1
|
Fernandez A, Monsen PJ, Platanias LC, Schiltz GE. Medicinal chemistry approaches to target the MNK-eIF4E axis in cancer. RSC Med Chem 2023; 14:1060-1087. [PMID: 37360400 PMCID: PMC10285747 DOI: 10.1039/d3md00121k] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/08/2023] [Indexed: 06/28/2023] Open
Abstract
Aberrant translation of proteins that promote cell proliferation is an essential factor that defines oncogenic processes and cancer. The process for ribosomal translation of proteins from mRNA requires an essential initiation step which is controlled by the protein eIF4E, which binds the RNA 5'-cap and forms the eIF4F complex that subsequently translates protein. Typically, eIF4E is activated by phosphorylation on Ser209 by MNK1 and MNK2 kinases. Substantial work has shown that eIF4E and MNK1/2 are dysregulated in many cancers and this axis has therefore become an active area of interest for developing new cancer therapeutics. This review summarizes and discusses recent work to develop small molecules that target different steps in the MNK-eIF4E axis as potential cancer therapeutics. The aim of this review is to cover the breadth of different molecular approaches being taken and the medicinal chemistry basis for their optimization and testing as new cancer therapeutics.
Collapse
Affiliation(s)
- Ann Fernandez
- Department of Chemistry, Northwestern University Evanston IL 60208 USA
| | - Paige J Monsen
- Department of Chemistry, Northwestern University Evanston IL 60208 USA
| | - Leonidas C Platanias
- Robert H. Lurie Comprehensive Cancer Center Chicago IL 60611 USA
- Division of Hematology-Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University Chicago IL 60611 USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center Chicago IL 60612 USA
| | - Gary E Schiltz
- Department of Chemistry, Northwestern University Evanston IL 60208 USA
- Robert H. Lurie Comprehensive Cancer Center Chicago IL 60611 USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine Chicago IL 60611 USA
| |
Collapse
|
2
|
He Q, Yang C, Xiang Z, Huang G, Wu H, Chen T, Dou R, Song J, Han L, Song T, Wang S, Xiong B. LINC00924-induced fatty acid metabolic reprogramming facilitates gastric cancer peritoneal metastasis via hnRNPC-regulated alternative splicing of Mnk2. Cell Death Dis 2022; 13:987. [PMID: 36418856 PMCID: PMC9684446 DOI: 10.1038/s41419-022-05436-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/25/2022]
Abstract
The molecular mechanism underlying gastric cancer (GC) peritoneal metastasis (PM) remains unclear. Here, we identified LINC00924 as a GC PM-related lncRNA through Microarray sequencing. LINC00924 was highly expressed in GC, and its high expression is associated with a broad range of PM. Via RNA sequencing, RNA pulldown assay, mass spectrometry, Seahorse, Lipidomics, spheroid formation and cell viability assays, we found that LINC00924 promoted fatty acid (FA) oxidation (FAO) and FA uptake, which was essential for matrix-detached GC cell survival and spheroid formation. Regarding the mechanism, LINC00924 regulated the alternative splicing (AS) of Mnk2 pre-mRNA by binding to hnRNPC. Specifically, LINC00924 enhanced the binding of hnRNPC to Mnk2 pre-mRNA at e14a, thus downregulating Mnk2a splicing and regulating the p38 MAPK/PPARα signaling pathway. Collectively, our results demonstrate that LINC00924 plays a role in promoting GC PM and could serve as a drug target.
Collapse
Affiliation(s)
- Qiuming He
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Chaogang Yang
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Zhenxian Xiang
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Guoquan Huang
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Haitao Wu
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Tingna Chen
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Rongzhang Dou
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Jialing Song
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Lei Han
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - TianTian Song
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Shuyi Wang
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| | - Bin Xiong
- grid.413247.70000 0004 1808 0969Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,grid.413247.70000 0004 1808 0969Department of Gastric and Colorectal Surgical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071 China ,Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071 China ,Hubei Cancer Clinical Study Center, Wuhan, 430071 China
| |
Collapse
|
3
|
Adamson AL, Jeffus D, Davis A, Greengrove E. Epstein-Barr virus lytic replication activates and is dependent upon MAPK-interacting kinase 1/2 in a cell-type dependent manner. Virology 2022; 572:72-85. [DOI: 10.1016/j.virol.2022.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/17/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022]
|
4
|
MNK2 deficiency potentiates β-cell regeneration via translational regulation. Nat Chem Biol 2022; 18:942-953. [PMID: 35697798 PMCID: PMC7613404 DOI: 10.1038/s41589-022-01047-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 04/26/2022] [Indexed: 02/08/2023]
Abstract
Regenerating pancreatic β-cells is a potential curative approach for diabetes. We previously identified the small molecule CID661578 as a potent inducer of β-cell regeneration, but its target and mechanism of action have remained unknown. We now screened 257 million yeast clones and determined that CID661578 targets MAP kinase-interacting serine/threonine kinase 2 (MNK2), an interaction we genetically validated in vivo. CID661578 increased β-cell neogenesis from ductal cells in zebrafish, neonatal pig islet aggregates and human pancreatic ductal organoids. Mechanistically, we found that CID661578 boosts protein synthesis and regeneration by blocking MNK2 from binding eIF4G in the translation initiation complex at the mRNA cap. Unexpectedly, this blocking activity augmented eIF4E phosphorylation depending on MNK1 and bolstered the interaction between eIF4E and eIF4G, which is necessary for both hypertranslation and β-cell regeneration. Taken together, our findings demonstrate a targetable role of MNK2-controlled translation in β-cell regeneration, a role that warrants further investigation in diabetes.
Collapse
|
5
|
Xie J, Kusnadi EP, Furic L, Selth LA. Regulation of mRNA Translation by Hormone Receptors in Breast and Prostate Cancer. Cancers (Basel) 2021; 13:3254. [PMID: 34209750 PMCID: PMC8268847 DOI: 10.3390/cancers13133254] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Breast and prostate cancer are the second and third leading causes of death amongst all cancer types, respectively. Pathogenesis of these malignancies is characterised by dysregulation of sex hormone signalling pathways, mediated by the estrogen receptor-α (ER) in breast cancer and androgen receptor (AR) in prostate cancer. ER and AR are transcription factors whose aberrant function drives oncogenic transcriptional programs to promote cancer growth and progression. While ER/AR are known to stimulate cell growth and survival by modulating gene transcription, emerging findings indicate that their effects in neoplasia are also mediated by dysregulation of protein synthesis (i.e., mRNA translation). This suggests that ER/AR can coordinately perturb both transcriptional and translational programs, resulting in the establishment of proteomes that promote malignancy. In this review, we will discuss relatively understudied aspects of ER and AR activity in regulating protein synthesis as well as the potential of targeting mRNA translation in breast and prostate cancer.
Collapse
Affiliation(s)
- Jianling Xie
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Eric P Kusnadi
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luc Furic
- Translational Prostate Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Cancer Program, Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Luke A Selth
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
- Freemasons Centre for Male Health and Wellbeing, Flinders University, Bedford Park, SA 5042, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
6
|
Pacheco JM, Canal MV, Pereyra CM, Welchen E, Martínez-Noël GMA, Estevez JM. The tip of the iceberg: emerging roles of TORC1, and its regulatory functions in plant cells. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:4085-4101. [PMID: 33462577 DOI: 10.1093/jxb/eraa603] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
Target of Rapamycin (TOR) is an evolutionarily conserved protein kinase that plays a central role in coordinating cell growth with light availability, the diurnal cycle, energy availability, and hormonal pathways. TOR Complex 1 (TORC1) controls cell proliferation, growth, metabolism, and defense in plants. Sugar availability is the main signal for activation of TOR in plants, as it also is in mammals and yeast. Specific regulators of the TOR kinase pathway in plants are inorganic compounds in the form of major nutrients in the soils, and light inputs via their impact on autotrophic metabolism. The lack of TOR is embryo-lethal in plants, whilst dysregulation of TOR signaling causes major alterations in growth and development. TOR exerts control as a regulator of protein translation via the action of proteins such as S6K, RPS6, and TAP46. Phytohormones are central players in the downstream systemic physiological TOR effects. TOR has recently been attributed to have roles in the control of DNA methylation, in the abundance of mRNA splicing variants, and in the variety of regulatory lncRNAs and miRNAs. In this review, we summarize recent discoveries in the plant TOR signaling pathway in the context of our current knowledge of mammalian and yeast cells, and highlight the most important gaps in our understanding of plants that need to be addressed in the future.
Collapse
Affiliation(s)
| | - María Victoria Canal
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas,, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Cintia M Pereyra
- Instituto de Investigaciones en Biodiversidad y Biotecnología (INBIOTEC-CONICET) and Fundación para Investigaciones Biológicas Aplicadas (FIBA), Vieytes, Mar Del Plata, Argentina
| | - Elina Welchen
- Instituto de Agrobiotecnología del Litoral (CONICET-UNL), Cátedra de Biología Celular y Molecular, Facultad de Bioquímica y Ciencias Biológicas,, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Giselle M A Martínez-Noël
- Instituto de Investigaciones en Biodiversidad y Biotecnología (INBIOTEC-CONICET) and Fundación para Investigaciones Biológicas Aplicadas (FIBA), Vieytes, Mar Del Plata, Argentina
| | - José M Estevez
- Fundación Instituto Leloir and IIBBA-CONICET, Buenos Aires CP, Argentina
- Centro de Biotecnología Vegetal (CBV), Facultad de Ciencias de la Vida (FCsV), Universidad Andres Bello, Santiago, Chile and Millennium Institute for Integrative Biology (iBio), Santiago, Chile
| |
Collapse
|
7
|
Inhibitory effects of Tomivosertib in acute myeloid leukemia. Oncotarget 2021; 12:955-966. [PMID: 34012509 PMCID: PMC8121614 DOI: 10.18632/oncotarget.27952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/19/2021] [Indexed: 12/26/2022] Open
Abstract
The MAPK-interacting kinases 1 and 2 (MNK1/2) have generated increasing interest as therapeutic targets for acute myeloid leukemia (AML). We evaluated the therapeutic potential of the highly-selective MNK1/2 inhibitor Tomivosertib on AML cells. Tomivosertib was highly effective at blocking eIF4E phosphorylation on serine 209 in AML cells. Such inhibitory effects correlated with dose-dependent suppression of cellular viability and leukemic progenitor colony formation. Moreover, combination of Tomivosertib and Venetoclax resulted in synergistic anti-leukemic responses in AML cell lines. Mass spectrometry studies identified novel putative MNK1/2 interactors, while in parallel studies we demonstrated that MNK2 - RAPTOR - mTOR complexes are not disrupted by Tomivosertib. Overall, these findings demonstrate that Tomivosertib exhibits potent anti-leukemic properties on AML cells and support the development of clinical translational efforts involving the use of this drug, alone or in combination with other therapies for the treatment of AML.
Collapse
|
8
|
Majeed ST, Batool A, Majeed R, Bhat NN, Zargar MA, Andrabi KI. mTORC1 induces eukaryotic translation initiation factor 4E interaction with TOS-S6 kinase 1 and its activation. Cell Cycle 2021; 20:839-854. [PMID: 33938392 DOI: 10.1080/15384101.2021.1901038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Eukaryotic translation initiation factor 4E was recently shown to be a substrate of mTORC1, suggesting it may be a mediator of mTORC1 signaling. Here, we present evidence that eIF4E phosphorylated at S209 interacts with TOS motif of S6 Kinase1 (S6K1). We also show that this interaction is sufficient to overcome rapamycin sensitivity and mTORC1 dependence of S6K1. Furthermore, we show that eIF4E-TOS interaction relieves S6K1 from auto-inhibition due to carboxy terminal domain (CTD) and primes it for hydrophobic motif (HM) phosphorylation and activation in mTORC1 independent manner. We conclude that the role of mTORC1 is restricted to engaging eIF4E with S6K1-TOS motif to influence its state of HM phosphorylation and inducing its activation.
Collapse
Affiliation(s)
- Sheikh Tahir Majeed
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India.,Department of Biotechnology, Central University of Kashmir, Ganderbal, India
| | - Asiya Batool
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India.,Division of Cancer Pharmacology, Indian Institute of Integrative Medicine, Srinagar, India
| | - Rabiya Majeed
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India.,Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Nadiem Nazir Bhat
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India
| | | | - Khurshid Iqbal Andrabi
- Growth Factor Signaling Laboratory, Department of Biotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
9
|
Liu H, Gong Z, Li K, Zhang Q, Xu Z, Xu Y. SRPK1/2 and PP1α exert opposite functions by modulating SRSF1-guided MKNK2 alternative splicing in colon adenocarcinoma. J Exp Clin Cancer Res 2021; 40:75. [PMID: 33602301 PMCID: PMC7893936 DOI: 10.1186/s13046-021-01877-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/11/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The Mnk2 kinase, encoded by MKNK2 gene, plays critical roles in MAPK signaling and was involved in oncogenesis. Human MKNK2 pre-mRNA can be alternatively spliced into two splicing isoforms, the MKNK2a and MKNK2b, thus yielding Mnk2a and Mnk2b proteins with different domains. The involvement of Mnk2 alternative splicing in colon cancer has been implicated based on RNA-sequencing data from TCGA database. This study aimed at investigating the upstream modulators and clinical relevance of Mnk2 alternative splicing in colon adenocarcinoma (CAC). METHODS PCR, western blotting and immunohistochemistry (IHC) were performed to assess the expression of Mnk2 and upstream proteins in CAC. The function of Mnk2 and its regulators were demonstrated in different CAC cell lines as well as in xenograft models. Two independent cohorts of CAC patients were used to reveal the clinical significance of MKNK2 alternative splicing. RESULTS Comparing with adjacent nontumorous tissue, CAC specimen showed a decreased MKNK2a level and an increased MKNK2b level, which were correlated with KRAS mutation and tumor size. The SRSF1 (serine/arginine-rich splicing factor 1) was further confirmed to be the major splicing factor targeting MKNK2 in CAC cells. Higher expression of SRPK1/2 or decreased activity of PP1α were responsible for enhancing SRSF1 phosphorylation and nucleus translocation, subsequently resulted in a switch of MKNK2 alternative splicing. CONCLUSIONS Our data showed that phosphorylation and subcellular localization of SRSF1 were balanced by SRPK1/2 and PP1α in CAC cells. High nucleus SRSF1 promoted MKNK2 splicing into MKNK2b instead of MNK2a, consequently enhanced tumor proliferation.
Collapse
Affiliation(s)
- Hongda Liu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Zheng Gong
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Kangshuai Li
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Qun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China.
| | - Yunfei Xu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
10
|
Yousuf MS, Shiers SI, Sahn JJ, Price TJ. Pharmacological Manipulation of Translation as a Therapeutic Target for Chronic Pain. Pharmacol Rev 2021; 73:59-88. [PMID: 33203717 PMCID: PMC7736833 DOI: 10.1124/pharmrev.120.000030] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dysfunction in regulation of mRNA translation is an increasingly recognized characteristic of many diseases and disorders, including cancer, diabetes, autoimmunity, neurodegeneration, and chronic pain. Approximately 50 million adults in the United States experience chronic pain. This economic burden is greater than annual costs associated with heart disease, cancer, and diabetes combined. Treatment options for chronic pain are inadequately efficacious and riddled with adverse side effects. There is thus an urgent unmet need for novel approaches to treating chronic pain. Sensitization of neurons along the nociceptive pathway causes chronic pain states driving symptoms that include spontaneous pain and mechanical and thermal hypersensitivity. More than a decade of preclinical research demonstrates that translational mechanisms regulate the changes in gene expression that are required for ongoing sensitization of nociceptive sensory neurons. This review will describe how key translation regulation signaling pathways, including the integrated stress response, mammalian target of rapamycin, AMP-activated protein kinase (AMPK), and mitogen-activated protein kinase-interacting kinases, impact the translation of different subsets of mRNAs. We then place these mechanisms of translation regulation in the context of chronic pain states, evaluate currently available therapies, and examine the potential for developing novel drugs. Considering the large body of evidence now published in this area, we propose that pharmacologically manipulating specific aspects of the translational machinery may reverse key neuronal phenotypic changes causing different chronic pain conditions. Therapeutics targeting these pathways could eventually be first-line drugs used to treat chronic pain disorders. SIGNIFICANCE STATEMENT: Translational mechanisms regulating protein synthesis underlie phenotypic changes in the sensory nervous system that drive chronic pain states. This review highlights regulatory mechanisms that control translation initiation and how to exploit them in treating persistent pain conditions. We explore the role of mammalian/mechanistic target of rapamycin and mitogen-activated protein kinase-interacting kinase inhibitors and AMPK activators in alleviating pain hypersensitivity. Modulation of eukaryotic initiation factor 2α phosphorylation is also discussed as a potential therapy. Targeting specific translation regulation mechanisms may reverse changes in neuronal hyperexcitability associated with painful conditions.
Collapse
Affiliation(s)
- Muhammad Saad Yousuf
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - Stephanie I Shiers
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - James J Sahn
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| | - Theodore J Price
- Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas (M.S.Y., S.I.S., T.J.P.) and 4E Therapeutics Inc, Austin, Texas (J.J.S.)
| |
Collapse
|
11
|
Xie J, Shen K, Jones AT, Yang J, Tee AR, Shen MH, Yu M, Irani S, Wong D, Merrett JE, Lenchine RV, De Poi S, Jensen KB, Trim PJ, Snel MF, Kamei M, Martin SK, Fitter S, Tian S, Wang X, Butler LM, Zannettino ACW, Proud CG. Reciprocal signaling between mTORC1 and MNK2 controls cell growth and oncogenesis. Cell Mol Life Sci 2021; 78:249-270. [PMID: 32170339 PMCID: PMC11068017 DOI: 10.1007/s00018-020-03491-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/23/2020] [Accepted: 02/17/2020] [Indexed: 12/21/2022]
Abstract
eIF4E plays key roles in protein synthesis and tumorigenesis. It is phosphorylated by the kinases MNK1 and MNK2. Binding of MNKs to eIF4G enhances their ability to phosphorylate eIF4E. Here, we show that mTORC1, a key regulator of mRNA translation and oncogenesis, directly phosphorylates MNK2 on Ser74. This suppresses MNK2 activity and impairs binding of MNK2 to eIF4G. These effects provide a novel mechanism by which mTORC1 signaling impairs the function of MNK2 and thereby decreases eIF4E phosphorylation. MNK2[S74A] knock-in cells show enhanced phosphorylation of eIF4E and S6K1 (i.e., increased mTORC1 signaling), enlarged cell size, and increased invasive and transformative capacities. MNK2[Ser74] phosphorylation was inversely correlated with disease progression in human prostate tumors. MNK inhibition exerted anti-proliferative effects in prostate cancer cells in vitro. These findings define a novel feedback loop whereby mTORC1 represses MNK2 activity and oncogenic signaling through eIF4E phosphorylation, allowing reciprocal regulation of these two oncogenic pathways.
Collapse
Affiliation(s)
- Jianling Xie
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
| | - Kaikai Shen
- Medical Research Council Toxicology Unit, Leicester, UK
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ashley T Jones
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, UK
| | - Jian Yang
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, UK
| | - Andrew R Tee
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, UK
| | - Ming Hong Shen
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, UK
| | - Mengyuan Yu
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Swati Irani
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Derick Wong
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
| | - James E Merrett
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
- Department of Molecular and Cellular Biology, University of Adelaide, Adelaide, Australia
| | - Roman V Lenchine
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
- Department of Molecular and Cellular Biology, University of Adelaide, Adelaide, Australia
| | - Stuart De Poi
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
- Department of Molecular and Cellular Biology, University of Adelaide, Adelaide, Australia
| | - Kirk B Jensen
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
- Department of Molecular and Cellular Biology, University of Adelaide, Adelaide, Australia
| | - Paul J Trim
- Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Marten F Snel
- Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Makoto Kamei
- Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Sally Kim Martin
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Science, University of Adelaide, Adelaide, Australia
| | - Stephen Fitter
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Science, University of Adelaide, Adelaide, Australia
| | - Shuye Tian
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Xuemin Wang
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
- Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Lisa M Butler
- Adelaide Medical School and Freemasons Foundation Centre for Men's Health, University of Adelaide, Adelaide, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Andrew C W Zannettino
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Science, University of Adelaide, Adelaide, Australia
| | - Christopher G Proud
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia.
- Hopwood Centre for Neurobiology, South Australian Health and Medical Research Institute, Adelaide, Australia.
| |
Collapse
|
12
|
Ackerman EE, Shoemaker JE. Network Controllability-Based Prioritization of Candidates for SARS-CoV-2 Drug Repositioning. Viruses 2020; 12:v12101087. [PMID: 32993136 PMCID: PMC7650805 DOI: 10.3390/v12101087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/12/2022] Open
Abstract
In a short time, the COVID-19 pandemic has left the world with over 25 million cases and staggering death tolls that are still rising. Treatments for SARS-CoV-2 infection are desperately needed as there are currently no approved drug therapies. With limited knowledge of viral mechanisms, a network controllability method of prioritizing existing drugs for repurposing efforts is optimal for quickly moving through the drug approval pipeline using limited, available, virus-specific data. Based on network topology and controllability, 16 proteins involved in translation, cellular transport, cellular stress, and host immune response are predicted as regulators of the SARS-CoV-2 infected cell. Of the 16, eight are prioritized as possible drug targets where two, PVR and SCARB1, are previously unexplored. Known compounds targeting these genes are suggested for viral inhibition study. Prioritized proteins in agreement with previous analysis and viral inhibition studies verify the ability of network controllability to predict biologically relevant candidates.
Collapse
Affiliation(s)
- Emily E. Ackerman
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Jason E. Shoemaker
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA 15260, USA;
- The McGowan Institute for Regenerative Medicine (MIRM), University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Correspondence:
| |
Collapse
|
13
|
Pinto-Díez C, Ferreras-Martín R, Carrión-Marchante R, González VM, Martín ME. Deeping in the Role of the MAP-Kinases Interacting Kinases (MNKs) in Cancer. Int J Mol Sci 2020; 21:2967. [PMID: 32340135 PMCID: PMC7215568 DOI: 10.3390/ijms21082967] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 02/05/2023] Open
Abstract
The mitogen-activated protein kinase (MAPK)-interacting kinases (MNKs) are involved in oncogenic transformation and can promote metastasis and tumor progression. In human cells, there are four MNKs isoforms (MNK1a/b and MNK2a/b), derived from two genes by alternative splicing. These kinases play an important role controlling the expression of specific proteins involved in cell cycle, cell survival and cell motility via eukaryotic initiation factor 4E (eIF4E) regulation, but also through other substrates such as heterogeneous nuclear ribonucleoprotein A1, polypyrimidine tract-binding protein-associated splicing factor and Sprouty 2. In this review, we provide an overview of the role of MNK in human cancers, describing the studies conducted to date to elucidate the mechanism involved in the action of MNKs, as well as the development of MNK inhibitors in different hematological cancers and solid tumors.
Collapse
Affiliation(s)
| | | | | | | | - María Elena Martín
- Grupo de Aptámeros, Servicio de Bioquímica-Investigación, IRYCIS-Hospital Ramón y Cajal, Madrid, Ctra. Colmenar Km. 9100, 28034 Madrid, Spain; (C.P.-D.); (R.F.-M.); (R.C.-M.); (V.M.G.)
| |
Collapse
|
14
|
Martin P, Wagh V, Reis SA, Erdin S, Beauchamp RL, Shaikh G, Talkowski M, Thiele E, Sheridan SD, Haggarty SJ, Ramesh V. TSC patient-derived isogenic neural progenitor cells reveal altered early neurodevelopmental phenotypes and rapamycin-induced MNK-eIF4E signaling. Mol Autism 2020; 11:2. [PMID: 31921404 PMCID: PMC6945400 DOI: 10.1186/s13229-019-0311-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/29/2019] [Indexed: 12/21/2022] Open
Abstract
Background Tuberous sclerosis complex (TSC) is a neurodevelopmental disorder with frequent occurrence of epilepsy, autism spectrum disorder (ASD), intellectual disability (ID), and tumors in multiple organs. The aberrant activation of mTORC1 in TSC has led to treatment with mTORC1 inhibitor rapamycin as a lifelong therapy for tumors, but TSC-associated neurocognitive manifestations remain unaffected by rapamycin. Methods Here, we generated patient-specific, induced pluripotent stem cells (iPSCs) from a TSC patient with a heterozygous, germline, nonsense mutation in exon 15 of TSC1 and established an isogenic set of heterozygous (Het), null and corrected wildtype (Corr-WT) iPSCs using CRISPR/Cas9-mediated gene editing. We differentiated these iPSCs into neural progenitor cells (NPCs) and examined neurodevelopmental phenotypes, signaling and changes in gene expression by RNA-seq. Results Differentiated NPCs revealed enlarged cell size in TSC1-Het and Null NPCs, consistent with mTORC1 activation. TSC1-Het and Null NPCs also revealed enhanced proliferation and altered neurite outgrowth in a genotype-dependent manner, which was not reversed by rapamycin. Transcriptome analyses of TSC1-NPCs revealed differentially expressed genes that display a genotype-dependent linear response, i.e., genes upregulated/downregulated in Het were further increased/decreased in Null. In particular, genes linked to ASD, epilepsy, and ID were significantly upregulated or downregulated warranting further investigation. In TSC1-Het and Null NPCs, we also observed basal activation of ERK1/2, which was further activated upon rapamycin treatment. Rapamycin also increased MNK1/2-eIF4E signaling in TSC1-deficient NPCs. Conclusion MEK-ERK and MNK-eIF4E pathways regulate protein translation, and our results suggest that aberrant translation distinct in TSC1/2-deficient NPCs could play a role in neurodevelopmental defects. Our data showing upregulation of these signaling pathways by rapamycin support a strategy to combine a MEK or a MNK inhibitor with rapamycin that may be superior for TSC-associated CNS defects. Importantly, our generation of isogenic sets of NPCs from TSC patients provides a valuable platform for translatome and large-scale drug screening studies. Overall, our studies further support the notion that early developmental events such as NPC proliferation and initial process formation, such as neurite number and length that occur prior to neuronal differentiation, represent primary events in neurogenesis critical to disease pathogenesis of neurodevelopmental disorders such as ASD.
Collapse
Affiliation(s)
- Pauline Martin
- 1Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Vilas Wagh
- 2MERCK Research Laboratories, Boston, MA 02115 USA
| | - Surya A Reis
- 1Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Serkan Erdin
- 1Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Roberta L Beauchamp
- 1Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Ghalib Shaikh
- 1Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Michael Talkowski
- 1Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114 USA.,3Department of Neurology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA
| | - Elizabeth Thiele
- 3Department of Neurology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA
| | - Steven D Sheridan
- 1Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114 USA.,4Center for Quantitative Health, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Stephen J Haggarty
- 1Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114 USA.,3Department of Neurology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA
| | - Vijaya Ramesh
- 1Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114 USA.,3Department of Neurology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA
| |
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
|
Proud CG. Phosphorylation and Signal Transduction Pathways in Translational Control. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a033050. [PMID: 29959191 DOI: 10.1101/cshperspect.a033050] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein synthesis, including the translation of specific messenger RNAs (mRNAs), is regulated by extracellular stimuli such as hormones and by the levels of certain nutrients within cells. This control involves several well-understood signaling pathways and protein kinases, which regulate the phosphorylation of proteins that control the translational machinery. These pathways include the mechanistic target of rapamycin complex 1 (mTORC1), its downstream effectors, and the mitogen-activated protein (MAP) kinase (extracellular ligand-regulated kinase [ERK]) signaling pathway. This review describes the regulatory mechanisms that control translation initiation and elongation factors, in particular the effects of phosphorylation on their interactions or activities. It also discusses current knowledge concerning the impact of these control systems on the translation of specific mRNAs or subsets of mRNAs, both in physiological processes and in diseases such as cancer.
Collapse
Affiliation(s)
- Christopher G Proud
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, North Terrace, Adelaide SA5000, Australia; and School of Biological Sciences, University of Adelaide, Adelaide SA5000, Australia
| |
Collapse
|
17
|
Ichiyanagi O, Ito H, Naito S, Kabasawa T, Kanno H, Narisawa T, Ushijima M, Kurota Y, Ozawa M, Sakurai T, Nishida H, Kato T, Yamakawa M, Tsuchiya N. Impact of eIF4E phosphorylation at Ser209 via MNK2a on tumour recurrence after curative surgery in localized clear cell renal cell carcinoma. Oncotarget 2019; 10:4053-4068. [PMID: 31258849 PMCID: PMC6592294 DOI: 10.18632/oncotarget.27017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/20/2019] [Indexed: 01/01/2023] Open
Abstract
Background: We investigated the roles of eIF4E phosphorylation (Ser209) in tumour recurrence after curative nephrectomy for localized clear cell renal cell carcinoma (ccRCC). Methods: Expression of eIF4E, p eIF4E and MNKs (MAPK interacting kinases), was evaluated in surgical specimens obtained from consecutive non metastatic ccRCC patients (n = 290) by immunohistochemistry (IHC), immunoblotting, and qRT PCR at the protein and mRNA levels. In human RCC cell lines, the effects of eIF4E phosphorylation were examined using immunoblotting, proliferation, migration and invasion assays with pharmacological inhibitors (CGP57380 or ETP45835) and specific small interfering (si) RNAs against MNK1/2(a/b). Results: In postoperative follow-up (median, 7.9 y), 40 patients experienced metastatic recurrence. In multivariate Cox analyses, higher IHC expression of p eIF4E in ccRCC significantly predicted a longer recurrence-free interval. eIF4E is phosphorylated mainly by MNK2a in tumour specimens and cell lines. In 786-O and A-498 cell lines, pharmacological inhibition of MNKs decreased p-eIF4E and increased vimentin and N cadherin but did not influence proliferation. Similarly, MNK2 or MNK2a inhibition with siRNA reduced p-eIF4E and enhanced vimentin translation, cell migration and invasion in the cell lines. Conclusions: MNK2a-induced eIF4E phosphorylation may suppress metastatic recurrence of ccRCC, partially due to vimentin downregulation at the translational level, consequently leading to inhibition of epithelial–mesenchymal transition.
Collapse
Affiliation(s)
- Osamu Ichiyanagi
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan.,Department of Urology, Yamagata Prefectural Kahoku Hospital, Kahoku, Japan
| | - Hiromi Ito
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Sei Naito
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Takanobu Kabasawa
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Hidenori Kanno
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Takafumi Narisawa
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Masaki Ushijima
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yuta Kurota
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Michinobu Ozawa
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Toshihiko Sakurai
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Hayato Nishida
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Tomoyuki Kato
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Mitsunori Yamakawa
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Norihiko Tsuchiya
- Department of Urology, Yamagata University Faculty of Medicine, Yamagata, Japan
| |
Collapse
|
18
|
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
|
19
|
Src Family Kinase Inhibitors Block Translation of Alphavirus Subgenomic mRNAs. Antimicrob Agents Chemother 2019; 63:AAC.02325-18. [PMID: 30917980 PMCID: PMC6496153 DOI: 10.1128/aac.02325-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/14/2019] [Indexed: 01/01/2023] Open
Abstract
Alphaviruses are arthropod-transmitted RNA viruses that can cause arthralgia, myalgia, and encephalitis in humans. Since the role of cellular kinases in alphavirus replication is unknown, we profiled kinetic changes in host kinase abundance and phosphorylation following chikungunya virus (CHIKV) infection of fibroblasts. Alphaviruses are arthropod-transmitted RNA viruses that can cause arthralgia, myalgia, and encephalitis in humans. Since the role of cellular kinases in alphavirus replication is unknown, we profiled kinetic changes in host kinase abundance and phosphorylation following chikungunya virus (CHIKV) infection of fibroblasts. Based upon the results of this study, we treated CHIKV-infected cells with kinase inhibitors targeting the Src family kinase (SFK)–phosphatidylinositol 3-kinase (PI3K)–AKT–mTORC signaling pathways. Treatment of cells with SFK inhibitors blocked the replication of CHIKV as well as multiple other alphaviruses, including Mayaro virus, O’nyong-nyong virus, Ross River virus, and Venezuelan equine encephalitis virus. Dissecting the effect of SFK inhibition on alphavirus replication, we found that viral structural protein levels were significantly reduced, but synthesis of viral genomic and subgenomic RNAs was unaffected. By measuring the association of viral RNA with polyribosomes, we found that the SFK inhibitor dasatinib blocks alphavirus subgenomic RNA translation. Our results demonstrate a role for SFK signaling in alphavirus subgenomic RNA translation and replication. Targeting host factors involved in alphavirus replication represents an innovative, perhaps paradigm-shifting, strategy for exploring the replication of CHIKV and other alphaviruses while promoting antiviral therapeutic development.
Collapse
|
20
|
Batool A, Majeed ST, Aashaq S, Majeed R, Shah G, Nazir N, Andrabi KI. Eukaryotic Initiation Factor 4E (eIF4E) sequestration mediates 4E-BP1 response to rapamycin. Int J Biol Macromol 2019; 125:651-659. [DOI: 10.1016/j.ijbiomac.2018.12.102] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 10/27/2022]
|
21
|
Xie J, Merrett JE, Jensen KB, Proud CG. The MAP kinase-interacting kinases (MNKs) as targets in oncology. Expert Opin Ther Targets 2019; 23:187-199. [DOI: 10.1080/14728222.2019.1571043] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Jianling Xie
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - James E. Merrett
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Kirk B. Jensen
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, Adelaide, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Christopher G. Proud
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, Adelaide, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
22
|
Huang XB, Yang CM, Han QM, Ye XJ, Lei W, Qian WB. MNK1 inhibitor CGP57380 overcomes mTOR inhibitor-induced activation of eIF4E: the mechanism of synergic killing of human T-ALL cells. Acta Pharmacol Sin 2018; 39:1894-1901. [PMID: 30297804 DOI: 10.1038/s41401-018-0161-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/30/2018] [Indexed: 01/05/2023]
Abstract
Although the treatment of adult T-cell acute lymphoblastic leukemia (T-ALL) has been significantly improved, the heterogeneous genetic landscape of the disease often causes relapse. Aberrant activation of mammalian target of rapamycin (mTOR) pathway in T-ALL is responsible for treatment failure and relapse, suggesting that mTOR inhibition may represents a new therapeutic strategy. In this study, we investigated whether the mTOR complex 1 (mTORC1) inhibitor everolimus could be used as a therapeutic agent against human T-ALL. We showed that rapamycin and its analog RAD001 (everolimus) exerted only mild inhibition on the viability of Jurkat, CEM and Molt-4 cell lines (for everolimus the maximum inhibition was <40% at 100 nM), but greatly enhanced the phosphorylation of eIF4E, a downstream substrate of MAPK-interacting kinase (MNK) that was involved in promoting cell survival. Furthermore, we demonstrated in Jurkat cells that mTOR inhibitor-induced eIF4E phosphorylation was independent of insulin-like growth factor-1/insulin-like growth factor-1 receptor axis, but was secondary to mTOR inhibition. Then we examined the antileukemia effects of CGP57380, a MNK1 inhibitor, and we found that CGP57380 (4-16 μM) dose-dependently suppressed the expression of both phosphor-MNK1 and phosphor-eIF4E, thereby inhibiting downstream targets such as c-Myc and survivin in T-ALL cells. Importantly, CGP57380 produced a synergistic growth inhibitory effect with everolimus in T-ALL cells, and treatment with this targeted therapy overcame everolimus-induced eIF4E phosphorylation. In conclusion, our results suggest that dual-targeting of mTOR and MNK1/eIF4E signaling pathways may represent a novel therapeutic strategy for the treatment of human T-ALL.
Collapse
|
23
|
D'Abronzo LS, Ghosh PM. eIF4E Phosphorylation in Prostate Cancer. Neoplasia 2018; 20:563-573. [PMID: 29730477 PMCID: PMC5994774 DOI: 10.1016/j.neo.2018.04.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/09/2018] [Accepted: 04/10/2018] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PCa) progression involves a shift from endocrine to paracrine and eventually autocrine control resulting from alterations in molecular mechanisms in the cells. Deregulation of RNA translation is crucial for tumor cells to grow and proliferate; therefore, overactivation of the translation machinery is often observed in cancer. The two most important signal transduction pathways regulating PCa progression are PI3K/Akt/mTOR and Ras/MAPK. These two pathways converge on the eukaryotic translation initiation factor 4E (eIF4E) which binds to the protein scaffold eIF4G upon mechanistic target of rapamycin (mTOR) activation and is phosphorylated by the mitogen-activated protein kinase (MAPK) interacting protein kinases (Mnk1/2). This review describes the role of eIF4E in mRNA translation initiation mediated by its binding to the methylated 5′ terminal structure (m7G-cap) of many mRNAs, and the ability of many tumor cells to bypass this mechanism. Hormonal therapy and chemotherapy are two of the most prevalent therapies used in patients with advanced PCa, and studies have implicated a role for eIF4E phosphorylation in promoting resistance to both these therapies. It appears that eIF4E phosphorylation enhances the rate of translation of oncogene mRNAs to increase tumorigenicity.
Collapse
Affiliation(s)
- Leandro S D'Abronzo
- VA Northern California Health Care System, Mather, CA; Department of Urological Surgery, University of California at Davis, Sacramento, CA
| | - Paramita M Ghosh
- VA Northern California Health Care System, Mather, CA; Department of Urological Surgery, University of California at Davis, Sacramento, CA; Department of Biochemistry and Molecular Medicine, University of California at Davis, Sacramento, CA.
| |
Collapse
|
24
|
Lineham E, Tizzard GJ, Coles SJ, Spencer J, Morley SJ. Synergistic effects of inhibiting the MNK-eIF4E and PI3K/AKT/ mTOR pathways on cell migration in MDA-MB-231 cells. Oncotarget 2018; 9:14148-14159. [PMID: 29581834 PMCID: PMC5865660 DOI: 10.18632/oncotarget.24354] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/25/2018] [Indexed: 01/25/2023] Open
Abstract
The study of eukaryotic initiation factor 4E (eIF4E) is a key focus in cancer research due to its role in controlling the translation of tumour-associated proteins, that drive an aggressive migratory phenotype. eIF4E is a limiting component of the eIF4F complex which is a critical determinant for the translation of mRNAs. Mitogen-activated protein kinase interacting protein kinases (MNK1/2) phosphorylate eIF4E on Ser209, promoting the expression of oncogenic proteins, whereas mTORC1 phosphorylates and de-activates the eIF4E inhibitor, 4E-BP1, to release translational repression. Here we show that inhibiting these pathways simultaneously effectively slows the rate of cell migration in breast cancer cells. However, a molecular hybridisation approach using novel, cleavable dual MNK1/2 and PI3K/mTOR inhibiting hybrid agents was less effective at slowing cell migration.
Collapse
Affiliation(s)
- Ella Lineham
- Department of Biochemistry, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| | - Graham J Tizzard
- UK National Crystallography Service, School of Chemistry, University of Southampton, Highfield, Southampton, UK
| | - Simon J Coles
- UK National Crystallography Service, School of Chemistry, University of Southampton, Highfield, Southampton, UK
| | - John Spencer
- Department of Chemistry, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| | - Simon J Morley
- Department of Biochemistry, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
| |
Collapse
|
25
|
Dual abrogation of MNK and mTOR: a novel therapeutic approach for the treatment of aggressive cancers. Future Med Chem 2017; 9:1539-1555. [PMID: 28841037 DOI: 10.4155/fmc-2017-0062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Targeting the translational machinery has emerged as a promising therapeutic option for cancer treatment. Cancer cells require elevated protein synthesis and exhibit augmented activity to meet the increased metabolic demand. Eukaryotic translation initiation factor 4E is necessary for mRNA translation, its availability and phosphorylation are regulated by the PI3K/AKT/mTOR and MNK1/2 pathways. The phosphorylated form of eIF4E drives the expression of oncogenic proteins including those involved in metastasis. In this article, we will review the role of eIF4E in cancer, its regulation and discuss the benefit of dual inhibition of upstream pathways. The discernible interplay between the MNK and mTOR signaling pathways provides a novel therapeutic opportunity to target aggressive migratory cancers through the development of hybrid molecules.
Collapse
|
26
|
D'Abronzo LS, Bose S, Crapuchettes ME, Beggs RE, Vinall RL, Tepper CG, Siddiqui S, Mudryj M, Melgoza FU, Durbin-Johnson BP, deVere White RW, Ghosh PM. The androgen receptor is a negative regulator of eIF4E phosphorylation at S209: implications for the use of mTOR inhibitors in advanced prostate cancer. Oncogene 2017; 36:6359-6373. [PMID: 28745319 PMCID: PMC5690844 DOI: 10.1038/onc.2017.233] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 05/22/2017] [Accepted: 06/06/2017] [Indexed: 01/25/2023]
Abstract
The anti-androgen bicalutamide is widely used in the treatment of advanced prostate cancer (PCa) in many countries, but its effect on castration resistant PCa (CRPC) is limited. We previously showed that resistance to bicalutamide results from activation of mechanistic target of rapamycin (mTOR). Interestingly, clinical trials testing combinations of the mTOR inhibitor RAD001 with bicalutamide were effective in bicalutamide-naïve CRPC patients, but not in bicalutamide-pre-treated ones. Here we investigate causes for their difference in response. Evaluation of CRPC cell lines identified resistant vs sensitive in-vitro models, and revealed that increased eIF4E(S209) phosphorylation is associated with resistance to the combination. We confirmed using a human-derived tumor-xenograft mouse model that bicalutamide pre-treatment is associated with an increase in eIF4E(S209) phosphorylation. Thus, AR suppressed eIF4E phosphorylation, while the use of anti-androgens relieved this suppression, thereby triggering its increase. Additional investigation in human prostatectomy samples showed that increased eIF4E phosphorylation strongly correlated with the cell proliferation marker Ki67. SiRNA-mediated knock-down of eIF4E sensitized CRPC cells to RAD001+bicalutamide, while eIF4E overexpression induced resistance. Inhibition of eIF4E phosphorylation by treatment with CGP57380 (an inhibitor of MAPK interacting serine-threonine kinases Mnk1/2, the eIF4E upstream kinase) or inhibitors of ERK1/2, the upstream kinase regulating Mnk1/2, also sensitized CRPC cells to RAD001+bicalutamide. Examination of downstream targets of eIF4E-mediated translation, including survivin, demonstrated that eIF4E(S209) phosphorylation increased cap-independent translation whereas its inhibition restored cap-dependent translation which could be inhibited by mTOR inhibitors. Thus, our results demonstrate that while combinations of AR and mTOR inhibitors were effective in suppressing tumor growth by inhibiting both AR-induced transcription and mTOR-induced cap-dependent translation, pre-treatment with AR antagonists including bicalutamide increased eIF4E phosphorylation that induced resistance to combinations of AR and mTOR inhibitors by inducing cap-independent translation. We conclude that this resistance can be overcome by inhibiting eIF4E phosphorylation with Mnk1/2 or ERK1/2 inhibitors.
Collapse
Affiliation(s)
- L S D'Abronzo
- VA Northern California Health Care System, University of California at Davis, Sacramento, CA, USA.,Department of Urology, University of California Davis School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - S Bose
- Department of Urology, University of California Davis School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - M E Crapuchettes
- VA Northern California Health Care System, University of California at Davis, Sacramento, CA, USA
| | - R E Beggs
- VA Northern California Health Care System, University of California at Davis, Sacramento, CA, USA
| | - R L Vinall
- Department of Urology, University of California Davis School of Medicine, University of California at Davis, Sacramento, CA, USA.,California Northstate University College of Pharmacy, Elk Grove, CA, USA
| | - C G Tepper
- Department of Biochemistry and Molecular Medicine, University of California at Davis, Sacramento, CA, USA
| | - S Siddiqui
- VA Northern California Health Care System, University of California at Davis, Sacramento, CA, USA
| | - M Mudryj
- Department of Medical Microbiology and Immunology, University of California at Davis, Sacramento, CA, USA
| | - F U Melgoza
- VA Northern California Health Care System, University of California at Davis, Sacramento, CA, USA
| | - B P Durbin-Johnson
- Department of Public Health, Division of Biostatistics, University of California at Davis, Sacramento, CA, USA
| | - R W deVere White
- Department of Urology, University of California Davis School of Medicine, University of California at Davis, Sacramento, CA, USA
| | - P M Ghosh
- VA Northern California Health Care System, University of California at Davis, Sacramento, CA, USA.,Department of Urology, University of California Davis School of Medicine, University of California at Davis, Sacramento, CA, USA.,Department of Biochemistry and Molecular Medicine, University of California at Davis, Sacramento, CA, USA
| |
Collapse
|
27
|
Bramham CR, Jensen KB, Proud CG. Tuning Specific Translation in Cancer Metastasis and Synaptic Memory: Control at the MNK-eIF4E Axis. Trends Biochem Sci 2016; 41:847-858. [PMID: 27527252 DOI: 10.1016/j.tibs.2016.07.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 02/07/2023]
Abstract
The eukaryotic translation initiation factor (eIF) 4E, which binds to the 5'-cap of mRNA, undergoes phosphorylation on a single conserved serine, executed by the mitogen-activated protein kinase (MAPK)-interacting kinases (MNKs). However, the functional consequences and physiological roles of MNK signalling have remained obscure. Now, new pharmacological and genetic tools have provided unprecedented insights into the function of MNKs and eIF4E phosphorylation. The studies suggest that MNKs control the translation of specific mRNAs in cancer metastasis and neuronal synaptic plasticity by a novel mechanism involving the regulation of the translational repressor, cytoplasmic fragile-X protein-interacting protein 1 (CYFIP1). These recent breakthroughs go a long way to resolving the longstanding enigma and controversy surrounding the function of the MNK-eIF4E axis in cancer cell biology and neurobiology.
Collapse
Affiliation(s)
- Clive R Bramham
- Department of Biomedicine and KG Jebsen Centre for Neuropsychiatric Disorders, University of Bergen, 5009 Bergen, Norway.
| | - Kirk B Jensen
- South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Christopher G Proud
- South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
28
|
Hussein NA, Delaney TL, Tounsel BL, Liebl FLW. The Extracellular-Regulated Kinase Effector Lk6 is Required for Glutamate Receptor Localization at the Drosophila Neuromuscular Junction. J Exp Neurosci 2016; 10:77-91. [PMID: 27199570 PMCID: PMC4866800 DOI: 10.4137/jen.s32840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 11/16/2022] Open
Abstract
The proper localization and synthesis of postsynaptic glutamate receptors are essential for synaptic plasticity. Synaptic translation initiation is thought to occur via the target of rapamycin (TOR) and mitogen-activated protein kinase signal-integrating kinase (Mnk) signaling pathways, which is downstream of extracellular-regulated kinase (ERK). We used the model glutamatergic synapse, the Drosophila neuromuscular junction, to better understand the roles of the Mnk and TOR signaling pathways in synapse development. These synapses contain non-NMDA receptors that are most similar to AMPA receptors. Our data show that Lk6, the Drosophila homolog of Mnk1 and Mnk2, is required in either presynaptic neurons or postsynaptic muscle for the proper localization of the GluRIIA glutamate receptor subunit. Lk6 may signal through eukaryotic initiation factor (eIF) 4E to regulate the synaptic levels of GluRIIA as either interfering with eIF4E binding to eIF4G or expression of a nonphosphorylatable isoform of eIF4E resulted in a significant reduction in GluRIIA at the synapse. We also find that Lk6 and TOR may independently regulate synaptic levels of GluRIIA.
Collapse
Affiliation(s)
- Nizar A Hussein
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Taylor L Delaney
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Brittany L Tounsel
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Faith L W Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| |
Collapse
|
29
|
Gopaul K, Koorbanally NA. Sulfoximine substituted quinazolines for pharmaceutical compositions US 20150005278 (A1): a patent evaluation. Expert Opin Ther Pat 2016; 26:861-9. [DOI: 10.1080/13543776.2016.1180366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Kaalin Gopaul
- School of Chemistry, University of KwaZulu-Natal, Durban, South Africa
| | | |
Collapse
|
30
|
Abstract
The discovery of small molecules that selectively inhibit Mnks is considered of paramount importance towards deciphering the exact role of these proteins in carcinogenesis and to further validate them as anti-cancer drug targets. However, the dearth of structural information of Mnks is a major hurdle. This study unveils the 7H-pyrrolo[2,3-d]pyrimidine derivatives as potent inhibitors of Mnks. ATP and substrate competition assays showed that this scaffold interacts with the ATP binding site, but not with the substrate site. Screened against a panel of cancer cells, Mnk inhibitors were most potent against MV4-11 acute myeloid leukemia cells. The induction of apoptosis was shown to be mediated by downregulation of Mcl-1.
Collapse
|
31
|
Joubert PE, Stapleford K, Guivel-Benhassine F, Vignuzzi M, Schwartz O, Albert ML. Inhibition of mTORC1 Enhances the Translation of Chikungunya Proteins via the Activation of the MnK/eIF4E Pathway. PLoS Pathog 2015; 11:e1005091. [PMID: 26317997 PMCID: PMC4552638 DOI: 10.1371/journal.ppat.1005091] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 07/16/2015] [Indexed: 01/20/2023] Open
Abstract
Chikungunya virus (CHIKV), the causative agent of a major epidemic spanning five continents, is a positive stranded mRNA virus that replicates using the cell's cap-dependent translation machinery. Despite viral infection inhibiting mTOR, a metabolic sensor controls cap-dependent translation, viral proteins are efficiently translated. Rapalog treatment, silencing of mtor or raptor genes, but not rictor, further enhanced CHIKV infection in culture cells. Using biochemical assays and real time imaging, we demonstrate that this effect is independent of autophagy or type I interferon production. Providing in vivo evidence for the relevance of our findings, mice treated with mTORC1 inhibitors exhibited increased lethality and showed a higher sensitivity to CHIKV. A systematic evaluation of the viral life cycle indicated that inhibition of mTORC1 has a specific positive effect on viral proteins, enhancing viral replication by increasing the translation of both structural and nonstructural proteins. Molecular analysis defined a role for phosphatidylinositol-3 kinase (PI3K) and MAP kinase-activated protein kinase (MnKs) activation, leading to the hyper-phosphorylation of eIF4E. Finally, we demonstrated that in the context of CHIKV inhibition of mTORC1, viral replication is prioritized over host translation via a similar mechanism. Our study reveals an unexpected bypass pathway by which CHIKV protein translation overcomes viral induced mTORC1 inhibition.
Collapse
Affiliation(s)
- Pierre-Emmanuel Joubert
- Unité Immunobiologie des Cellules Dendritiques, Département d’Immunologie, Institut Pasteur, Paris, Cedex 15, France
- INSERM U818, Paris, France
| | - Kenneth Stapleford
- Unité des populations virales et Pathogenèse, Département de Virologie, Institut Pasteur, Paris, Cedex 15, France
| | | | - Marco Vignuzzi
- Unité des populations virales et Pathogenèse, Département de Virologie, Institut Pasteur, Paris, Cedex 15, France
| | - Olivier Schwartz
- Unité Virus et Immunité, Département de Virologie Institut Pasteur, Paris, Cedex 15, France
| | - Matthew L. Albert
- Unité Immunobiologie des Cellules Dendritiques, Département d’Immunologie, Institut Pasteur, Paris, Cedex 15, France
- INSERM U818, Paris, France
- Centre d’Immunologie Humaine, Département d’Immunologie, Institut Pasteur, Paris, Cedex 15, France
| |
Collapse
|
32
|
Wu H, Hu C, Wang A, Weisberg EL, Chen Y, Yun CH, Wang W, Liu Y, Liu X, Tian B, Wang J, Zhao Z, Liang Y, Li B, Wang L, Wang B, Chen C, Buhrlage SJ, Qi Z, Zou F, Nonami A, Li Y, Fernandes SM, Adamia S, Stone RM, Galinsky IA, Wang X, Yang G, Griffin JD, Brown JR, Eck MJ, Liu J, Gray NS, Liu Q. Discovery of a BTK/MNK dual inhibitor for lymphoma and leukemia. Leukemia 2015; 30:173-81. [PMID: 26165234 DOI: 10.1038/leu.2015.180] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 06/28/2015] [Accepted: 06/29/2015] [Indexed: 12/14/2022]
Abstract
Bruton's tyrosine kinase (BTK) kinase is a member of the TEC kinase family and is a key regulator of the B-cell receptor (BCR)-mediated signaling pathway. It is important for B-cell maturation, proliferation, survival and metastasis. Pharmacological inhibition of BTK is clinically effective against a variety of B-cell malignances, such as mantle cell lymphoma, chronic lymphocytic leukemia (CLL), acute myeloid leukemia (AML) and activated B-cell-diffuse large B-cell lymphoma. MNK kinase is one of the key downstream regulators in the RAF-MEK-ERK signaling pathway and controls protein synthesis via regulating the activity of eIF4E. Inhibition of MNK activity has been observed to moderately inhibit the proliferation of AML cells. Through a structure-based drug-design approach, we have discovered a selective and potent BTK/MNK dual kinase inhibitor (QL-X-138), which exhibits covalent binding to BTK and noncovalent binding to MNK. Compared with the BTK kinase inhibitor (PCI-32765) and the MNK kinase inhibitor (cercosporamide), QL-X-138 enhanced the antiproliferative efficacies in vitro against a variety of B-cell cancer cell lines, as well as AML and CLL primary patient cells, which respond moderately to BTK inhibitor in vitro. The agent can effectively arrest the growth of lymphoma and leukemia cells at the G0-G1 stage and can induce strong apoptotic cell death. These primary results demonstrate that simultaneous inhibition of BTK and MNK kinase activity might be a new therapeutic strategy for B-cell malignances.
Collapse
Affiliation(s)
- H Wu
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China.,University of Science and Technology of China, Anhui, Hefei, P. R. China
| | - C Hu
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - A Wang
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China.,University of Science and Technology of China, Anhui, Hefei, P. R. China
| | - E L Weisberg
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Y Chen
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - C-H Yun
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - W Wang
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Y Liu
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - X Liu
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China.,University of Science and Technology of China, Anhui, Hefei, P. R. China
| | - B Tian
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - J Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Z Zhao
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Y Liang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - B Li
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - L Wang
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - B Wang
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - C Chen
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - S J Buhrlage
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Z Qi
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - F Zou
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - A Nonami
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Y Li
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - S M Fernandes
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - S Adamia
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - R M Stone
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - I A Galinsky
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - X Wang
- Department of Lymphoma, Sino-US Center for Lymphoma and Leukemia, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - G Yang
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - J D Griffin
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - J R Brown
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - M J Eck
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - J Liu
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China
| | - N S Gray
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Q Liu
- High Magnetic Field laboratory, Chinese Academy of Sciences, Hefei, Anhui, China.,University of Science and Technology of China, Anhui, Hefei, P. R. China.,Hefei Science Center, Chinese Academy of Sciences, Hefei, Anhui, China
| |
Collapse
|
33
|
Regulatory effects of a Mnk2-eIF4E feedback loop during mTORC1 targeting of human medulloblastoma cells. Oncotarget 2015; 5:8442-51. [PMID: 25193863 PMCID: PMC4226695 DOI: 10.18632/oncotarget.2319] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The mTOR pathway controls mRNA translation of mitogenic proteins and is a central regulator of metabolism in malignant cells. Development of malignant cell resistance is a limiting factor to the effects of mTOR inhibitors, but the mechanisms accounting for such resistance are not well understood. We provide evidence that mTORC1 inhibition by rapamycin results in engagement of a negative feedback regulatory loop in malignant medulloblastoma cells, involving phosphorylation of the eukaryotic translation-initiation factor eIF4E. This eIF4E phosphorylation is Mnk2- mediated, but Mnk1-independent, and acts as a survival mechanism for medulloblastoma cells. Pharmacological targeting of Mnk1/2 or siRNA-mediated knockdown of Mnk2 sensitizes medulloblastoma cells to mTOR inhibition and promotes suppression of malignant cell proliferation and anchorage-independent growth. Altogether, these findings provide evidence for the existence of a Mnk2-controlled feedback loop in medulloblastoma cells that accounts for resistance to mTOR inhibitors, and raise the potential for combination treatments of mTOR and Mnk inhibitors for the treatment of medulloblastoma.
Collapse
|
34
|
Martínez A, Sesé M, Losa JH, Robichaud N, Sonenberg N, Aasen T, Ramón y Cajal S. Phosphorylation of eIF4E Confers Resistance to Cellular Stress and DNA-Damaging Agents through an Interaction with 4E-T: A Rationale for Novel Therapeutic Approaches. PLoS One 2015; 10:e0123352. [PMID: 25923732 PMCID: PMC4414544 DOI: 10.1371/journal.pone.0123352] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 03/02/2015] [Indexed: 12/25/2022] Open
Abstract
Phosphorylation of the eukaryotic translation initiation factor eIF4E is associated with malignant progression and poor cancer prognosis. Accordingly, here we have analyzed the association between eIF4E phosphorylation and cellular resistance to oxidative stress, starvation, and DNA-damaging agents in vitro. Using immortalized and cancer cell lines, retroviral expression of a phosphomimetic (S209D) form of eIF4E, but not phospho-dead (S209A) eIF4E or GFP control, significantly increased cellular resistance to stress induced by DNA-damaging agents (cisplatin), starvation (glucose+glutamine withdrawal), and oxidative stress (arsenite). De novo accumulation of eIF4E-containing cytoplasmic bodies colocalizing with the eIF4E-binding protein 4E-T was observed after expression of phosphomimetic S209D, but not S209A or wild-type eIF4E. Increased resistance to cellular stress induced by eIF4E-S209D was lost upon knockdown of endogenous 4E-T or use of an eIF4E-W73A-S209D mutant unable to bind 4E-T. Cancer cells treated with the Mnk1/2 inhibitor CGP57380 to prevent eIF4E phosphorylation and mouse embryonic fibroblasts derived from Mnk1/2 knockout mice were also more sensitive to arsenite and cisplatin treatment. Polysome analysis revealed an 80S peak 2 hours after arsenite treatment in cells overexpressing phosphomimetic eIF4E, indicating translational stalling. Nonetheless, a selective increase was observed in the synthesis of some proteins (cyclin D1, HuR, and Mcl-1). We conclude that phosphorylation of eIF4E confers resistance to various cell stressors and that a direct interaction or regulation of 4E-T by eIF4E is required. Further delineation of this process may identify novel therapeutic avenues for cancer treatment, and these results support the use of modern Mnk1/2 inhibitors in conjunction with standard therapy.
Collapse
Affiliation(s)
- Alba Martínez
- Molecular Pathology, Hospital Universitari Vall d’Hebron, Vall d'Hebron Institut de Recerca, VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Sesé
- Molecular Pathology, Hospital Universitari Vall d’Hebron, Vall d'Hebron Institut de Recerca, VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Nathaniel Robichaud
- McGill University, Department of Biochemistry, Goodman Cancer Research Centre, Montréal, Québec, Canada
| | - Nahum Sonenberg
- McGill University, Department of Biochemistry, Goodman Cancer Research Centre, Montréal, Québec, Canada
| | - Trond Aasen
- Molecular Pathology, Hospital Universitari Vall d’Hebron, Vall d'Hebron Institut de Recerca, VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
- * E-mail: (SRC); (TA)
| | - Santiago Ramón y Cajal
- Molecular Pathology, Hospital Universitari Vall d’Hebron, Vall d'Hebron Institut de Recerca, VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Pathology, Vall d’Hebron University Hospital, Barcelona, Spain
- * E-mail: (SRC); (TA)
| |
Collapse
|
35
|
BDNF stimulation of protein synthesis in cortical neurons requires the MAP kinase-interacting kinase MNK1. J Neurosci 2015; 35:972-84. [PMID: 25609615 DOI: 10.1523/jneurosci.2641-14.2015] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although the MAP kinase-interacting kinases (MNKs) have been known for >15 years, their roles in the regulation of protein synthesis have remained obscure. Here, we explore the involvement of the MNKs in brain-derived neurotrophic factor (BDNF)-stimulated protein synthesis in cortical neurons from mice. Using a combination of pharmacological and genetic approaches, we show that BDNF-induced upregulation of protein synthesis requires MEK/ERK signaling and the downstream kinase, MNK1, which phosphorylates eukaryotic initiation factor (eIF) 4E. Translation initiation is mediated by the interaction of eIF4E with the m(7)GTP cap of mRNA and with eIF4G. The latter interaction is inhibited by the interactions of eIF4E with partner proteins, such as CYFIP1, which acts as a translational repressor. We find that BDNF induces the release of CYFIP1 from eIF4E, and that this depends on MNK1. Finally, using a novel combination of BONCAT and SILAC, we identify a subset of proteins whose synthesis is upregulated by BDNF signaling via MNK1 in neurons. Interestingly, this subset of MNK1-sensitive proteins is enriched for functions involved in neurotransmission and synaptic plasticity. Additionally, we find significant overlap between our subset of proteins whose synthesis is regulated by MNK1 and those encoded by known FMRP-binding mRNAs. Together, our data implicate MNK1 as a key component of BDNF-mediated translational regulation in neurons.
Collapse
|
36
|
The MAP kinase-interacting kinases regulate cell migration, vimentin expression and eIF4E/CYFIP1 binding. Biochem J 2015; 467:63-76. [DOI: 10.1042/bj20141066] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The MAP kinase-interacting kinases (Mnk1 and Mnk2) are activated by ERK and are best known for phosphorylating the translation initiation factor eIF4E. Genetic knockout of the Mnks impaired the migration of embryonic fibroblasts both in two-dimensional wound-healing experiments and in three-dimensional migration assays. Furthermore, a novel and selective Mnk inhibitor, Mnk-I1, which potently blocks eIF4E phosphorylation, blocked the migration of fibroblasts and cancer cells, without exerting ‘off-target’ effects on other signalling pathways such as Erk. Mnk-I1 or genetic knockout of the Mnks decreased the expression of vimentin, a marker of mesenchymal cells, without affecting vimentin mRNA levels. Vimentin protein levels were much lower in Mnk1/2-knockout cells than in controls, although mRNA levels were similar. Our data suggest that the Mnks regulate the translation of the vimentin mRNA and the stability of the vimentin protein. Inhibition or genetic knockout of the Mnks increased the binding of eIF4E to the cytoplasmic FMRP-interacting protein 1 (CYFIP1), which binds the fragile-X mental retardation protein, FMRP, a translational repressor. Since FMRP binds mRNAs for proteins involved in metastasis, the Mnk-dependent release of CYFIP1 from eIF4E is expected to release the repression of translation of FMRP-bound mRNAs, potentially providing a molecular mechanism for the control of cell migration by the Mnks. As Mnk1/2 are not essential for viability, inhibition of the Mnks may be a useful approach to tackling cancer metastasis, a key process contributing to mortality in cancer patients.
Collapse
|
37
|
Sakamoto KM, Grant S, Saleiro D, Crispino JD, Hijiya N, Giles F, Platanias L, Eklund EA. Targeting novel signaling pathways for resistant acute myeloid leukemia. Mol Genet Metab 2015; 114:397-402. [PMID: 25533111 PMCID: PMC4355162 DOI: 10.1016/j.ymgme.2014.11.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 11/28/2014] [Accepted: 11/28/2014] [Indexed: 01/23/2023]
Abstract
Acute myeloid leukemia (AML) is a hematologic malignancy that is the most common type of acute leukemia diagnosed in adults and the second most common type in children. The overall survival is poor and treatment is associated with significant complications and even death. In addition, a significant number of patients will not respond to therapy or relapse. In this review, several new signaling proteins aberrantly regulated in AML are described, including CREB, Triad1, Bcl-2 family members, Stat3, and mTOR/MEK. Identifying more effective and less toxic agents will provide novel approaches to treat AML.
Collapse
Affiliation(s)
- Kathleen M Sakamoto
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven Grant
- Division of Hematology/Oncology and Palliative Care, Department of Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Diana Saleiro
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Department of Medicine, Northwestern University Medical School of Medicine, Chicago, IL, USA
| | - John D Crispino
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Department of Medicine, Northwestern University Medical School of Medicine, Chicago, IL, USA
| | - Nobuko Hijiya
- Division of Hematology/Oncology, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Francis Giles
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Department of Medicine, Northwestern University Medical School of Medicine, Chicago, IL, USA
| | - Leonidas Platanias
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Department of Medicine, Northwestern University Medical School of Medicine, Chicago, IL, USA; Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Elizabeth A Eklund
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Department of Medicine, Northwestern University Medical School of Medicine, Chicago, IL, USA; Division of Hematology-Oncology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
38
|
Teo T, Yu M, Yang Y, Gillam T, Lam F, Sykes MJ, Wang S. Pharmacologic co-inhibition of Mnks and mTORC1 synergistically suppresses proliferation and perturbs cell cycle progression in blast crisis-chronic myeloid leukemia cells. Cancer Lett 2015; 357:612-23. [DOI: 10.1016/j.canlet.2014.12.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 01/03/2023]
|
39
|
Proud CG. Mnks, eIF4E phosphorylation and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:766-73. [PMID: 25450520 DOI: 10.1016/j.bbagrm.2014.10.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/06/2014] [Accepted: 10/18/2014] [Indexed: 01/18/2023]
Abstract
The MAP kinase signal-integrating kinases or MAP kinase-interacting protein kinases (Mnks) are activated by signaling through the oncogenic MAP kinase (ERK) pathway. The best-known Mnk substrate is eukaryotic initiation factor eIF4E, the protein which binds the 5'-cap structure of eukaryotic mRNAs and helps to recruit ribosomes to them. eIF4E is a well-established proto-oncogene, whose expression or activation is associated with transformation and tumorigenesis. Mnks phosphorylate eIF4E at a single site. Increasing evidence implicates the Mnks and/or phosphorylation of eIF4E in cell transformation, tumorigenesis or tumor progression, in a growing range of settings. Mnks and/or the phosphorylation of eIF4E have been suggested to regulate the expression of proteins involved in cell cycle progression, cell survival and cell motility. Further work is needed to extend our understanding of the impact of the Mnks on gene expression, explore the biochemical mechanisms involved and evaluate the utility of targeting the Mnks in cancer therapy. This article is part of a Special Issue entitled: Translation and Cancer.
Collapse
Affiliation(s)
- Christopher G Proud
- South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia.
| |
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
|
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
|