4G1 or TbEIF4G2 are implicated in post-transcriptional regulation in Trypanosoma brucei. RNA (NEW YORK, N.Y.) 2014; 20:1272-86. [PMID: 24962368 PMCID: PMC4105752 DOI: 10.1261/rna.045534.114] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 05/12/2014] [Indexed: 05/19/2023]
Abstract
Members of the eIF4E mRNA cap-binding family are involved in translation and the modulation of transcript availability in other systems as part of a three-component complex including eIF4G and eIF4A. The kinetoplastids possess four described eIF4E and five eIF4G homologs. We have identified two new eIF4E family proteins in Trypanosoma brucei, and define distinct complexes associated with the fifth member, TbEIF4E5. The cytosolic TbEIF4E5 protein binds cap 0 in vitro. TbEIF4E5 was found in association with two of the five TbEIF4Gs. TbIF4EG1 bound TbEIF4E5, a 47.5-kDa protein with two RNA-binding domains, and either the regulatory protein 14-3-3 II or a 117.5-kDa protein with guanylyltransferase and methyltransferase domains in a potentially dynamic interaction. The TbEIF4G2/TbEIF4E5 complex was associated with a 17.9-kDa hypothetical protein and both 14-3-3 variants I and II. Knockdown of TbEIF4E5 resulted in the loss of productive cell movement, as evidenced by the inability of the cells to remain in suspension in liquid culture and the loss of social motility on semisolid plating medium, as well as a minor reduction of translation. Cells appeared lethargic, as opposed to compromised in flagellar function per se. The minimal use of transcriptional control in kinetoplastids requires these organisms to implement downstream mechanisms to regulate gene expression, and the TbEIF4E5/TbEIF4G1/117.5-kDa complex in particular may be a key player in that process. We suggest that a pathway involved in cell motility is affected, directly or indirectly, by one of the TbEIF4E5 complexes.
Collapse
Affiliation(s)
- Eden R Freire
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Ajay A Vashisht
- Department of Biological Chemistry, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Amaranta M Malvezzi
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA Department of Microbiology, Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Pernambuco 50670-420, Brazil
| | - Joanna Zuberek
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-089 Warsaw, Poland
| | - Gerasimos Langousis
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Edwin A Saada
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Janaína De F Nascimento
- Department of Microbiology, Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Pernambuco 50670-420, Brazil
| | - Janusz Stepinski
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-089 Warsaw, Poland
| | - Edward Darzynkiewicz
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-089 Warsaw, Poland Centre of New Technologies, University of Warsaw, 02-089 Warsaw, Poland
| | - Kent Hill
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Osvaldo P De Melo Neto
- Department of Microbiology, Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz, Recife, Pernambuco 50670-420, Brazil
| | - James A Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Nancy R Sturm
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - David A Campbell
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
40
|
Pettersson F, del Rincon SV, Miller WH. Eukaryotic translation initiation factor 4E as a novel therapeutic target in hematological malignancies and beyond. Expert Opin Ther Targets 2014; 18:1035-48. [DOI: 10.1517/14728222.2014.937426] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
41
|
Mahalingam P, Takrouri K, Chen T, Sahoo R, Papadopoulos E, Chen L, Wagner G, Aktas BH, Halperin JA, Chorev M. Synthesis of rigidified eIF4E/eIF4G inhibitor-1 (4EGI-1) mimetic and their in vitro characterization as inhibitors of protein-protein interaction. J Med Chem 2014; 57:5094-111. [PMID: 24827861 PMCID: PMC4216204 DOI: 10.1021/jm401733v] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
![]()
The
4EGI-1 is the prototypic inhibitor of eIF4E/eIF4G interaction,
a potent inhibitor of translation initiation in vitro and in vivo
and an efficacious anticancer agent in animal models of human cancers.
We report on the design, synthesis, and in vitro characterization
of a series of rigidified mimetic of this prototypic inhibitor in
which the phenyl in the 2-(4-(3,4-dichlorophenyl)thiazol-2-yl) moiety
was bridged into a tricyclic system. The bridge consisted one of the
following: ethylene, methylene oxide, methylenesulfide, methylenesulfoxide,
and methylenesulfone. Numerous analogues in this series were found
to be markedly more potent than the parent prototypic inhibitor in
the inhibition of eIF4E/eIF4G interaction, thus preventing the eIF4F
complex formation, a rate limiting step in the translation initiation
cascade in eukaryotes, and in inhibition of human cancer cell proliferation.
Collapse
Affiliation(s)
- Poornachandran Mahalingam
- Laboratory for Translational Research, Harvard Medical School , One Kendall Square, Cambridge, Massachusetts 02139, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
León K, Boulo T, Musnier A, Morales J, Gauthier C, Dupuy L, Heyne S, Backofen R, Poupon A, Cormier P, Reiter E, Crepieux P. Activation of a GPCR leads to eIF4G phosphorylation at the 5' cap and to IRES-dependent translation. J Mol Endocrinol 2014; 52:373-82. [PMID: 24711644 DOI: 10.1530/jme-14-0009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The control of mRNA translation has been mainly explored in response to activated tyrosine kinase receptors. In contrast, mechanistic details on the translational machinery are far less available in the case of ligand-bound G protein-coupled receptors (GPCRs). In this study, using the FSH receptor (FSH-R) as a model receptor, we demonstrate that part of the translational regulations occurs by phosphorylation of the translation pre-initiation complex scaffold protein, eukaryotic initiation factor 4G (eIF4G), in HEK293 cells stably expressing the FSH-R. This phosphorylation event occurred when eIF4G was bound to the mRNA 5' cap, and probably involves mammalian target of rapamycin. This regulation might contribute to cap-dependent translation in response to FSH. The cap-binding protein eIF4E also had its phosphorylation level enhanced upon FSH stimulation. We also show that FSH-induced signaling not only led to cap-dependent translation but also to internal ribosome entry site (IRES)-dependent translation of some mRNA. These data add detailed information on the molecular bases underlying the regulation of selective mRNA translation by a GPCR, and a topological model recapitulating these mechanisms is proposed.
Collapse
Affiliation(s)
- Kelly León
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Thomas Boulo
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Astrid Musnier
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Julia Morales
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, Germany
| | - Christophe Gauthier
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Laurence Dupuy
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Steffen Heyne
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, Germany
| | - Rolf Backofen
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, Germany
| | - Anne Poupon
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Patrick Cormier
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, Germany
| | - Eric Reiter
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| | - Pascale Crepieux
- UMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling Systems (BIOS)»CNRS, UMR7247, F-37380 Nouzilly, FranceUniversité François RabelaisF-37041 Tours, FranceIFCEF-37380 Nouzilly, FranceUniversité Pierre et Marie CurieUniversity of Paris VI, CNRS, UMR 7150 Mer et Santé, Equipe Traduction, Cycle Cellulaire, et Développement, Station Biologique de Roscoff, F-29239 Roscoff, FranceUniversité Européenne de BretagneF-29239 Roscoff, FranceBioinformatics GroupDepartment of Computer Science, University of Freiburg, Freiburg, GermanyUMR85Unité Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, FranceGroup «Biology and Bioinformatics of Signaling System
| |
Collapse
|
43
|
Tang Y, Jacobi A, Vater C, Zou X, Stiehler M. Salvianolic acid B protects human endothelial progenitor cells against oxidative stress-mediated dysfunction by modulating Akt/mTOR/4EBP1, p38 MAPK/ATF2, and ERK1/2 signaling pathways. Biochem Pharmacol 2014; 90:34-49. [PMID: 24780446 DOI: 10.1016/j.bcp.2014.04.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/14/2014] [Accepted: 04/14/2014] [Indexed: 12/01/2022]
Abstract
The vascular endothelium is specifically sensitive to oxidative stress, and this is one of the mechanisms that causes widespread endothelial dysfunction in most cardiovascular diseases and disorders. Protection against reactive oxygen species (ROS)-mediated oxidative damage via antioxidant mechanisms is essential for tissue maintenance and shows therapeutic potential for patients suffering from cardiovascular and metabolic disorders. Salvianolic acid B (SalB), a natural bioactive component known from Traditional Chinese Medicine, has been reported to exert cellular protection in various types of cells. However, the underlying mechanisms involved are not fully understood. Here, we showed that SalB significantly promoted the migratory and tube formation abilities of human bone marrow derived-endothelial progenitor cells (BM-EPCs) in vitro, and substantially abrogated hydrogen peroxide (H2O2)-induced cell damage. SalB down-regulated Nox4 and eNOS, as well as nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase expression upon H2O2 induction that in turn prevents oxidative-induced endothelial dysfunction. Moreover, SalB suppressed the Bax/Bcl-xL ratio and caspase-3 activation after H2O2 induction. Furthermore, our results provide mechanistic evidence that activation of the mTOR/p70S6K/4EBP1 pathways is required for both SalB-mediated angiogenic and protective effects against oxidative stress-induced cell injury in BM-EPCs. Suppression of MKK3/6-p38 MAPK-ATF2 and ERK1/2 signaling pathways by SalB significantly protected BM-EPCs against cell injury caused by oxidative stress via reduction of intracellular ROS levels and apoptosis. Taken together, by providing a mechanistic insight into the modulation of redox states in BM-EPCs by SalB, we suggest that SalB has a strong potential of being a new proangiogenic and cytoprotective therapeutic agent with applications in the field of endothelial injury-mediated vascular diseases.
Collapse
Affiliation(s)
- Yubo Tang
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Centre for Orthopaedics and Trauma Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany; Department of Pharmacy, the First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, China.
| | - Angela Jacobi
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Centre for Orthopaedics and Trauma Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany.
| | - Corina Vater
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Centre for Orthopaedics and Trauma Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany.
| | - Xuenong Zou
- Department of Spinal Surgery, the First Affiliated Hospital of Sun Yat-sen University, 510080 Guangzhou, China.
| | - Maik Stiehler
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Centre for Orthopaedics and Trauma Surgery, University Hospital Carl Gustav Carus at Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
44
|
Abstract
The 7mG (7-methylguanosine cap) formed on mRNA is fundamental to eukaryotic gene expression. Protein complexes recruited to 7mG mediate key processing events throughout the lifetime of the transcript. One of the most important mediators of 7mG functions is CBC (cap-binding complex). CBC has a key role in several gene expression mechanisms, including transcription, splicing, transcript export and translation. Gene expression can be regulated by signalling pathways which influence CBC function. The aim of the present review is to discuss the mechanisms by which CBC mediates and co-ordinates multiple gene expression events.
Collapse
|
45
|
Glutamatergic candidate genes in autism spectrum disorder: an overview. J Neural Transm (Vienna) 2014; 121:1081-106. [PMID: 24493018 DOI: 10.1007/s00702-014-1161-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/13/2014] [Indexed: 12/22/2022]
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental disorders with early onset in childhood. Most of the risk for ASD can be explained by genetic variants that act in interaction with biological environmental risk factors. However, the architecture of the genetic components is still unclear. Genetic studies and subsequent systems biological approaches described converging functional effects of identified genes towards pathways relevant for neuronal signalling. Mouse models suggest an aberrant synaptic plasticity at the neuropathological level, which is believed to be conferred by dysregulation of long-term potentiation or depression of neuronal connections. A central pathway regulating these mechanisms is glutamatergic signalling. Here, we hypothesized that susceptibility genes for ASD are enriched for components of this pathway. To further understand the impact of ASD risk genes on the glutamatergic pathway, we performed a systematic review using the literature database "pubmed" and the "AutismKB" knowledgebase. We provide an overview of the glutamatergic system in typical brain function and development, and summarize findings from linkage, association, copy number variants, and sequencing studies in ASD to provide a comprehensive picture of the glutamatergic landscape of ASD genetics. Genetic variants associated with ASD were enriched in glutamatergic pathways, affecting receptor signalling, metabolism and transport. Furthermore, in genetically modified mouse models for ASD, pharmacological compounds acting on ionotropic or metabotropic receptor activity are able to rescue ASD reminscent phenotypes. We conclude that glutamatergic genetic risk factors for ASD show a complex pattern and further studies are needed to fully understand its mechanisms, before translation of findings into clinical applications and individualized treatment approaches will be possible.
Collapse
|
46
|
Wu YY, Kumar R, Haque MS, Castillejo-López C, Alarcón-Riquelme ME. BANK1 controls CpG-induced IL-6 secretion via a p38 and MNK1/2/eIF4E translation initiation pathway. THE JOURNAL OF IMMUNOLOGY 2013; 191:6110-6. [PMID: 24227780 DOI: 10.4049/jimmunol.1301203] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BANK1, an adaptor protein expressed in B cells, plays a little understood role in B cell signaling. Because BANK1 contains an N-terminal putative Toll/IL-1R receptor domain, we used mouse Bank1(-/-) splenic B cells to test whether BANK1 affects signaling induced by the TLR9 agonist CpG. Following CpG stimulation, BANK1 deficiency reduced p38 phosphorylation without affecting that of ERK or JNK and reduced IL-6 secretion. Bank1(-/-) B cells showed reduced phosphorylation of MNK1/2 and eIF4E, suggesting an effect on translation initiation, whereas Bank1(-/-) had no effect on IL-6 mRNA stability, thus suggesting that BANK1 has no effect on MK2 signaling. IL-6 secretion observed when CpG stimulation was combined with anti-CD40 was reduced in the absence of BANK1. Whereas in the presence of anti-CD40 stimulation CpG induced a stronger phosphorylation of AKT, mTOR, and 4E-BP1, Bank1(-/-) had no effect on phosphorylation of mTOR and 4E-BP1, and a weak effect on AKT, implying that BANK1 does not affect the release of eIF4E by phospho-4E-BP1. Taken together, these data establish a previously unrecognized role for BANK1 in CpG-induced responses by splenic B cells on p38 signaling and control of translation initiation of IL-6 via MNK1/2 and eIF4E.
Collapse
Affiliation(s)
- Ying-Yu Wu
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | | | | | | | | |
Collapse
|
47
|
Soung YH, Korneeva N, Kim TH, Chung J. The role of c-Src in integrin (α6β4) dependent translational control. BMC Cell Biol 2013; 14:49. [PMID: 24180592 PMCID: PMC4228388 DOI: 10.1186/1471-2121-14-49] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 10/25/2013] [Indexed: 11/10/2022] Open
Abstract
Background Integrin α6β4 contributes to cancer progression by stimulating transcription as well as translation of cancer related genes. Our previous study demonstrated that α6β4 stimulates translation initiation of survival factors such as VEGF by activating mTOR pathway. However, the immediate early signaling events that link α6β4 to mTOR activation needs to be defined. Results In the current studies, we demonstrated that c-Src is an immediate early signaling molecule that acts upstream of α6β4 dependent mTOR activation and subsequent translation of VEGF in MDA-MB-435/β4 and MDA-MB-231 cancer cells. m7GTP-Sepharose–binding assay revealed that Src activity is required to form eIF4F complex which is necessary for Cap-dependent translation in α6β4 expressing human cancer cells. Conclusions Overall, our studies suggest that integrin β4 and c-Src activation is important early signaling events to lead mTOR activation and cap-dependent translation of VEGF.
Collapse
Affiliation(s)
| | | | | | - Jun Chung
- Department of Physiology and Stephenson Cancer Center, The University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA.
| |
Collapse
|
48
|
Chakravarthy R, Clemens MJ, Pirianov G, Perdios N, Mudan S, Cartwright JE, Elia A. Role of the eIF4E binding protein 4E-BP1 in regulation of the sensitivity of human pancreatic cancer cells to TRAIL and celastrol-induced apoptosis. Biol Cell 2013; 105:414-29. [PMID: 23734772 DOI: 10.1111/boc.201300021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 05/29/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND INFORMATION Tumour cells can be induced to undergo apoptosis after treatment with the tumour necrosis factor α-related death-inducing ligand (TRAIL). Although human pancreatic cancer cells show varying degrees of response they can be sensitised to the pro-apoptotic effects of TRAIL in the presence of celastrol, a natural compound extracted from the plant Tripterygium wilfordii Hook F. One important aspect of the cellular response to TRAIL is the control of protein synthesis, a key regulator of which is the eukaryotic initiation factor 4E-binding protein, 4E-BP1. RESULTS We examined the effects of celastrol and TRAIL in several pancreatic cancer cell lines. In cells that are normally resistant to TRAIL, synergistic effects of TRAIL plus celastrol on commitment to apoptosis and inhibition of protein synthesis were observed. These were associated with a strong up-regulation and dephosphorylation of 4E-BP1. The enhancement of 4E-BP1 expression, which correlated with a threefold increase in the level of the 4E-BP1 transcript, was blocked by inhibitors of reactive oxygen species and the JNK protein kinase. When the expression of 4E-BP1 was reduced by an inducible micro-RNA, TRAIL-mediated apoptosis was inhibited. CONCLUSION These results suggest that 4E-BP1 plays a critical role in the mechanism by which TRAIL and celastrol together cause apoptotic cell death in human pancreatic tumour cells.
Collapse
Affiliation(s)
- Reka Chakravarthy
- Translational Control Group, Division of Biomedical Sciences, St George's, University of London, London, SW17 0RE, UK
| | | | | | | | | | | | | |
Collapse
|
49
|
Ochs MJ, Ossipova E, Oliynyk G, Steinhilber D, Suess B, Jakobsson PJ. Mass Spectrometry-Based Proteomics Identifies UPF1 as a Critical Gene Expression Regulator in MonoMac 6 Cells. J Proteome Res 2013; 12:2622-9. [DOI: 10.1021/pr301193f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Meike J. Ochs
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438
Frankfurt/M., Germany
- Institute
of Molecular Biosciences, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt/M., Germany
| | - Elena Ossipova
- Department of Medicine, Rheumatology Unit, Karolinska Institute, Stockholm, Sweden
| | - Ganna Oliynyk
- Department of Medicine, Rheumatology Unit, Karolinska Institute, Stockholm, Sweden
- Department of Microbiology, Tumor
and Cell Biology, Karolinska Institute,
Stockholm, Sweden
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry/ZAFES, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438
Frankfurt/M., Germany
| | - Beatrix Suess
- Institute
of Molecular Biosciences, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt/M., Germany
- Department of
Biology, Technical University Darmstadt, Schnittspahnstraße 1064287 Darmstadt, Germany
| | - Per-Johan Jakobsson
- Department of Medicine, Rheumatology Unit, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
50
|
Nicaise V, Joe A, Jeong BR, Korneli C, Boutrot F, Westedt I, Staiger D, Alfano JR, Zipfel C. Pseudomonas HopU1 modulates plant immune receptor levels by blocking the interaction of their mRNAs with GRP7. EMBO J 2013; 32:701-12. [PMID: 23395902 DOI: 10.1038/emboj.2013.15] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 01/09/2013] [Indexed: 02/01/2023] Open
Abstract
Pathogens target important components of host immunity to cause disease. The Pseudomonas syringae type III-secreted effector HopU1 is a mono-ADP-ribosyltransferase required for full virulence on Arabidopsis thaliana. HopU1 targets several RNA-binding proteins including GRP7, whose role in immunity is still unclear. Here, we show that GRP7 associates with translational components, as well as with the pattern recognition receptors FLS2 and EFR. Moreover, GRP7 binds specifically FLS2 and EFR transcripts in vivo through its RNA recognition motif. HopU1 does not affect the protein-protein associations between GRP7, FLS2 and translational components. Instead, HopU1 blocks the interaction between GRP7 and FLS2 and EFR transcripts in vivo. This inhibition correlates with reduced FLS2 protein levels upon Pseudomonas infection in a HopU1-dependent manner. Our results reveal a novel virulence strategy used by a microbial effector to interfere with host immunity.
Collapse
Affiliation(s)
- Valerie Nicaise
- The Sainsbury Laboratory, Norwich Research Park, Norwich, UK
| | | | | | | | | | | | | | | | | |
Collapse
|