Translation Initiation Factor Eif4f Modifies the Dexamethasone Response in Multiple Myeloma

Total Page:16

File Type:pdf, Size:1020Kb

Translation Initiation Factor Eif4f Modifies the Dexamethasone Response in Multiple Myeloma Translation initiation factor eIF4F modifies the dexamethasone response in multiple myeloma Francis Roberta, William Romanb, Alexandre Bramoulléc, Christof Fellmannd, Anne Roulstonc, Chaim Shustikb, John A. Porco, Jr.e, Gordon C. Shorea,c, Michael Sebagb,1, and Jerry Pelletiera,c,f,1 Departments of aBiochemistry, bMedicine and Hematology, and fOncology, and cThe Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, QC, Canada H3G 1Y6; dMirimus, Inc., Cold Spring Harbor, NY 11724; and eCenter for Chemical Methodology and Library Development, Boston University, Boston, MA 02215 Edited* by David E. Housman, Massachusetts Institute of Technology, Cambridge, MA, and approved August 5, 2014 (received for review February 24, 2014) Enhanced protein synthesis capacity is associated with increased PI3K/mTOR and MAPK pathways intimately impinge on the tumor cell survival, proliferation, and resistance to chemother- translation pathway, linking this energetically demanding process apy. Cancers like multiple myeloma (MM), which display elevated to intra- and extracellular proliferation and survival cues (4). As activity in key translation regulatory nodes, such as the PI3K/ well, overexpression of MYC, a master regulator of ribosome mammalian target of rapamycin and MYC-eukaryotic initiation biogenesis and translation, is frequently observed in MM (5–7) factor (eIF) 4E pathways, are predicted to be particularly sensitive and is associated with a poor prognosis (7, 8). Deregulated to therapeutic strategies that target this process. To identify novel translational control is therefore a central feature of MM, with vulnerabilities in MM, we undertook a focused RNAi screen in perturbations occurring at the level of core components and which components of the translation apparatus were targeted. regulatory pathways. Our screen was designed to identify synthetic lethal relationships One of the best-studied MYC effectors and a downstream between translation factors or regulators and dexamethasone target of the PI3K/mTOR pathway implicated in translational (DEX), a corticosteroid used as frontline therapy in this disease. We control is eIF4F (4, 5, 9). eIF4F is a heterotrimeric complex find that suppression of all three subunits of the eIF4F cap-binding consisting of: (i) eIF4E, a cap-binding protein; (ii) eIF4A, an complex synergizes with DEX in MM to induce cell death. Using RNA helicase implicated in remodeling mRNA structure; and GENETICS a suite of small molecules that target various activities of eIF4F, we (iii) eIF4G, a large scaffolding protein. The association of eIF4E observed that cell survival and DEX resistance are attenuated with the eIF4F complex is regulated by mTOR, with mTOR upon eIF4F inhibition in MM cell lines and primary human samples. activation leading to stimulation of eIF4F formation. Elevated Levels of MYC and myeloid cell leukemia 1, two known eIF4F- PI3K/mTOR signaling flux or increased MYC levels exert pro- responsive transcripts and key survival factors in MM, were found activating effects on cap-dependent translation, and con- reduced upon eIF4F inhibition, and their independent suppres- sequently on cellular proliferation and survival (10). sion also synergized with DEX. Inhibition of eIF4F in MM exerts The PI3K/mTOR axis is being probed as a therapeutic target pleotropic effects unraveling a unique therapeutic opportunity. in MM (11). MM cells are sensitive to mTOR inhibition by rapa- mycin-related molecules (12), but the presence of an mTOR- silvestrol | hippuristanol | eIF4A | RNAi screening S6K-insulin receptor substrate-1 negative-feedback loop, which reactivates PI3K and AKT upon mTOR inhibition, diminishes ultiple myeloma (MM) is a bone marrow-derived malig- the efficacy of rapalogs (13). Second-generation mTOR complex Mnancy of plasma cells that typically produce a monoclonal (mTORC) kinase inhibitors (KIs) (e.g., OSI-027) and dual-spec- immunoglobulin (Ig). It is the second most frequent hema- ificity PI3K/mTOR KIs [e.g., (NVP)-BEZ235] avoid PI3K/AKT tological neoplasm in adults, with ∼20,000 newly diagnosed cases annually in the United States. Corticosteroids [predni- Significance sone and dexamethasone (DEX)] are common to all treat- ment regimens, usually in combination with an alkylating Multiple myeloma (MM) is a cancer that develops in the bone agent (e.g., melphalan) or more recently introduced agents, marrow and remains incurable to this day. It is a cancer type such as proteasome inhibitors (bortezomib and carfilzomib) that shows hallmarks of deregulated protein synthesis control. and immunomodulatory agents (thalidomide, lenalidomide, and To uncover new vulnerabilities in this disease, we performed pomalidomide). Despite the single-agent activity of these novel a focused RNAi screen to identify components of the trans- treatment options, enhanced clinical activity is observed when lation apparatus that, when depleted, would sensitize tumor they are used in combination with DEX. cells to dexamethasone (DEX), a component of frontline ther- The MM genomic landscape includes recurrent DNA trans- apy in this cancer. We found that suppression of eukaryotic locations involving mostly the IgH locus, chromosomal gains and initiation factor 4F, a heterotrimeric complex required for cap- losses, and a significant number of mutations in genes involved in dependent translation initiation, is a modifier of the DEX re- translation and its regulation (1). In nearly 50% of patients an- sponse in MM. Our efforts uncover a previously unidentified alyzed, mutations were documented in the following: DIS3, vulnerability in MM that should be explored clinically. a component of the exosome and predicted to increase mRNA content; FAM46C, a gene product functionally related to a reg- Author contributions: F.R., A.R., C.S., G.C.S., M.S., and J.P. designed research; F.R., W.R., ulator of protein synthesis; XBP1, an unfolded response protein and M.S. performed research; A.B., C.F., and J.A.P. contributed new reagents/analytic tools; F.R., W.R., A.B., C.F., A.R., C.S., J.A.P., G.C.S., M.S., and J.P. analyzed data; and F.R. linked to translational control; LRRK2, a eukaryotic initiation and J.P. wrote the paper. factor (eIF) 4E binding protein kinase; and, less frequently, Conflict of interest statement: C.F. is a founder and employee of Mirimus, Inc., a company eIF3B, rpL10, and rpS6KA1 (1). In addition, several signaling that has licensed shRNA technology based on the mir30 system used in this report. pathways are aberrantly activated in MM. These signaling *This Direct Submission article had a prearranged editor. pathways include the PI3K/mammalian target of rapamycin 1To whom correspondence may be addressed. Email: [email protected] or jerry. (mTOR), NF-κB, Ras, Raf, MAPK, and Janus kinase pathways, [email protected]. all of which promote proliferation, evasion of apoptosis, and This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. resistance to therapy (2, 3). Among these signaling pathways, the 1073/pnas.1402650111/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1402650111 PNAS Early Edition | 1of6 Downloaded by guest on September 27, 2021 activation via the aforementioned feedback loop and show higher Because DEX is used as frontline therapy in this disease, we activity against MM cells than rapalogs (11, 14–16). Rapalogs and designed the screen to identify potential modifiers of the DEX PI3K/mTOR inhibitors have also been shown to enhance the response (Fig. 1A). Among five human MM cell lines tested for cytotoxic effects of DEX (14, 17, 18), which is an effect that can their ability to be infected by our modified pGmP lentivirus, JJN- also be obtained by sequestering eIF4E from the eIF4F complex 3, and KMS-11 were the most efficiently and reproducibly by overexpressing an inhibitory binding partner (18). Herein, we infected, with rates attaining 50–60% following one round of describe the results of a focused RNAi-based screen aimed at infection (Fig. S1A). Whereas KMS-11 cells were sensitive to identifying druggable targets among components of the trans- DEX (IC50 = 50 nM), JJN-3 cells were quite resistant to con- lation apparatus to identify DEX synthetic lethal partners in centrations as high as 3 μM(Fig. S1B), as previously reported MM. Our results define eIF4F as a target for the treatment of (19). Upon exposure to DEX, JJN-3 cells activated expression MM, identify a small-molecule inhibitor with low nanomolar of the DEX-responsive glucocorticoid-induced leucine zipper potency against human MM cells, and demonstrate that in- (GILZ) (20), indicating that DEX resistance in JJN-3 cells is hibition of translation is synthetic lethal with DEX when applied not due to defective glucocorticoid receptor (GR) signaling to MM tumor cells. (Fig. S1C). GILZ induction is mediated by DEX in our setting because it is blocked by the antagonist RU-486 (Fig. S1C). Results These results establish JJN-3 cells as a DEX-resistant cell line RNAi-Based Synthetic Lethal Screen Identifies Modifiers of DEX with a functional GR/GILZ signaling axis. Sensitivity in MM. Given the profound deregulation at the level We generated a custom, sequence-verified and arrayed, miR30- of translation that has been documented in MM (Introduction), we based shRNA library targeting amino acyl-tRNA synthetases, large chose to perform an RNAi-based screen targeting this pathway and small ribosomal proteins, initiation factors, elongation fac- to identify vulnerabilities for potential therapeutic intervention. tors, termination factors, RNA helicases, and components known Fig. 1. DEX-dependent synthetic lethal RNAi screen in the JJN-3 cell line. (A) Schematic outline of the DEX-dependent synthetic lethal screen performed in this study. A diagram of the pGmP vector and a time line of the infection and cell-harvesting schedule are presented. Puro, puromycin; SIN, self-inactivating. (B) Categories of genes evaluated in the RNAi screen. The number of genes targeted in each category is indicated in parentheses. DHX, DEAH-box. (C) Pooled synthetic lethal shRNA screen in JJN-3 cells showing changes in overall representation of 1,099 informative shRNAs during 12 d of culture.
Recommended publications
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • A Chemical-Genetic Screen for Identifying Substrates of the Er Kinase Perk
    University of Pennsylvania ScholarlyCommons Publicly Accessible Penn Dissertations 2014 A Chemical-Genetic Screen for Identifying Substrates of the Er Kinase Perk Nancy L. Maas University of Pennsylvania, [email protected] Follow this and additional works at: https://repository.upenn.edu/edissertations Part of the Biology Commons, Cell Biology Commons, and the Molecular Biology Commons Recommended Citation Maas, Nancy L., "A Chemical-Genetic Screen for Identifying Substrates of the Er Kinase Perk" (2014). Publicly Accessible Penn Dissertations. 1354. https://repository.upenn.edu/edissertations/1354 This paper is posted at ScholarlyCommons. https://repository.upenn.edu/edissertations/1354 For more information, please contact [email protected]. A Chemical-Genetic Screen for Identifying Substrates of the Er Kinase Perk Abstract Cells constantly encounter changing environments that challenge the ability to adapt and survive. Signal transduction networks enable cells to appropriately sense and respond to these changes, and are often mediated through the activity of protein kinases. Protein kinases are a class of enzyme responsible for regulating a broad spectrum of cellular functions by transferring phosphate groups from ATP to substrate proteins, thereby altering substrate activity and function. PERK is a resident kinase of the endoplasmic reticulum, and is responsible for sensing perturbations in the protein folding capacity of the ER. When the influx of unfolded, nascent proteins exceeds the folding capacity of the ER, PERK initiates a cascade of signaling events that enable cell adaptation and ER stress resolution. These signaling pathways are not only essential for the survival of normal cells undergoing ER stress, but are also co-opted by tumor cells in order to survive the oxygen and nutrient-restricted conditions of the tumor microenvironment.
    [Show full text]
  • Systematically Profiling the Expression of Eif3 Subunits in Glioma Reveals
    Chai et al. Cancer Cell Int (2019) 19:155 https://doi.org/10.1186/s12935-019-0867-1 Cancer Cell International PRIMARY RESEARCH Open Access Systematically profling the expression of eIF3 subunits in glioma reveals the expression of eIF3i has prognostic value in IDH-mutant lower grade glioma Rui‑Chao Chai1,4,6†, Ning Wang2†, Yu‑Zhou Chang3, Ke‑Nan Zhang1,6, Jing‑Jun Li1,6, Jun‑Jie Niu5, Fan Wu1,6*, Yu‑Qing Liu1,6* and Yong‑Zhi Wang1,3,4,6* Abstract Background: Abnormal expression of the eukaryotic initiation factor 3 (eIF3) subunits plays critical roles in tumo‑ rigenesis and progression, and also has potential prognostic value in cancers. However, the expression and clinical implications of eIF3 subunits in glioma remain unknown. Methods: Expression data of eIF3 for patients with gliomas were obtained from the Chinese Glioma Genome Atlas (CGGA) (n 272) and The Cancer Genome Atlas (TCGA) (n 595). Cox regression, the receiver operating characteristic (ROC) curves= and Kaplan–Meier analysis were used to study= the prognostic value. Gene oncology (GO) and gene set enrichment analysis (GSEA) were utilized for functional prediction. Results: In both the CGGA and TCGA datasets, the expression levels of eIF3d, eIF3e, eIF3f, eIF3h and eIF3l highly were associated with the IDH mutant status of gliomas. The expression of eIF3b, eIF3i, eIF3k and eIF3m was increased with the tumor grade, and was associated with poorer overall survival [All Hazard ratio (HR) > 1 and P < 0.05]. By contrast, the expression of eIF3a and eIF3l was decreased in higher grade gliomas and was associated with better overall sur‑ vival (Both HR < 1 and P < 0.05).
    [Show full text]
  • The J-Subunit of Human Translation Initiation Factor Eif3 Is Required for the Stable Binding of Eif3 and Its Subcomplexes to 40 S Ribosomal Subunits in Vitro*
    THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 279, No. 10, Issue of March 5, pp. 8946–8956, 2004 © 2004 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A. The j-Subunit of Human Translation Initiation Factor eIF3 Is Required for the Stable Binding of eIF3 and Its Subcomplexes to 40 S Ribosomal Subunits in Vitro* Received for publication, November 21, 2003, and in revised form, December 18, 2003 Published, JBC Papers in Press, December 19, 2003, DOI 10.1074/jbc.M312745200 Christopher S. Fraser‡, Jennifer Y. Lee, Greg L. Mayeur, Martin Bushell§, Jennifer A. Doudna¶, and John W. B. Hersheyʈ From the Department of Biological Chemistry, School of Medicine, University of California, Davis, California 95616 Eukaryotic initiation factor 3 (eIF3) is a 12-subunit protein subunits, named in order of decreasing molecular protein complex that plays a central role in binding of weight as recommended (4): eIF3a, eIF3b, eIF3c, eIF3d, eIF3l, initiator methionyl-tRNA and mRNA to the 40 S riboso- eIF3e, eIF3f, eIF3g, eIF3h, eIF3i, eIF3j, and eIF3k (5, 6). Spe- mal subunit to form the 40 S initiation complex. The cific functions for mammalian eIF3 have been identified by a molecular mechanisms by which eIF3 exerts these func- variety of in vitro experiments. It binds directly to 40 S ribo- tions are poorly understood. To learn more about the somal subunits in the absence of other initiation components structure and function of eIF3 we have expressed and (1), and affects the association/dissociation of ribosomes (7–10). purified individual human eIF3 subunits or complexes It promotes the binding of Met-tRNA and mRNA to the 40 S of eIF3 subunits using baculovirus-infected Sf9 cells.
    [Show full text]
  • Translation Initiation Factor Eif3h Targets Specific Transcripts To
    Translation initiation factor eIF3h targets specific transcripts to polysomes during embryogenesis Avik Choudhuria,b, Umadas Maitraa,1, and Todd Evansb,1 aDepartment of Developmental and Molecular Biology, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461; and bDepartment of Surgery, Weill Cornell Medical College, New York, NY 10065 Edited by Igor B. Dawid, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, and approved April 30, 2013 (received for review February 14, 2013) Eukaryotic translation initiation factor 3 (eIF3) plays a central role eukaryotes. These—eIF3d, eIF3e, eIF3f, eIF3h, eIF3j, eIF3k, in translation initiation and consists of five core (conserved) sub- eIF3l, and eIF3m—were designated “non-core” subunits (4). units present in both budding yeast and higher eukaryotes. Higher In contrast to the budding yeast, the genome of the fission eukaryotic eIF3 contains additional (noncore or nonconserved) yeast Schizosaccharomyces pombe contains structural homologs subunits of poorly defined function, including sub-unit h (eIF3h), of at least five noncore (nonconserved) eIF3 subunits—eIF3d, which in zebrafish is encoded by two distinct genes (eif3ha and eIF3e, eIF3f, eIF3h, and eIF3m. The gene encoding eIF3f is eif3hb). Previously we showed that eif3ha encodes the predominant essential for growth, whereas eIF3d, eIF3e, and eIF3h are dis- isoform during zebrafish embryogenesis and that depletion of this pensable for growth and viability (5–11). However, deleted factor causes defects in the development of the brain and eyes. To strains show specific phenotypes including defects in meiosis/ investigate the molecular mechanism governing this regulation, we sporulation (6, 9, 11).
    [Show full text]
  • Supplementary Material Contents
    Supplementary Material Contents Immune modulating proteins identified from exosomal samples.....................................................................2 Figure S1: Overlap between exosomal and soluble proteomes.................................................................................... 4 Bacterial strains:..............................................................................................................................................4 Figure S2: Variability between subjects of effects of exosomes on BL21-lux growth.................................................... 5 Figure S3: Early effects of exosomes on growth of BL21 E. coli .................................................................................... 5 Figure S4: Exosomal Lysis............................................................................................................................................ 6 Figure S5: Effect of pH on exosomal action.................................................................................................................. 7 Figure S6: Effect of exosomes on growth of UPEC (pH = 6.5) suspended in exosome-depleted urine supernatant ....... 8 Effective exosomal concentration....................................................................................................................8 Figure S7: Sample constitution for luminometry experiments..................................................................................... 8 Figure S8: Determining effective concentration .........................................................................................................
    [Show full text]
  • 1 1 2 Pharmacological Dimerization and Activation of the Exchange
    1 2 3 Pharmacological dimerization and activation of the exchange factor eIF2B antagonizes the 4 integrated stress response 5 6 7 *Carmela Sidrauski1,2, *Jordan C. Tsai1,2, Martin Kampmann2,3, Brian R. Hearn4, Punitha 8 Vedantham4, Priyadarshini Jaishankar4 , Masaaki Sokabe5, Aaron S. Mendez1,2, Billy W. 9 Newton6, Edward L. Tang6.7, Erik Verschueren6, Jeffrey R. Johnson6,7, Nevan J. Krogan6,7,, 10 Christopher S. Fraser5, Jonathan S. Weissman2,3, Adam R. Renslo4, and Peter Walter 1,2 11 12 1Department of Biochemistry and Biophysics, University of California, San Francisco, United 13 States 14 2Howard Hughes Medical Institute, University of California, San Francisco, United States 15 3Department of Cellular and Molecular Pharmacology, University of California, San Francisco, 16 United States 17 4Department of Pharmaceutical Chemistry and the Small Molecule Discovery Center, University 18 of California at San Francisco, United States 19 5Department of Molecular and Cellular Biology, College of Biological Sciences, University of 20 California, Davis, United States 21 6QB3, California Institute for Quantitative Biosciences, University of California, San Francisco, 22 United States 23 7Gladstone Institutes, San Francisco, United States 24 25 * Both authors contributed equally to this work 26 27 28 Abstract 29 30 The general translation initiation factor eIF2 is a major translational control point. Multiple 31 signaling pathways in the integrated stress response phosphorylate eIF2 serine-51, inhibiting 32 nucleotide exchange by eIF2B. ISRIB, a potent drug-like small molecule, renders cells 33 insensitive to eIF2α phosphorylation and enhances cognitive function in rodents by blocking 34 long-term depression. ISRIB was identified in a phenotypic cell-based screen, and its mechanism 35 of action remained unknown.
    [Show full text]
  • Relevance of Translation Initiation in Diffuse Glioma Biology and Its
    cells Review Relevance of Translation Initiation in Diffuse Glioma Biology and its Therapeutic Potential Digregorio Marina 1, Lombard Arnaud 1,2, Lumapat Paul Noel 1, Scholtes Felix 1,2, Rogister Bernard 1,3 and Coppieters Natacha 1,* 1 Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences Research Centre, University of Liège, 4000 Liège, Belgium; [email protected] (D.M.); [email protected] (L.A.); [email protected] (L.P.N.); [email protected] (S.F.); [email protected] (R.B.) 2 Department of Neurosurgery, CHU of Liège, 4000 Liège, Belgium 3 Department of Neurology, CHU of Liège, 4000 Liège, Belgium * Correspondence: [email protected] Received: 18 October 2019; Accepted: 26 November 2019; Published: 29 November 2019 Abstract: Cancer cells are continually exposed to environmental stressors forcing them to adapt their protein production to survive. The translational machinery can be recruited by malignant cells to synthesize proteins required to promote their survival, even in times of high physiological and pathological stress. This phenomenon has been described in several cancers including in gliomas. Abnormal regulation of translation has encouraged the development of new therapeutics targeting the protein synthesis pathway. This approach could be meaningful for glioma given the fact that the median survival following diagnosis of the highest grade of glioma remains short despite current therapy. The identification of new targets for the development of novel therapeutics is therefore needed in order to improve this devastating overall survival rate. This review discusses current literature on translation in gliomas with a focus on the initiation step covering both the cap-dependent and cap-independent modes of initiation.
    [Show full text]
  • Expression of Eukaryotic Translation Initiation Factor 3 Subunit B in Liver Cancer and Its Prognostic Significance
    436 EXPERIMENTAL AND THERAPEUTIC MEDICINE 20: 436-446, 2020 Expression of eukaryotic translation initiation factor 3 subunit B in liver cancer and its prognostic significance QING YUE1*, LINGYU MENG2*, BAOXING JIA2 and WEI HAN2 Departments of 1Oncology and 2Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China Received July 16, 2019; Accepted December 19, 2019 DOI: 10.3892/etm.2020.8726 Abstract. Liver cancer is one of the major malignancies with in high EIF3B expression and low EIF3B expression groups. the worst prognosis among all solid tumor types. It is therefore In conclusion, high EIF3B expression was indicated to be an ponderable to explore prognostic biomarkers and therapeutic independent prognostic biomarker for patients with liver cancer. targets for liver cancer. Eukaryotic translation initiation factor 3 subunit B (EIF3B) is closely linked to the transcription initia- Introduction tion of cancer-associated genes. In the present study, EIF3B was indicated to be a potential prognostic biomarker of liver cancer. Liver cancer is a common malignant tumor type with high The mRNA expression level of EIF3B in liver cancer was morbidity and mortality (1). Although various treatments have assessed by analyzing the Cancer Genome Atlas dataset. χ2 and been improved, the mortality rate of liver cancer is still high Fisher's exact tests were used to assess the association of EIF3B and the prognosis remains poor (2,3). Therefore, prognostic expression with clinical parameters. Receiver-operating char- biomarkers of liver cancer have become one of the hotspots acteristic curve analysis was used for evaluating the diagnostic of current research (4).
    [Show full text]
  • Loss of Eif4e Phosphorylation Engenders Depression-Like Behaviors Via Selective Mrna Translation
    This Accepted Manuscript has not been copyedited and formatted. The final version may differ from this version. A link to any extended data will be provided when the final version is posted online. Research Articles: Neurobiology of Disease Loss of eIF4E phosphorylation engenders depression-like behaviors via selective mRNA translation Inês Silva Amorim1,2, Sonal Kedia1,2, Stella Kouloulia1,2, Konstanze Simbriger1,2, Ilse Gantois3, Seyed Mehdi Jafarnejad3, Yupeng Li1,2, Agniete Kampaite1,2, Tine Pooters1, Nicola Romanò1 and Christos G. Gkogkas1,2,4 1Centre for Discovery Brain Sciences, EH8 9XD, Edinburgh, Scotland, UK 2The Patrick Wild Centre, EH8 9XD, Edinburgh, Scotland, UK 3Goodman Cancer Research Centre and Biochemistry Department, McGill University, H3A 1A3, Montréal, QC, Canada 4Simons Initiative for the Developing Brain, EH8 9XD, Edinburgh, Scotland, UK DOI: 10.1523/JNEUROSCI.2673-17.2018 Received: 16 September 2017 Revised: 3 December 2017 Accepted: 8 January 2018 Published: 24 January 2018 Author contributions: I.S.A., S. Kedia, S. Kouloulia, S.M.J., N.R., and C.G.G. designed research; I.S.A., S. Kedia, S. Kouloulia, K.S., I.G., S.M.J., Y.L., A.K., N.R., and C.G.G. performed research; I.S.A., S. Kedia, S. Kouloulia, K.S., I.G., S.M.J., A.K., T.P., and C.G.G. analyzed data; N.R. and C.G.G. contributed unpublished reagents/analytic tools; C.G.G. wrote the paper. Conflict of Interest: The authors declare no competing financial interests. This work was supported by grants to CGG: Sir Henry Dale Fellowship from the Wellcome Trust and Royal Society (107687/Z/15/Z), a NARSAD Young Investigator grant from the Brain & Behavior Research Foundation, the RS Macdonald Charitable Trust and the Patrick Wild Centre.
    [Show full text]
  • Inhibition of the MID1 Protein Complex
    Matthes et al. Cell Death Discovery (2018) 4:4 DOI 10.1038/s41420-017-0003-8 Cell Death Discovery ARTICLE Open Access Inhibition of the MID1 protein complex: a novel approach targeting APP protein synthesis Frank Matthes1,MoritzM.Hettich1, Judith Schilling1, Diana Flores-Dominguez1, Nelli Blank1, Thomas Wiglenda2, Alexander Buntru2,HannaWolf1, Stephanie Weber1,InaVorberg 1, Alina Dagane2, Gunnar Dittmar2,3,ErichWanker2, Dan Ehninger1 and Sybille Krauss1 Abstract Alzheimer’s disease (AD) is characterized by two neuropathological hallmarks: senile plaques, which are composed of amyloid-β (Aβ) peptides, and neurofibrillary tangles, which are composed of hyperphosphorylated tau protein. Aβ peptides are derived from sequential proteolytic cleavage of the amyloid precursor protein (APP). In this study, we identified a so far unknown mode of regulation of APP protein synthesis involving the MID1 protein complex: MID1 binds to and regulates the translation of APP mRNA. The underlying mode of action of MID1 involves the mTOR pathway. Thus, inhibition of the MID1 complex reduces the APP protein level in cultures of primary neurons. Based on this, we used one compound that we discovered previously to interfere with the MID1 complex, metformin, for in vivo experiments. Indeed, long-term treatment with metformin decreased APP protein expression levels and consequently Aβ in an AD mouse model. Importantly, we have initiated the metformin treatment late in life, at a time-point where mice were in an already progressed state of the disease, and could observe an improved behavioral phenotype. These 1234567890 1234567890 findings together with our previous observation, showing that inhibition of the MID1 complex by metformin also decreases tau phosphorylation, make the MID1 complex a particularly interesting drug target for treating AD.
    [Show full text]
  • Distinct Interactions of Eif4a and Eif4e with RNA Helicase Ded1 Stimulate Translation in Vivo Suna Gulay, Neha Gupta, Jon R Lorsch, Alan G Hinnebusch*
    RESEARCH ARTICLE Distinct interactions of eIF4A and eIF4E with RNA helicase Ded1 stimulate translation in vivo Suna Gulay, Neha Gupta, Jon R Lorsch, Alan G Hinnebusch* Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States Abstract Yeast DEAD-box helicase Ded1 stimulates translation initiation, particularly of mRNAs with structured 5’UTRs. Interactions of the Ded1 N-terminal domain (NTD) with eIF4A, and Ded1- CTD with eIF4G, subunits of eIF4F, enhance Ded1 unwinding activity and stimulation of preinitiation complex (PIC) assembly in vitro. However, the importance of these interactions, and of Ded1-eIF4E association, in vivo were poorly understood. We identified separate amino acid clusters in the Ded1-NTD required for binding to eIF4A or eIF4E in vitro. Disrupting each cluster selectively impairs native Ded1 association with eIF4A or eIF4E, and reduces cell growth, polysome assembly, and translation of reporter mRNAs with structured 5’UTRs. It also impairs Ded1 stimulation of PIC assembly on a structured mRNA in vitro. Ablating Ded1 interactions with eIF4A/eIF4E unveiled a requirement for the Ded1-CTD for robust initiation. Thus, Ded1 function in vivo is stimulated by independent interactions of its NTD with eIF4E and eIF4A, and its CTD with eIF4G. Introduction Eukaryotic translation initiation is an intricate process that ensures accurate selection and decoding *For correspondence: of the mRNA start codon. Initiation
    [Show full text]