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
|
Xu W, Kannan S, Verma CS, Nacro K. Update on the Development of MNK Inhibitors as Therapeutic Agents. J Med Chem 2021; 65:983-1007. [PMID: 34533957 DOI: 10.1021/acs.jmedchem.1c00368] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mitogen-activated protein kinase-interacting kinases 1 and 2 (MNK1/2) represent a central class of enzymes that are activated by extracellular signal-regulated kinase (ERK) or p38 mitogen-activated protein (MAP) kinases. MNK1 and MNK2 coordinate cellular signaling, control production of inflammatory chemokines, and regulate cell proliferation and survival. MNK1/2 are referred to as serine/threonine kinases as they phosphorylate serine or threonine residues on their substrates. Upon activation, MNK1/2 phosphorylate eukaryotic translation initiation factor 4E (eIF4E) at Ser209, which in turn initiates ribosome assembly and protein translation. Deleterious overexpression of MNK1/2 and/or eIF4E have been reported in several diseases including cancers, neurological disorders, autism, and inflammation. Recently, there have been intense efforts toward the development of potent and selective inhibitors of MNK1/2 in both academia and industry. Herein, we review the current understanding of the structural and biological aspects of MNK1/2 and provide an update of pharmacological inhibitors of MNK1/2 including candidates in clinical trials.
Collapse
Affiliation(s)
- Weijun Xu
- Experimental Drug Development Centre (EDDC), A*STAR, 10 Biopolis Road, Chromos #05-01, 138670, Singapore
| | | | - Chandra S Verma
- Bioinformatics Institute (BII), A*STAR, 30 Biopolis Street, #07-01 Matrix, 138671, Singapore.,Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, 117558, Singapore.,School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore
| | - Kassoum Nacro
- Experimental Drug Development Centre (EDDC), A*STAR, 10 Biopolis Road, Chromos #05-01, 138670, Singapore
| |
Collapse
|
3
|
Jin X, Yu R, Wang X, Proud CG, Jiang T. Progress in developing MNK inhibitors. Eur J Med Chem 2021; 219:113420. [PMID: 33892273 DOI: 10.1016/j.ejmech.2021.113420] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/19/2022]
Abstract
The MNKs (mitogen-activated protein kinase-interacting protein kinases) phosphorylate eIF4E (eukaryotic initiation factor 4 E) at serine 209; eIF4E plays an important role in the translation of cytoplasmic mRNAs, all of which possess a 5' 'cap' structure to which eIF4E binds. Elevated levels of eIF4E, p-eIF4E and/or the MNK protein kinases have been found in many types of cancer, including solid tumors and leukemia. MNKs also play a role in metabolic disease. Regulation of the activities of MNKs (MNK1 and MNK2), control the phosphorylation of eIF4E, which in turn has a close relationship with the processes of tumor development, cell migration and invasion, and energy metabolism. MNK knock-out mice display no adverse effects on normal cells or phenotypes suggesting that MNK may be a potentially safe targets for the treatment of various cancers. Several MNK inhibitors or 'degraders' have been identified. Initially, some of the inhibitors were developed from natural products or based on other protein kinase inhibitors which inhibit multiple kinases. Subsequently, more potent and selective inhibitors for MNK1/2 have been designed and synthesized. Currently, three inhibitors (BAY1143269, eFT508 and ETC-206) are in various stages of clinical trials for the treatment of solid cancers or leukemia, either alone or combined with inhibitors of other protein kinase. In this review, we summarize the diverse MNK inhibitors that have been reported in patents and other literature, including those with activities in vitro and/or in vivo.
Collapse
Affiliation(s)
- Xin Jin
- School of Medicine and Pharmacy, Ocean University of China and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Rilei Yu
- School of Medicine and Pharmacy, Ocean University of China and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xuemin Wang
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA5000, Australia; School of Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Christopher G Proud
- Lifelong Health, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA5000, Australia; School of Biomedical Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Tao Jiang
- School of Medicine and Pharmacy, Ocean University of China and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
4
|
Shukla T, de la Peña JB, Perish JM, Ploski JE, Stumpf CR, Webster KR, Thorn CA, Campbell ZT. A Highly Selective MNK Inhibitor Rescues Deficits Associated with Fragile X Syndrome in Mice. Neurotherapeutics 2021; 18:624-639. [PMID: 33006091 PMCID: PMC8116363 DOI: 10.1007/s13311-020-00932-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2020] [Indexed: 12/22/2022] Open
Abstract
Fragile X syndrome (FXS) is the most common inherited source of intellectual disability in humans. FXS is caused by mutations that trigger epigenetic silencing of the Fmr1 gene. Loss of Fmr1 results in increased activity of the mitogen-activated protein kinase (MAPK) pathway. An important downstream consequence is activation of the mitogen-activated protein kinase interacting protein kinase (MNK). MNK phosphorylates the mRNA cap-binding protein, eukaryotic initiation factor 4E (eIF4E). Excessive phosphorylation of eIF4E has been directly implicated in the cognitive and behavioral deficits associated with FXS. Pharmacological reduction of eIF4E phosphorylation is one potential strategy for FXS treatment. We demonstrate that systemic dosing of a highly specific, orally available MNK inhibitor, eFT508, attenuates numerous deficits associated with loss of Fmr1 in mice. eFT508 resolves a range of phenotypic abnormalities associated with FXS including macroorchidism, aberrant spinogenesis, and alterations in synaptic plasticity. Key behavioral deficits related to anxiety, social interaction, obsessive and repetitive activities, and object recognition are ameliorated by eFT508. Collectively, this work establishes eFT508 as a potential means to reverse deficits associated with FXS.
Collapse
Affiliation(s)
- Tarjani Shukla
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - June Bryan de la Peña
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - John M Perish
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Jonathan E Ploski
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | | | | | - Catherine A Thorn
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Zachary T Campbell
- Department of Biological Sciences, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA.
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
5
|
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
|
6
|
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: 70] [Impact Index Per Article: 11.7] [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
|
7
|
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
|
8
|
Jin X, Merrett J, Tong S, Flower B, Xie J, Yu R, Tian S, Gao L, Zhao J, Wang X, Jiang T, Proud CG. Design, synthesis and activity of Mnk1 and Mnk2 selective inhibitors containing thieno[2,3-d]pyrimidine scaffold. Eur J Med Chem 2018; 162:735-751. [PMID: 30496989 DOI: 10.1016/j.ejmech.2018.10.070] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/25/2018] [Accepted: 10/31/2018] [Indexed: 01/10/2023]
Abstract
The mitogen-activated protein kinase-interacting kinases 1 and 2 (MNK1 and MNK2) phosphorylate eukaryotic initiation factor 4E (eIF4E) and play important roles in promoting tumorigenesis and metabolic disease. Thus, inhibiting these enzymes might be valuable in the treatment of such conditions. We designed and synthesized a series of 4-((4-fluoro-2-isopropoxyphenyl)amino)-5-methylthieno[2,3-d]pyrimidine derivatives, and evaluated their inhibitory activity against the MNKs. We found 15 compounds that were active as MNK inhibitors and that one in particular, designated MNK-7g, which was potent against MNK1 and substantially more potent against MNK2. The compound MNK-7g did not affect other signaling pathways tested and had no adverse effects on cell viability. As expected from earlier studies, MNK-7g also inhibited cell migration. Therefore, the compound MNK-7g, which forms an ionic bond with Asp226 in MNK2 and possesses a substituted aniline in a thieno[2,3-d] pyrimidine structure, is a promising starting point for the future development of novel drugs for treating or managing cancer and metabolic disease.
Collapse
Affiliation(s)
- Xin Jin
- School of Medicine and Pharmacy, Ocean University of China, and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - James Merrett
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
| | - Sheng Tong
- School of Medicine and Pharmacy, Ocean University of China, and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Bartholomew Flower
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
| | - Jianling Xie
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
| | - Rilei Yu
- School of Medicine and Pharmacy, Ocean University of China, and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shuye Tian
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong, China
| | - Xuemin Wang
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Tao Jiang
- School of Medicine and Pharmacy, Ocean University of China, and Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Christopher G Proud
- Nutrition & Metabolism, South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
9
|
Yang H, Chennamaneni LR, Ho MWT, Ang SH, Tan ESW, Jeyaraj DA, Yeap YS, Liu B, Ong EH, Joy JK, Wee JLK, Kwek P, Retna P, Dinie N, Nguyen TTH, Tai SJ, Manoharan V, Pendharkar V, Low CB, Chew YS, Vuddagiri S, Sangthongpitag K, Choong ML, Lee MA, Kannan S, Verma CS, Poulsen A, Lim S, Chuah C, Ong TS, Hill J, Matter A, Nacro K. Optimization of Selective Mitogen-Activated Protein Kinase Interacting Kinases 1 and 2 Inhibitors for the Treatment of Blast Crisis Leukemia. J Med Chem 2018; 61:4348-4369. [PMID: 29683667 DOI: 10.1021/acs.jmedchem.7b01714] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease caused by bcr-abl1, a constitutively active tyrosine kinase fusion gene responsible for an abnormal proliferation of leukemic stem cells (LSCs). Inhibition of BCR-ABL1 kinase activity offers long-term relief to CML patients. However, for a proportion of them, BCR-ABL1 inhibition will become ineffective at treating the disease, and CML will progress to blast crisis (BC) CML with poor prognosis. BC-CML is often associated with excessive phosphorylated eukaryotic translation initiation factor 4E (eIF4E), which renders LSCs capable of proliferating via self-renewal, oblivious to BCR-ABL1 inhibition. In vivo, eIF4E is exclusively phosphorylated on Ser209 by MNK1/2. Consequently, a selective inhibitor of MNK1/2 should reduce the level of phosphorylated eIF4E and re-sensitize LSCs to BCR-ABL1 inhibition, thus hindering the proliferation of BC LSCs. We report herein the structure-activity relationships and pharmacokinetic properties of a selective MNK1/2 inhibitor clinical candidate, ETC-206, which in combination with dasatinib prevents BC-CML LSC self-renewal in vitro and enhances dasatinib antitumor activity in vivo.
Collapse
Affiliation(s)
- Haiyan Yang
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Lohitha Rao Chennamaneni
- Organic Chemistry, Institute of Chemical and Engineering Sciences (ICES), A*STAR , 8 Biomedical Grove, Neuros, #07-01 , 138665 Singapore
| | - Melvyn Wai Tuck Ho
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Shi Hua Ang
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Eldwin Sum Wai Tan
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | | | - Yoon Sheng Yeap
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Boping Liu
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Esther Hq Ong
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Joma Kanikadu Joy
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - John Liang Kuan Wee
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Perlyn Kwek
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Priya Retna
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Nurul Dinie
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Thuy Thi Hanh Nguyen
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Shi Jing Tai
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Vithya Manoharan
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Vishal Pendharkar
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Choon Bing Low
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Yun Shan Chew
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Susmitha Vuddagiri
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Kanda Sangthongpitag
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Meng Ling Choong
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - May Ann Lee
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | | | - Chandra S Verma
- Bioinformatics Institute (BII) , A*STAR , 30 Biopolis Street, #07-01 Matrix , 138671 Singapore.,School of Biological Sciences , Nanyang Technological University , 60 Nanyang Drive , 637551 Singapore.,Department of Biological Sciences , National University of Singapore , 14 Science Drive 4 , 117543 Singapore
| | - Anders Poulsen
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Sharon Lim
- Duke-NUS Medical School , 8 College Road , 169857 Singapore
| | - Charles Chuah
- Duke-NUS Medical School , 8 College Road , 169857 Singapore
| | - Tiong Sin Ong
- Duke-NUS Medical School , 8 College Road , 169857 Singapore.,Department of Medicine , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Jeffrey Hill
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Alex Matter
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| | - Kassoum Nacro
- Experimental Therapeutics Centre (ETC) , A*STAR , 31 Biopolis Way, Nanos #03-01 , 138669 Singapore
| |
Collapse
|
10
|
Tian S, Wang X, Proud CG. Oncogenic MNK signalling regulates the metastasis suppressor NDRG1. Oncotarget 2018; 8:46121-46135. [PMID: 28545025 PMCID: PMC5542254 DOI: 10.18632/oncotarget.17555] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/28/2017] [Indexed: 12/18/2022] Open
Abstract
The protein N-myc down-regulated gene 1 (NDRG1) represses tumour metastasis. It is phosphorylated at several sites by serum and glucocorticoid-regulated kinase 1 (SGK1). Here we show that NDRG1 is also regulated by the oncogenic MAP kinase-interacting kinase (MNK) pathway, a target for cancer therapy.Inhibiting MNKs increases the expression of NDRG1 protein and mRNA in breast cancer cells. MNK inhibition also decreases the phosphorylation of NDRG1. Phosphorylation of NDRG1 is reduced in cells lacking MNK1, but not MNK2-knockout cells, indicating that NDRG1 phosphorylation is a specific target for MNK1. However, MNK1 cannot directly phosphorylate NDRG1 in vitro, indicating that additional signalling connections are involved. Taken together, our data indicate that MNK signaling regulates NDRG1 at transcriptional and post-translational levels.We show that SGK1 phosphorylates MNK1 at a conserved site, which represses its activity. NDRG1, SGK1 and the MNKs are implicated in cell migration and metastasis. As expected, knocking-down NDRG1 promoted cell migration. However, whereas MNK inhibition impairs these processes irrespective of NDRG1 levels, SGK inhibition only did so in NDRG1-depleted cells. Thus, MNKs and SGK affect migration/invasion through distinct mechanisms.Our data reveal several novel connections between signalling pathways important for tumour biology.
Collapse
Affiliation(s)
- Shuye Tian
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide SA5000, Australia.,School of Biological Sciences, University of Adelaide, Adelaide SA5005, Australia
| | - Xuemin Wang
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide SA5000, Australia.,School of Biological Sciences, University of Adelaide, Adelaide SA5005, Australia
| | - Christopher G Proud
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide SA5000, Australia.,School of Biological Sciences, University of Adelaide, Adelaide SA5005, Australia
| |
Collapse
|
11
|
Abstract
As obligate parasites, viruses strictly depend on host cell translation for the production of new progeny, yet infected cells also synthesize antiviral proteins to limit virus infection. Modulation of host cell translation therefore represents a frequent strategy by which viruses optimize their replication and spread. Here we sought to define how host cell translation is regulated during infection of human cells with dengue virus (DENV) and Zika virus (ZIKV), two positive-strand RNA flaviviruses. Polysome profiling and analysis of de novo protein synthesis revealed that flavivirus infection causes potent repression of host cell translation, while synthesis of viral proteins remains efficient. Selective repression of host cell translation was mediated by the DENV polyprotein at the level of translation initiation. In addition, DENV and ZIKV infection suppressed host cell stress responses such as the formation of stress granules and phosphorylation of the translation initiation factor eIF2α (α subunit of eukaryotic initiation factor 2). Mechanistic analyses revealed that translation repression was uncoupled from the disruption of stress granule formation and eIF2α signaling. Rather, DENV infection induced p38-Mnk1 signaling that resulted in the phosphorylation of the eukaryotic translation initiation factor eIF4E and was essential for the efficient production of virus particles. Together, these results identify the uncoupling of translation suppression from the cellular stress responses as a conserved strategy by which flaviviruses ensure efficient replication in human cells. For efficient production of new progeny, viruses need to balance their dependency on the host cell translation machinery with potentially adverse effects of antiviral proteins produced by the infected cell. To achieve this, many viruses evolved mechanisms to manipulate host cell translation. Here we find that infection of human cells with two major human pathogens, dengue virus (DENV) and Zika virus (ZIKV), leads to the potent repression of host cell translation initiation, while the synthesis of viral protein remains unaffected. Unlike other RNA viruses, these flaviviruses concomitantly suppress host cell stress responses, thereby uncoupling translation suppression from stress granule formation. We identified that the p38-Mnk1 cascade regulating phosphorylation of eIF4E is a target of DENV infection and plays an important role in virus production. Our results define several molecular interfaces by which flaviviruses hijack host cell translation and interfere with stress responses to optimize the production of new virus particles.
Collapse
|
12
|
Roy A, Srivastava M, Saqib U, Liu D, Faisal SM, Sugathan S, Bishnoi S, Baig MS. Potential therapeutic targets for inflammation in toll-like receptor 4 (TLR4)-mediated signaling pathways. Int Immunopharmacol 2016; 40:79-89. [PMID: 27584057 DOI: 10.1016/j.intimp.2016.08.026] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 08/08/2016] [Accepted: 08/23/2016] [Indexed: 12/13/2022]
Abstract
Inflammation is set off when innate immune cells detect infection or tissue injury. Tight control of the severity, duration, and location of inflammation is an absolute requirement for an appropriate balance between clearance of injured tissue and pathogens versus damage to host cells. Impeding the risk associated with the imbalance in the inflammatory response requires precise identification of potential therapeutic targets involved in provoking the inflammation. Toll-like receptors (TLRs) primarily known for the pathogen recognition and subsequent immune responses are being investigated for their pathogenic role in various chronic diseases. A mammalian homologue of Drosophila Toll receptor 4 (TLR4) was shown to induce the expression of genes involved in inflammatory responses. Signaling pathways via TLR4 activate various transcription factors like Nuclear factor kappa-light-chain-enhancer (NF-κB), activator protein 1 (AP1), Signal Transducers and Activators of Transcription family of transcription factors (STAT1) and Interferon regulatory factors (IRF's), which are the key players regulating the inflammatory response. Inhibition of these targets and their upstream signaling molecules provides a potential therapeutic approach to treat inflammatory diseases. Here we review the therapeutic targets involved in TLR-4 signaling pathways that are critical for suppressing chronic inflammatory disorders.
Collapse
Affiliation(s)
- Anjali Roy
- Center for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology (IIT), Indore, MP, India
| | - Mansi Srivastava
- Center for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology (IIT), Indore, MP, India
| | - Uzma Saqib
- Discipline of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore (IITI), Indore, MP, India
| | - Dongfang Liu
- Center for Inflammation & Epigenetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Syed M Faisal
- National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Subi Sugathan
- Center for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology (IIT), Indore, MP, India
| | - Suman Bishnoi
- Center for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology (IIT), Indore, MP, India
| | - Mirza S Baig
- Center for Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology (IIT), Indore, MP, India.
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Moore CEJ, Pickford J, Cagampang FR, Stead RL, Tian S, Zhao X, Tang X, Byrne CD, Proud CG. MNK1 and MNK2 mediate adverse effects of high-fat feeding in distinct ways. Sci Rep 2016; 6:23476. [PMID: 27087055 PMCID: PMC4834573 DOI: 10.1038/srep23476] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/07/2016] [Indexed: 02/06/2023] Open
Abstract
The MAP kinase-interacting kinases (MNK1 and MNK2) are non-essential enzymes which are activated by MAP kinases. They are implicated in controlling protein synthesis. Here we show that mice in which the expression of either MNK1 or MNK2 has been knocked out (KO) are protected against adverse effects of high-fat feeding, and in distinct ways. High-fat diet (HFD)-fed MNK2-KO show less weight gain than wild-type animals, and improved glucose tolerance, better insulin sensitivity and markedly diminished adipose tissue inflammation. This suggests MNK2 plays a role in adipogenesis and/or lipogenesis and in macrophage biology. MNK1-KO/HFD mice show better glucose tolerance and insulin sensitivity, but gain weight and show similar adipose inflammation to WT animals. These data suggest MNK1 participates in mediating HFD-induced insulin resistance. Our findings reveal distinct roles for the MNKs in a novel area of disease biology, metabolic dysfunction, and suggests they are potential new targets for managing metabolic disease.
Collapse
Affiliation(s)
- C E J Moore
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom.,Nutrition and Metabolism, South Australian Health &Medical Research Institute, North Terrace, Adelaide, SA5000, Australia
| | - J Pickford
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - F R Cagampang
- Institute of Developmental Sciences, University of Southampton, Faculty of Medicine, Southampton SO16 6YD, United Kingdom
| | - R L Stead
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - S Tian
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom.,Nutrition and Metabolism, South Australian Health &Medical Research Institute, North Terrace, Adelaide, SA5000, Australia
| | - X Zhao
- Nutrition and Metabolism, South Australian Health &Medical Research Institute, North Terrace, Adelaide, SA5000, Australia
| | - X Tang
- Department of Biochemistry &Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058 China
| | - C D Byrne
- Nutrition and Metabolism, University of Southampton, Faculty of Medicine, Southampton SO16 6YD, United Kingdom.,Southampton National Institute for Health Research, Biomedical Research Centre, University Hospital, Southampton, UK
| | - C G Proud
- Centre for Biological Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom.,Nutrition and Metabolism, South Australian Health &Medical Research Institute, North Terrace, Adelaide, SA5000, Australia.,School of Biological Sciences, University of Adelaide, Adelaide SA 5005, Australia
| |
Collapse
|
16
|
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
|
17
|
Carter JH, Deddens JA, Spaulding NR, Lucas D, Colligan BM, Lewis TG, Hawkins E, Jones J, Pemberton JO, Douglass LE, Graff JR. Phosphorylation of eIF4E serine 209 is associated with tumour progression and reduced survival in malignant melanoma. Br J Cancer 2016; 114:444-53. [PMID: 26882068 PMCID: PMC4815768 DOI: 10.1038/bjc.2015.450] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/18/2015] [Accepted: 11/30/2015] [Indexed: 02/08/2023] Open
Abstract
Background: Melanoma is a disease that primarily arises in the skin but is a derivative of the neural crest. Eukaryotic translation initiation factor 4E (eIF4E) regulates translation of multiple malignancy-associated mRNAs and is overexpressed in many epithelial tumours. However, expression in human tumours derived from the neural crest is unknown. Here, we determined the association of eIF4E and phospho-eIF4E expression in melanocytic lesions with malignant conversion, metastatic potential and patient survival. Methods: Archived formalin-fixed, paraffin-embedded surgical specimens from 114 patients with melanocytic lesions were stained immunohistochemically for eIF4E and phospho-eIF4E and evaluated semiquantitatively. The relationship between cytoplasmic and nuclear eIF4E and phospho-eIF4E protein expression, melanocytic lesion subtype and tumour progression was determined. Kaplan–Meier survival analyses and Cox proportional hazard regression were performed. Results: Increased eIF4E and phospho-eIF4E expression was highly associated with malignancy (P<0.0001). High nuclear phospho-eIF4E was associated with synchronous or future metastasis (P=0.0059). Kaplan–Meier analyses demonstrated highly significant associations between high histoscores for cytoplasmic and nuclear phospho-eIF4E and reduced survival in all patients (P=0.0003 and 0.0009, respectively). Conclusions: Increased melanoma expression of eIF4E and phospho-eIF4E is associated with metastatic potential, reduced survival and increased risk of death.
Collapse
Affiliation(s)
- Julia H Carter
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - James A Deddens
- Department of Mathematical Sciences, University of Cincinnati, 4512 French Hall, ML 25, Cincinnati, OH 45221, USA
| | - Nelson Reed Spaulding
- Department of Pathology and Laboratory Medicine, University of Louisville School of Medicine, 627 South Preston Street, Louisville, KY 40292, USA
| | - Denise Lucas
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Bruce M Colligan
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Thomas Grant Lewis
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Elyse Hawkins
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Jordan Jones
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Jackson O Pemberton
- St Elizabeth Healthcare, Department of Laboratory Medicine, 20 Medical Village Drive, Edgewood, KY 41018, USA
| | - Larry E Douglass
- Wood Hudson Cancer Research Laboratory, 931 Isabella Street, Newport, KY 41071, USA
| | - Jeremy R Graff
- Eli Lilly & Company, Lilly Corporate Center, DC 0546, Indianapolis, IN 46285, USA
| |
Collapse
|
18
|
Abstract
The translation initiation factor eIF4E mediates a rate-limiting process that drives selective translation of many oncongenic proteins such as cyclin D1, survivin and VEGF, thereby contributing to tumour growth, metastasis and therapy resistance. As an essential regulatory hub in cancer signalling network, many oncogenic signalling pathways appear to converge on eIF4E. Therefore, targeting eIF4E-mediated cap-dependent translation is considered a promising anticancer strategy. This paper reviews the strategies that can be used to target eIF4E, highlighting agents that target eIF4E activity at each distinct level.
Collapse
|
19
|
Korneeva NL, Song A, Gram H, Edens MA, Rhoads RE. Inhibition of Mitogen-activated Protein Kinase (MAPK)-interacting Kinase (MNK) Preferentially Affects Translation of mRNAs Containing Both a 5'-Terminal Cap and Hairpin. J Biol Chem 2015; 291:3455-67. [PMID: 26668315 DOI: 10.1074/jbc.m115.694190] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Indexed: 12/22/2022] Open
Abstract
The MAPK-interacting kinases 1 and 2 (MNK1 and MNK2) are activated by extracellular signal-regulated kinases 1 and 2 (ERK1/2) or p38 in response to cellular stress and extracellular stimuli that include growth factors, cytokines, and hormones. Modulation of MNK activity affects translation of mRNAs involved in the cell cycle, cancer progression, and cell survival. However, the mechanism by which MNK selectively affects translation of these mRNAs is not understood. MNK binds eukaryotic translation initiation factor 4G (eIF4G) and phosphorylates the cap-binding protein eIF4E. Using a cell-free translation system from rabbit reticulocytes programmed with mRNAs containing different 5'-ends, we show that an MNK inhibitor, CGP57380, affects translation of only those mRNAs that contain both a cap and a hairpin in the 5'-UTR. Similarly, a C-terminal fragment of human eIF4G-1, eIF4G(1357-1600), which prevents binding of MNK to intact eIF4G, reduces eIF4E phosphorylation and inhibits translation of only capped and hairpin-containing mRNAs. Analysis of proteins bound to m(7)GTP-Sepharose reveals that both CGP and eIF4G(1357-1600) decrease binding of eIF4E to eIF4G. These data suggest that MNK stimulates translation only of mRNAs containing both a cap and 5'-terminal RNA duplex via eIF4E phosphorylation, thereby enhancing the coupled cap-binding and RNA-unwinding activities of eIF4F.
Collapse
Affiliation(s)
- Nadejda L Korneeva
- From the Departments of Emergency Medicine and Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, and
| | - Anren Song
- Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, and
| | - Hermann Gram
- the Novartis Institute for Biomedical Research, Forum 1, CH-4002 Basel, Switzerland
| | | | - Robert E Rhoads
- Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130-3932, and
| |
Collapse
|
20
|
Chu J, Cencic R, Wang W, Porco JA, Pelletier J. Translation Inhibition by Rocaglates Is Independent of eIF4E Phosphorylation Status. Mol Cancer Ther 2015; 15:136-41. [PMID: 26586722 DOI: 10.1158/1535-7163.mct-15-0409] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/29/2015] [Indexed: 02/06/2023]
Abstract
Rocaglates are natural products that inhibit protein synthesis in eukaryotes and exhibit antineoplastic activity. In vitro biochemical assays, affinity chromatography experiments coupled with mass spectrometry analysis, and in vivo genetic screens have identified eukaryotic initiation factor (eIF) 4A as a direct molecular target of rocaglates. eIF4A is the RNA helicase subunit of eIF4F, a complex that mediates cap-dependent ribosome recruitment to mRNA templates. The eIF4F complex has been implicated in tumor initiation and maintenance through elevated levels or increased phosphorylation status of its cap-binding subunit, eIF4E, thus furthering the interest toward developing rocaglates as antineoplastic agents. Recent experiments have indicated that rocaglates also interact with prohibitins 1 and 2, proteins implicated in c-Raf-MEK-ERK signaling. Because increased ERK signaling stimulates eIF4E phosphorylation status, rocaglates are also expected to inhibit eIF4E phosphorylation status, a point that has not been thoroughly investigated. It is currently unknown whether the effects on translation observed with rocaglates are solely through eIF4A inhibition or also a feature of blocking eIF4E phosphorylation. Here, we show that rocaglates inhibit translation through an eIF4E phosphorylation-independent mechanism.
Collapse
Affiliation(s)
- Jennifer Chu
- Department of Biochemistry, McGill University, Montreal, Québec, Canada
| | - Regina Cencic
- Department of Biochemistry, McGill University, Montreal, Québec, Canada
| | - Wenyu Wang
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts
| | - John A Porco
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Québec, Canada. The Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, Québec, Canada. Department of Oncology, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
21
|
An integrated approach for discovery of highly potent and selective Mnk inhibitors: Screening, synthesis and SAR analysis. Eur J Med Chem 2015; 103:539-50. [DOI: 10.1016/j.ejmech.2015.09.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/07/2014] [Accepted: 09/05/2015] [Indexed: 02/02/2023]
|
22
|
Pelletier J, Graff J, Ruggero D, Sonenberg N. Targeting the eIF4F translation initiation complex: a critical nexus for cancer development. Cancer Res 2015; 75:250-63. [PMID: 25593033 DOI: 10.1158/0008-5472.can-14-2789] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Elevated protein synthesis is an important feature of many cancer cells and often arises as a consequence of increased signaling flux channeled to eukaryotic initiation factor 4F (eIF4F), the key regulator of the mRNA-ribosome recruitment phase of translation initiation. In many cellular and preclinical models of cancer, eIF4F deregulation results in changes in translational efficiency of specific mRNA classes. Importantly, many of these mRNAs code for proteins that potently regulate critical cellular processes, such as cell growth and proliferation, enhanced cell survival and cell migration that ultimately impinge on several hallmarks of cancer, including increased angiogenesis, deregulated growth control, enhanced cellular survival, epithelial-to-mesenchymal transition, invasion, and metastasis. By being positioned as the molecular nexus downstream of key oncogenic signaling pathways (e.g., Ras, PI3K/AKT/TOR, and MYC), eIF4F serves as a direct link between important steps in cancer development and translation initiation. Identification of mRNAs particularly responsive to elevated eIF4F activity that typifies tumorigenesis underscores the critical role of eIF4F in cancer and raises the exciting possibility of developing new-in-class small molecules targeting translation initiation as antineoplastic agents.
Collapse
Affiliation(s)
- Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Québec, Canada. The Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, Québec, Canada. Department of Oncology, McGill University, Montreal, Québec, Canada.
| | - Jeremy Graff
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana
| | - Davide Ruggero
- School of Medicine and Department of Urology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montreal, Québec, Canada. The Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, Québec, Canada
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Yu M, Li P, Basnet SKC, Kumarasiri M, Diab S, Teo T, Albrecht H, Wang S. Discovery of 4-(dihydropyridinon-3-yl)amino-5-methylthieno[2,3-d]pyrimidine derivatives as potent Mnk inhibitors: synthesis, structure-activity relationship analysis and biological evaluation. Eur J Med Chem 2015; 95:116-26. [PMID: 25800647 DOI: 10.1016/j.ejmech.2015.03.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/15/2014] [Accepted: 03/13/2015] [Indexed: 12/22/2022]
Abstract
Phosphorylation of the eukaryotic initiation factor 4E (eIF4E) by mitogen-activated protein kinase (MAPK)-interacting kinases (Mnks) is essential for oncogenesis but unnecessary for normal development. Thus, pharmacological inhibition of Mnks may offer an effective and non-toxic anti-cancer therapeutic strategy. Herein, we report the discovery of 4-(dihydropyridinon-3-yl)amino-5-methylthieno[2,3-d]pyrimidine derivatives as potent Mnk inhibitors. Docking study of 7a in Mnk2 suggests that the compound is stabilised in the ATP binding site through multiple hydrogen bonds and hydrophobic interaction. Cellular mechanistic studies on MV-4-11 cells with leads 7a, 8e and 8f reveal that they are able to down-regulate the phosphorylated eIF4E, Mcl-1 and cyclin D1, and induce apoptosis.
Collapse
Affiliation(s)
- Mingfeng Yu
- Centre for Drug Discovery and Development, Sansom Institute for Health Research and Center for Cancer Biology, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Peng Li
- Centre for Drug Discovery and Development, Sansom Institute for Health Research and Center for Cancer Biology, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Sunita K C Basnet
- Centre for Drug Discovery and Development, Sansom Institute for Health Research and Center for Cancer Biology, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Malika Kumarasiri
- Centre for Drug Discovery and Development, Sansom Institute for Health Research and Center for Cancer Biology, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Sarah Diab
- Centre for Drug Discovery and Development, Sansom Institute for Health Research and Center for Cancer Biology, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Theodosia Teo
- Centre for Drug Discovery and Development, Sansom Institute for Health Research and Center for Cancer Biology, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Hugo Albrecht
- Centre for Drug Discovery and Development, Sansom Institute for Health Research and Center for Cancer Biology, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia
| | - Shudong Wang
- Centre for Drug Discovery and Development, Sansom Institute for Health Research and Center for Cancer Biology, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia 5001, Australia.
| |
Collapse
|
26
|
Abstract
Dysregulation of mRNA translation is a frequent feature of neoplasia. Many oncogenes and tumour suppressors affect the translation machinery, making aberrant translation a widespread characteristic of tumour cells, independent of the genetic make-up of the cancer. Therefore, therapeutic agents that target components of the protein synthesis apparatus hold promise as novel anticancer drugs that can overcome intra-tumour heterogeneity. In this Review, we discuss the role of translation in cancer, with a particular focus on the eIF4F (eukaryotic translation initiation factor 4F) complex, and provide an overview of recent efforts aiming to 'translate' these results to the clinic.
Collapse
|
27
|
Ramalingam S, Gediya L, Kwegyir-Afful AK, Ramamurthy VP, Purushottamachar P, Mbatia H, Njar VCO. First MNKs degrading agents block phosphorylation of eIF4E, induce apoptosis, inhibit cell growth, migration and invasion in triple negative and Her2-overexpressing breast cancer cell lines. Oncotarget 2015; 5:530-43. [PMID: 24504069 PMCID: PMC3964227 DOI: 10.18632/oncotarget.1528] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Some retinoic acid metabolism blocking agents (RAMBAs) are known to exhibit a wide range of anticancer activities by mechanisms that are still not completely resolved. This study investigated the anticancer efficacy and mechanism(s) of novel RAMBA retinamides (RRs) in triple negative and Her-2 overexpressing breast cancer cells. Specifically, we examined the possibility that RRs affect the translational machinery in these breast cancer (BC) cells. Recent findings suggest that overexpression of eukaryotic translation initiation factor 4E (eIF4E) in breast cancers critically augments CAP-dependent mRNA translation and synthesis of proteins involved in cell growth, cell proliferation, invasion and apoptosis evasion. The oncogenic potential of eIF4E is strictly dependent on serine209 phosphorylation by upstream MAPK-interacting kinases (Mnks). Targeting Mnk/eIF4E pathway for blocking Mnk function and eIF4E phosphorylation is therefore a novel approach for treating BCs, particularly for Her2-positive and triple negative breast cancers that have no indications for endocrine therapy or effective treatment regimes. We report for the first time that the degradation of Mnk1 by RRs in BC cells blocks eIF4E phosphorylation and subsequently inhibits cell growth, colonization, invasion, and migration and induce apoptosis. Most importantly, the anticancer efficacy of RRs was mediated via degrading Mnk rather than inhibiting its kinase activity like Mnk inhibitors (cercosporamide and CGP57380). Furthermore, RRs potencies on peIF4E down-regulation and growth inhibition were superior to those of two clinically relevant retinoids and the Mnk inhibitors. Together our findings provide the first preclinical proof-of-concept of novel Mnk degrading agents for Mnk/eIF4E based therapeutic treatment of breast cancers.
Collapse
|
28
|
Karaki S, Andrieu C, Ziouziou H, Rocchi P. The Eukaryotic Translation Initiation Factor 4E (eIF4E) as a Therapeutic Target for Cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 101:1-26. [PMID: 26572974 PMCID: PMC7185574 DOI: 10.1016/bs.apcsb.2015.09.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cancer cells depend on cap-dependent translation more than normal tissue. This explains the emergence of proteins involved in the cap-dependent translation as targets for potential anticancer drugs. Cap-dependent translation starts when eIF4E binds to mRNA cap domain. This review will present eIF4E's structure and functions. It will also expose the use of eIF4E as a therapeutic target in cancer.
Collapse
Affiliation(s)
- Sara Karaki
- INSERM, U1068, CRCM, Marseille, France,Institut Paoli-Calmettes, Marseille, France,Aix-Marseille University, Marseille, France,CNRS, UMR7258, Marseille, France
| | - Claudia Andrieu
- INSERM, U1068, CRCM, Marseille, France,Institut Paoli-Calmettes, Marseille, France,Aix-Marseille University, Marseille, France,CNRS, UMR7258, Marseille, France
| | - Hajer Ziouziou
- INSERM, U1068, CRCM, Marseille, France,Institut Paoli-Calmettes, Marseille, France,Aix-Marseille University, Marseille, France,CNRS, UMR7258, Marseille, France
| | - Palma Rocchi
- INSERM, U1068, CRCM, Marseille, France,Institut Paoli-Calmettes, Marseille, France,Aix-Marseille University, Marseille, France,CNRS, UMR7258, Marseille, France,Corresponding author:
| |
Collapse
|
29
|
Panja D, Kenney J, D’Andrea L, Zalfa F, Vedeler A, Wibrand K, Fukunaga R, Bagni C, Proud C, Bramham C. Two-Stage Translational Control of Dentate Gyrus LTP Consolidation Is Mediated by Sustained BDNF-TrkB Signaling to MNK. Cell Rep 2014; 9:1430-45. [DOI: 10.1016/j.celrep.2014.10.016] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 08/18/2014] [Accepted: 10/03/2014] [Indexed: 12/13/2022] Open
|
30
|
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: 90] [Impact Index Per Article: 8.2] [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
|
31
|
Joshi S, Platanias LC. Mnk kinase pathway: Cellular functions and biological outcomes. World J Biol Chem 2014; 5:321-333. [PMID: 25225600 PMCID: PMC4160526 DOI: 10.4331/wjbc.v5.i3.321] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/10/2014] [Accepted: 06/03/2014] [Indexed: 02/05/2023] Open
Abstract
The mitogen-activated protein kinase (MAPK) interacting protein kinases 1 and 2 (Mnk1 and Mnk2) play important roles in controlling signals involved in mRNA translation. In addition to the MAPKs (p38 or Erk), multiple studies suggest that the Mnk kinases can be regulated by other known kinases such as Pak2 and/or other unidentified kinases by phosphorylation of residues distinct from the sites phosphorylated by the MAPKs. Several studies have established multiple Mnk protein targets, including PSF, heterogenous nuclear ribonucleoprotein A1, Sprouty 2 and have lead to the identification of distinct biological functions and substrate specificity for the Mnk kinases. In this review we discuss the pathways regulating the Mnk kinases, their known substrates as well as the functional consequences of engagement of pathways controlled by Mnk kinases. These kinases play an important role in mRNA translation via their regulation of eukaryotic initiation factor 4E (eIF4E) and their functions have important implications in tumor biology as well as the regulation of drug resistance to anti-oncogenic therapies. Other studies have identified a role for the Mnk kinases in cap-independent mRNA translation, suggesting that the Mnk kinases can exert important functional effects independently of the phosphorylation of eIF4E. The role of Mnk kinases in inflammation and inflammation-induced malignancies is also discussed.
Collapse
|
32
|
Carroll M, Borden KLB. The oncogene eIF4E: using biochemical insights to target cancer. J Interferon Cytokine Res 2013; 33:227-38. [PMID: 23472659 DOI: 10.1089/jir.2012.0142] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The eukaryotic translation initiation factor eIF4E is overexpressed in many human malignancies where it is typically a harbinger of poor prognosis. eIF4E is positioned as a nexus in post-transcriptional gene expression. To carry out these functions, eIF4E needs to bind the m(7)G cap moiety on mRNAs. It plays critical roles in mRNA translation, mRNA export, and most likely in mRNA stability as well. Through these activities, eIF4E coordinately modulates the expression of many transcripts involved in proliferation and survival. eIF4E function is controlled by interactions with protein cofactors in concert with many signaling pathways, including Ras, Mnk, Erk, MAPK, PI3K, mTOR, and Akt. This review describes the eIF4E activity and provides several examples of cellular control mechanisms. Further, we describe some therapeutic strategies in preclinical and clinical development.
Collapse
Affiliation(s)
- Martin Carroll
- Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
33
|
Abstract
Deregulation of protein synthesis is a common event in human cancer and a key player in translational control is eIF4E. Elevated expression levels of eIF4E promote cancer development and progression. Recent findings suggest that eIF4E activity is a key determinant of the PI3K/Akt/mTOR and Ras/Raf/MEK/ERK mediated tumorigenic activity and targeting eIF4E should have a major impact on these pathways in human cancer. The function of eIF4E is modulated through phosphorylation of a conserved serine (Ser209) by Mnk1 and Mnk2 downstream of ERK. While the phosphorylation event is necessary for oncogenic transformation, it seems to be dispensable for normal development. Hence, pharmacologic Mnk inhibitors may provide non-toxic and effective anti-cancer strategy. Strong circumstantial evidence indicates that Mnk inhibition presents attractive therapeutic potential, but the lack of selective Mnk inhibitors has so far confounded pharmacological target validation and clinical development.
Collapse
Affiliation(s)
- Jinqiang Hou
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | | | | | | |
Collapse
|
34
|
Joshi S, Platanias LC. Mnk Kinases in Cytokine Signaling and Regulation of Cytokine Responses. Biomol Concepts 2012; 3:255-266. [PMID: 23710261 DOI: 10.1515/bmc-2011-0057] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The kinases Mnk1 and Mnk2 are activated downstream of the p38 MAPK and MEK/ERK signaling pathways. Extensive work over the years has shown that these kinases control phosphorylation of the eukaryotic initiation factor 4E (eIF4E) and regulate engagement of other effector elements, including hnRNPA1 and PSF. Mnk kinases are ubiquitously expressed and play critical roles in signaling for various cytokine receptors, while there is emerging evidence that they have important functions as mediators of pro-inflammatory cytokine production. In this review the mechanisms of activation of MNK pathways by cytokine receptors are addressed and their roles in diverse cytokine-dependent biological processes are reviewed. The clinical-translational implications of such work and the relevance of future development of specific MNK inhibitors for the treatment of malignancies and auto-immune disorders are discussed.
Collapse
Affiliation(s)
- Sonali Joshi
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Northwestern University Medical School, and Jesse Brown VA, Medical Center, Chicago, IL ; Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
35
|
Joshi S, Platanias LC. Mnk Kinases in Cytokine Signaling and Regulation of Cytokine Responses. Biomol Concepts 2012. [PMID: 23710261 DOI: 10.1515/bmc-2011-1057] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The kinases Mnk1 and Mnk2 are activated downstream of the p38 MAPK and MEK/ERK signaling pathways. Extensive work over the years has shown that these kinases control phosphorylation of the eukaryotic initiation factor 4E (eIF4E) and regulate engagement of other effector elements, including hnRNPA1 and PSF. Mnk kinases are ubiquitously expressed and play critical roles in signaling for various cytokine receptors, while there is emerging evidence that they have important functions as mediators of pro-inflammatory cytokine production. In this review the mechanisms of activation of MNK pathways by cytokine receptors are addressed and their roles in diverse cytokine-dependent biological processes are reviewed. The clinical-translational implications of such work and the relevance of future development of specific MNK inhibitors for the treatment of malignancies and auto-immune disorders are discussed.
Collapse
Affiliation(s)
- Sonali Joshi
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Northwestern University Medical School, and Jesse Brown VA, Medical Center, Chicago, IL ; Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
36
|
Kang MK, Han SJ. Post-transcriptional and post-translational regulation during mouse oocyte maturation. BMB Rep 2011; 44:147-57. [DOI: 10.5483/bmbrep.2011.44.3.147] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
37
|
|
38
|
Shiga toxin 2 and flagellin from shiga-toxigenic Escherichia coli superinduce interleukin-8 through synergistic effects on host stress-activated protein kinase activation. Infect Immun 2010; 78:2984-94. [PMID: 20439475 DOI: 10.1128/iai.00383-10] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Shiga toxins expressed in the intestinal lumen during infection with Shiga-toxigenic Escherichia coli must translocate across the epithelium and enter the systemic circulation to cause systemic (pathological) effects, including hemolytic uremic syndrome. The transepithelial migration of polymorphonuclear leukocytes in response to chemokine expression by intestinal epithelial cells is thought to promote uptake of Stx from the intestinal lumen by compromising the epithelial barrier. In the present study, we investigated the hypothesis that flagellin acts in conjunction with Shiga toxin to augment this chemokine expression. We investigated the relative contributions of nuclear factor kappaB (NF-kappaB) and mitogen-activated protein kinase (MAPK) signaling to transcription and translation of interleukin-8. Using reporter gene constructs, we showed that flagellin-mediated interleukin-8 gene transcription is heavily dependent on both NF-kappaB and extracellular signal-regulated kinase 1 and 2 (ERK-1/2) activation. In contrast, inhibition of p38 has no detectable effect on interleukin-8 gene transcription, even though flagellin-mediated activation of host p38 is critical for maximal interleukin-8 protein expression. Inhibition of MAPK-interacting kinase 1 suggests that p38 signaling affects the posttranscriptional regulation of interleukin-8 protein expression induced by flagellin. Cotreatment with Stx2 and flagellin results in a synergistic upregulation of c-Jun N-terminal protein kinases (JNKs), p38 activation, and a superinduction of interleukin-8 mRNA. This synergism was also evident at the protein level, with increased interleukin-8 protein detectable following cotreatment with flagellin and Stx2. We propose that flagellin, in conjunction with Shiga toxin, synergistically upregulates stress-activated protein kinases, resulting in superinduction of interleukin-8 and, ultimately, absorption of Stx into the systemic circulation.
Collapse
|
39
|
Van Der Kelen K, Beyaert R, Inzé D, De Veylder L. Translational control of eukaryotic gene expression. Crit Rev Biochem Mol Biol 2009; 44:143-68. [PMID: 19604130 DOI: 10.1080/10409230902882090] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Translational control mechanisms are, besides transcriptional control and mRNA stability, the most determining for final protein levels. A large number of accessory factors that assist the ribosome during initiation, elongation, and termination of translation are required for protein synthesis. Cap-dependent translational control occurs mainly during the initiation step, involving eukaryotic initiation factors (eIFs) and accessory proteins. Initiation is affected by various stimuli that influence the phosphorylation status of both eIF4E and eIF2 and through binding of 4E-binding proteins to eIF4E, which finally inhibits cap- dependent translation. Under conditions where cap-dependent translation is hampered, translation of transcripts containing an internal ribosome entry site can still be supported in a cap-independent manner. An interesting example of translational control is the switch between cap-independent and cap-dependent translation during the eukaryotic cell cycle. At the G1-to-S transition, translation occurs predominantly in a cap-dependent manner, while during the G2-to-M transition, cap-dependent translation is inhibited and transcripts are predominantly translated through a cap-independent mechanism.
Collapse
|
40
|
Son DO, Satsu H, Kiso Y, Totsuka M, Shimizu M. Inhibitory effect of carnosine on interleukin-8 production in intestinal epithelial cells through translational regulation. Cytokine 2008; 42:265-276. [DOI: 10.1016/j.cyto.2008.02.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 01/22/2008] [Accepted: 02/19/2008] [Indexed: 10/22/2022]
|
41
|
Dolniak B, Katsoulidis E, Carayol N, Altman JK, Redig AJ, Tallman MS, Ueda T, Watanabe-Fukunaga R, Fukunaga R, Platanias LC. Regulation of arsenic trioxide-induced cellular responses by Mnk1 and Mnk2. J Biol Chem 2008; 283:12034-42. [PMID: 18299328 DOI: 10.1074/jbc.m708816200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Arsenic trioxide (As(2)O(3)) is a potent inducer of apoptosis of malignant cells in vitro and in vivo, but the precise mechanisms by which it mediates such effects are not well defined. We provide evidence that As(2)O(3) induces phosphorylation/activation of the MAPK signal-integrating kinases (Mnks) 1 and 2 in leukemia cell lines. Such activation is defective in cells with targeted disruption of the p38alpha MAPK gene, indicating that it requires upstream engagement of the p38 MAPK pathway. Studies using Mnk1(-/-) or Mnk2(-/-), or double Mnk1(-/-)Mnk2(-/-) knock-out cells, establish that activation of Mnk1 and Mnk2 by arsenic trioxide regulates downstream phosphorylation of the eukaryotic initiation factor 4E at Ser-209. Importantly, arsenic-induced apoptosis is enhanced in cells with targeted disruption of the Mnk1 and/or Mnk2 genes, suggesting that these kinases are activated in a negative-feedback regulatory manner, to control generation of arsenic trioxide responses. Consistent with this, pharmacological inhibition of Mnk activity enhances the suppressive effects of arsenic trioxide on primary leukemic progenitors from patients with acute leukemias. Taken together, these findings indicate an important role for Mnk kinases, acting as negative regulators for signals that control generation of arsenic trioxide-dependent apoptosis and antileukemic responses.
Collapse
Affiliation(s)
- Blazej Dolniak
- Robert H. Lurie Comprehensive Cancer Center and Division of Hematology-Oncology, Department of Medicine, Northwestern University Medical School, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kjellerup RB, Kragballe K, Iversen L, Johansen C. Pro-inflammatory cytokine release in keratinocytes is mediated through the MAPK signal-integrating kinases. Exp Dermatol 2007; 17:498-504. [PMID: 18081851 DOI: 10.1111/j.1600-0625.2007.00672.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The mitogen-activated protein kinases (MAPKs) are known to play a key role in the regulation of cytokine expression in several cell types. MAPK signal-integrating kinase 1 (Mnk1) is a kinase activated through both the stress- and cytokine-activated p38 MAPK pathway and the classical extracellular signal-regulated kinase 1/2 (ERK1/2) pathway. In this study, we demonstrate that in cultured normal human keratinocytes Mnk1 and its downstream target eukaryotic initiation factor 4E (eIF4E) are phosphorylated in a time-dependent manner in response to stimulation with anisomycin or interleukin (IL)-1beta. Both the stimuli are well-recognized activators of the p38 MAPK pathway. Furthermore, we show that the Mnk inhibitor CGP57380 is capable of inhibiting the phosphorylation of eIF4E in keratinocytes, and that the abolishment of eIF4E phosphorylation dramatically decreases the anisomycin-induced protein release of the pro-inflammatory cytokines tumor necrosis factor-alpha (TNF-alpha), IL-1beta and IL-6 as well as the IL-1beta-induced protein release of TNF-alpha. Therefore, we propose that Mnk1 might contribute to the expression of pro-inflammatory cytokines found in inflammatory skin diseases.
Collapse
|
43
|
O'Loghlen A, González VM, Jurado T, Salinas M, Martín ME. Characterization of the activity of human MAP kinase-interacting kinase Mnk1b. BIOCHIMICA ET BIOPHYSICA ACTA 2007; 1773:1416-27. [PMID: 17590453 DOI: 10.1016/j.bbamcr.2007.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Revised: 05/18/2007] [Accepted: 05/21/2007] [Indexed: 02/08/2023]
Abstract
Human mitogen-activated protein (MAP) kinase interacting kinase 1b (Mnk1b) is a splice variant of human Mnk1a, which has been identified in our laboratory [A. O'Loghlen, V.M. Gonzalez, D. Pineiro, M.I. Perez-Morgado, M. Salinas, M.E. Martin, Identification and molecular characterization of Mnk1b, a splice variant of human MAP kinase-interacting kinase Mnk1, Exp. Cell Res. 299 (2004) 343-355]. Mnk1b has much higher basal eIF4E kinase activity than Mnk1a. Because Mnk1b presents different features in its C-terminus with respect to Mnk1a, we have studied in this paper the potential role of these structural differences in determining the higher basal eIF4E kinase activity as well as the subcellular localization of Mnk1b. In this paper, we demonstrate that phosphorylation of the Thr209 and Thr214 in the activation loop of Mnk1b is necessary for its activation. However, the different kinase activity between Mnk1a and Mnk1b is independent of the phosphorylation status of the activation loop residues. By deletion of the C-terminal tail in Mnk1a, we confirmed that the absence of this sequence is not responsible for the higher eIF4E kinase activity present in Mnk1b. Moreover, our findings support a crucial role of the 12 amino acids, particularly the Ala344, in the C-terminal specific region of Mnk1b (Mnk1bSR), on the kinase activity of the protein.
Collapse
Affiliation(s)
- Ana O'Loghlen
- Servicio de Bioquímica-Investigación, Hospital Ramón y Cajal, Ctra. Colmenar km. 9,100. E-28034 Madrid, Spain
| | | | | | | | | |
Collapse
|
44
|
Abstract
The translational machinery of mammalian cells is regulated through the phosphorylation of a number of its components, especially translation factor proteins. These include factors involved in the initiation and elongation stages of translation, and proteins that modify their activity. Examples include eukaryotic initiation factor (eIF) 4E, eukaryotic elongation factor (eEF) 2, and eIF4E-binding protein 1 (4E-BP1). Their phosphorylation is mediated by protein kinases that, in turn, are regulated by specific intracellular signaling pathways. These pathways include those mediated via the mammalian target of rapamycin (mTOR), the ERK and p38 MAP kinase pathways, and protein kinase B (Akt). These pathways are activated by hormones (e.g., insulin), growth factors, mitogens, and other extracellular stimuli. In some cases, amino acids also modulate the pathway (e.g., mTOR). Procedures are described for determining the states of phosphorylation and/or activity of several translation factors, and of kinases that phosphorylate them. We also outline procedures for assessing the states of activation of relevant signaling pathways. In addition, we provide guidelines on using small molecule inhibitors to assess the involvement of specific signaling pathways in controlling translation factors and protein synthesis.
Collapse
|
45
|
Chrestensen CA, Shuman JK, Eschenroeder A, Worthington M, Gram H, Sturgill TW. MNK1 and MNK2 regulation in HER2-overexpressing breast cancer lines. J Biol Chem 2006; 282:4243-4252. [PMID: 17130135 DOI: 10.1074/jbc.m607368200] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
MAPK-interacting protein kinases 1 and 2 (MNK1 and MNK2) function downstream of p38 and ERK MAPK, but there are large gaps in our knowledge of how MNKs are regulated and function. As proteins activated in the HER2/Ras/Raf/ERK pathway, the MNKs are of potential interest in HER2-overexpressing cancers. We utilized a panel of breast cell lines (HCC1419, AU565, SKBR3, MCF7, and MCF10A), three of which overexpress HER2, to characterize the amounts and activation status of MNKs and other pathway enzymes (ERKs and RSKs) in these cells. We generated a phosphospecific antibody to Thr(P)-214 in the T-loop of MNKs and found that phosphorylations of both Thr-209 and Thr-214 in human MNK1 are required for activation. Increased phosphorylation and activity of the MNKs correlate with HER2 overexpression, and inhibition of the MNKs reduces colony formation in soft agar. Our work identifies the MNKs as potential therapeutic targets for breast cancer treatments.
Collapse
Affiliation(s)
- Carol A Chrestensen
- Department of Pharmacology, University of Virginia Health Sciences Center, Charlottesville, Virginia 22908
| | - Jacquelyn K Shuman
- Department of Pharmacology, University of Virginia Health Sciences Center, Charlottesville, Virginia 22908
| | - Andrew Eschenroeder
- Department of Pharmacology, University of Virginia Health Sciences Center, Charlottesville, Virginia 22908
| | - Mark Worthington
- Digestive Health Center of Excellence, University of Virginia Health Sciences Center, Charlottesville, Virginia 22908, and
| | - Hermann Gram
- Arthritis and Bone Metabolism, Novartis Pharma AG, CH-4002 Basel, Switzerland
| | - Thomas W Sturgill
- Department of Pharmacology, University of Virginia Health Sciences Center, Charlottesville, Virginia 22908.
| |
Collapse
|
46
|
Ross G, Dyer JR, Castellucci VF, Sossin WS. Mnk is a negative regulator of cap-dependent translation in Aplysia neurons. J Neurochem 2006; 97:79-91. [PMID: 16515558 DOI: 10.1111/j.1471-4159.2006.03704.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To investigate the mechanisms underlying regulation of eukaryotic initiation factor 4E (eIF4E) phosphorylation in Aplysia neurons, we have cloned the Aplysia homolog of the vertebrate eIF4E kinases, Mnk1 and -2. Aplysia Mnk shares many conserved regions with vertebrate Mnk, including putative eukaryotic initiation factor 4G binding regions, activation loop phosphorylation sites, and a carboxy-terminal anchoring site for MAP kinases. As expected, purified Aplysia Mnk phosphorylated Aplysia eIF4E at a conserved carboxy-terminal serine and over-expression of Aplysia Mnk in sensory neurons led to increased phosphorylation of endogenous eIF4E. Over-expression of Aplysia Mnk led to strong decreases in cap-dependent translation, while generally sparing internal ribosomal entry site (IRES)-dependent translation. However, decreases in cap-dependent translation seen after expression of Aplysia Mnk could only be partly explained by increases in eIF4E phosphorylation. In Aplysia sensory neurons, phosphorylation of eIF4E is reduced during intermediate memory formation. However, we found that this physiological regulation of eIF4E phosphorylation was independent of changes in Aplysia Mnk phosphorylation. We propose that changes in eIF4E phosphorylation in Aplysia neurons are a consequence of changes in cap-dependent translation that are independent of regulation of Aplysia Mnk.
Collapse
Affiliation(s)
- Gabriel Ross
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
47
|
Wollenhaupt K, Dänicke S, Brüssow KP, Tiemann U. In vitro and in vivo effects of deoxynivalenol (DNV) on regulators of cap dependent translation control in porcine endometrium. Reprod Toxicol 2006; 21:60-73. [PMID: 16099139 DOI: 10.1016/j.reprotox.2005.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2005] [Revised: 06/07/2005] [Accepted: 06/10/2005] [Indexed: 11/17/2022]
Abstract
Deoxynivalenol (DNV) is the most frequently encountered trichothecene in grain-based foods, and is able to produce toxic effects resulting in various diseases in farm and laboratory animals. The molecular mechanisms that control this mycotoxin mediated effects in porcine endometrial cells are far from being completely understood. Recent results show that DNV inhibits protein synthesis in actively proliferating tissues. Therefore, the present study investigated the effects of this mycotoxin on a cellular level in an in vivo and in vitro system. The abundance and phosphorylation state (activity) of the cell cycle dependent kinases MAPk and Akt (PKB) and their potential targets eIF-4E (eukaryotic initiation factor 4E) and 4E-BP1 (4E binding protein, eIF4E repressor protein) were examined. In previous investigations it was found that these factors are involved in initiation of mRNA translation. The results show that DNV in vitro strongly reduce the abundance of p38 MAPk, protein kinase Akt and the alpha- and beta-4E-BP1 bands. The phosphorylation state of these proteins was obviously not modulated. In contrast, the eIF4E phosphorylation was strongly reduced in DNV treated cells. In summary, our in vitro results let assume that DNV potentially influences gene expression, but this work does not present a direct proof that DNV alters processes, which are involved in the initiation of mRNA translation. Surprisingly in vivo, an influence of DNV feeding on the investigated molecular events could not be demonstrated.
Collapse
Affiliation(s)
- K Wollenhaupt
- Unit of Reproductive Biology, Research Institute for the Biology of Farm Animals, 18196 Dummerstorf, Germany.
| | | | | | | |
Collapse
|
48
|
Worch J, Tickenbrock L, Schwäble J, Steffen B, Cauvet T, Mlody B, Buerger H, Koeffler HP, Berdel WE, Serve H, Müller-Tidow C. The serine-threonine kinase MNK1 is post-translationally stabilized by PML-RARalpha and regulates differentiation of hematopoietic cells. Oncogene 2005; 23:9162-72. [PMID: 15516979 DOI: 10.1038/sj.onc.1208164] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Microarray analyses were performed to identify target genes that are shared by the acute myeloid leukemia (AML) translocation products PML-RARalpha, PLZF-RARalpha and AML1-ETO in inducibly transfected U937 cell lines. The cytoplasmic serine and threonine kinase MNK1 was identified as one of the target genes. At the protein level, MNK1 was significantly induced by each of the three fusion proteins. Protein half-life analyses showed that PML-RARalpha enhanced MNK1 protein stability in U937 cells and ATRA exposure decreased MNK1 half-life in NB4 cells. EIF4E, the main MNK1 substrate, plays a role in the pathogenesis of a variety of cancers. Upon MNK1 overexpression, eIF4E phosphorylation increased as a sign of functional activation. Interestingly, MNK1 protein expression decreased during myeloid differentiation. Inhibition of MNK1 activity by a specific inhibitor (CGP57380) enhanced differentiation of HL60 and 32D cells, further suggesting a role for MNK1 in the myeloid differentiation. In addition, kinase dead mutants of MNK1 significantly impaired proliferation of 32D cells. Immunohistochemistry of primary AML bone marrow biopsies showed strong cytoplasmic MNK1 expression in 25 of 99 AML specimens (25%). MNK1 expression was associated with high levels of c-myc expression. Taken together, we identified MNK1 as a target gene of several leukemogenic fusion proteins in AML. MNK1 plays a role in myeloid differentiation. These data suggest a role for MNK1 in the AML fusion protein-associated differentiation block.
Collapse
Affiliation(s)
- Jennifer Worch
- Department of Medicine, Hematology and Oncology, University of Münster, Domagkstr. 3, 48129 Münster, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Feliers D, Duraisamy S, Barnes JL, Ghosh-Choudhury G, Kasinath BS. Translational regulation of vascular endothelial growth factor expression in renal epithelial cells by angiotensin II. Am J Physiol Renal Physiol 2005; 288:F521-9. [PMID: 15572520 DOI: 10.1152/ajprenal.00271.2004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ANG II regulates growth factor expression in the kidney. We investigated whether ANG II regulated vascular endothelial growth factor (VEGF) synthesis in proximal tubular epithelial (MCT) cells. ANG II (1 nM) increased VEGF protein expression within 5 min, the effect lasting for 30 min. There was no change in VEGF mRNA levels or mRNA stability, and transcription inhibitors did not affect ANG II-induced VEGF expression. Regulation of VEGF translation was investigated. Polyribosomal analysis revealed selective enrichment of heavy ribosomes (polysomes) with VEGF mRNA transcripts compared with light ribosomes in ANG II-treated cells, although distribution of GAPDH was unaltered. In vitro translation of total RNA from polysomal fractions showed selective increase in VEGF protein synthesis in ANG II-treated cells. Preincubation with LY-294002, a PI 3-kinase inhibitor, or expression of dominant-negative Akt prevented ANG II-stimulated increase in VEGF translation. ANG II increased phosphorylation of eukaryotic initiation factor 4E and its binding protein 4E-BP1, critical events that regulate the initiation phase of protein translation. ANG II failed to increase VEGF mRNA translation in cells stably expressing the phosphorylation mutant of 4E-BP1. Our data illustrate that a rapid increase in VEGF protein expression by ANG II is regulated at the initiation phase of translation of VEGF mRNA in renal epithelial cells. Regulation of VEGF translation by ANG II represents a novel pathway of renal response to injury.
Collapse
Affiliation(s)
- Denis Feliers
- Dept. of Medicine, Mail Code 7882, Univ. of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX 78229-3900, USA
| | | | | | | | | |
Collapse
|
50
|
Colpoys WE, Cochran BH, Carducci TM, Thorpe CM. Shiga toxins activate translational regulation pathways in intestinal epithelial cells. Cell Signal 2004; 17:891-9. [PMID: 15763431 DOI: 10.1016/j.cellsig.2004.11.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2004] [Revised: 11/03/2004] [Accepted: 11/03/2004] [Indexed: 11/20/2022]
Abstract
Shiga toxins (Stxs) cause irreversible damage to eukaryotic ribosomes, yet cellular intoxication of intestinal epithelial cells (IECs) results in increased synthesis of selected proteins, notably cytokines. How mRNA translation is maintained in this circumstance is unclear. This study was designed to assess whether Stx-induced alterations in host signal transduction machinery permit translation despite protein synthesis inhibition. A key step of translation is recruitment of initiation machinery to the 5' mRNA cap. This event occurs in part via interaction of the 5' cap with the cap binding protein, eIF4E, whose activity is positively regulated by phosphorylation and negatively regulated by binding to the translational repressor 4E-BP1. Following Stx treatment of IECs, eIF4E phosphorylation was detected by Western blotting using phospho-specific antibodies. Treatment with the p38 inhibitor, SB202190, or either of the ERK1/2 inhibitors, PD98059 and U0126, partially blocked Stx1-induced eIF4E phosphorylation. The Mnk1 inhibitor, CGP57380, blocked both basal and Stx-induced eIF4E phosphorylation. Interestingly, pretreatment with CGP57380 did not alter basal protein synthesis, but diminished the ability of cells to maintain translation following Stx1 challenge. Stx1 also induced hyperphosphorylation of 4E-BP1 and phosphorylation of S6Kinase; both effects were blocked by rapamycin. These data are novel observations showing that Stxs regulate multiple signal transduction pathways controlling translation in host cells, and support a role for eIF4E phosphorylation in maintaining host cell translation despite ribosomal intoxication.
Collapse
Affiliation(s)
- W E Colpoys
- Division of Geographic Medicine and Infectious Diseases, 750 Washington Street Box 041, Boston, MA 02111, USA
| | | | | | | |
Collapse
|