Differential Regulation of the Melanoma Proteome by Eif4a1 and Eif4e Cailin E

Total Page:16

File Type:pdf, Size:1020Kb

Differential Regulation of the Melanoma Proteome by Eif4a1 and Eif4e Cailin E Published OnlineFirst November 22, 2016; DOI: 10.1158/0008-5472.CAN-16-1298 Cancer Integrated Systems and Technologies Research Differential Regulation of the Melanoma Proteome by eIF4A1 and eIF4E Cailin E. Joyce1,2,3, Adrienne G. Yanez1,2,3, Akihiro Mori4,5,6, Akinori Yoda1,2,3, Johanna S. Carroll1,2,3, and Carl D. Novina1,2,3 Abstract Small molecules and antisense oligonucleotides that inhibit eIF4A1 and eIF4E with patient survival. Finally, comparative the translation initiation factors eIF4A1 and eIF4E have been proteomic and transcriptomic analyses reveal extensive mech- explored as broad-based therapeutic agents for cancer treat- anistic divergence in response to eIF4A1 or eIF4E silencing. ment, based on the frequent upregulation of these two sub- Current models indicate that eIF4A1 and eIF4E function units of the eIF4F cap-binding complex in many cancer cells. together through the 50UTR to increase translation of onco- Here, we provide support for these therapeutic approaches genes. In contrast, our data demonstrate that the common with mechanistic studies of eIF4F-driven tumor progression effects of eIF4A1 and eIF4E on translation are mediated by the in a preclinical model of melanoma. Silencing eIF4A1 or eIF4E coding region and 30UTR. Moreover, their divergent effects decreases melanoma proliferation and invasion. There were occur through the 50UTR. Overall, our work shows that it will common effects on the level of cell-cycle proteins that could be important to evaluate subunit-specific inhibitors of eIF4F in explain the antiproliferative effects in vitro.Usingclinical different disease contexts to fully understand their anticancer specimens, we correlate the common cell-cycle targets of actions. Cancer Res; 77(3); 1–10. Ó2016 AACR. Introduction Classic models dictate that eIF4E is the rate-limiting factor for translation initiation in most normal tissues (11). Under phys- Steady-state translation initiation is primarily regulated iologic conditions, mRNAs are thought to compete for limiting through the cytoplasmic mRNA cap–binding complex, eukary- amounts of eIF4E. In this model, mRNAs with longer and more otic initiation factor 4F (eIF4F), comprised of the cap-binding 0 structured 5 UTRs are weakly translated. In cancer cells, increased protein eIF4E, the RNA helicase eIF4A, and the scaffolding availability of eIF4E leads to selective translation of highly sen- protein eIF4G (reviewed in ref.1). Increased protein levels of sitive oncogenes, such as MYC (12) and VEGF (13). More recently, eIF4E (2), phospho-eIF4E (3), eIF4A1 (4), and eIF4G (5) are 0 this model was extended to incorporate 5 UTR sequence elements associated with poor clinical outcomes in multiple cancer that confer eIF4E sensitivity such as the terminal oligonucleotide contexts and are linked to aggressive phenotypes in cellular 0 tract (5 TOP), pyrimidine-rich translational element (PRTE), and and preclinical models (6–8). In melanoma, many of the cytosine-enriched regulator of translation (CERT; refs.14–16). known oncogenic signaling pathways converge on eIF4F (9). 0 However, 5 UTR features do not explain the full spectrum of Targeted therapies that deactivate these pathways are initially eIF4E targets (17), including the class of mRNAs for which eIF4E effective, but nearly universal acquired resistance leads to high negatively regulates translation. Improved models are necessary mortality rates (10). Thus, second-line inhibition of eIF4F to predict outcomes of therapeutic inhibition of eIF4F in cancer. could significantly extend the period of response to targeted Targets of eIF4A and eIF4E have been identified using poly- therapies for patients with melanoma. somal RNA association or ribosome profiling to estimate trans- lation efficiency (16, 18–21). Some mRNAs have different requirements for individual eIF4F subunits (19, 22), but global 1Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, effects of eIF4A1 and eIF4E depletion have not been assessed in Boston, Massachusetts. 2Department of Medicine, Harvard Medical School, parallel in the same cancer model. Here, we characterize the Boston, Massachusetts. 3Broad Institute of Harvard and MIT, Cambridge, Mas- phenotypic and global proteomic effects of eIF4A1 and eIF4E sachusetts. 4Program in Systems Biology and Program in Molecular Medicine, inhibition in melanoma. We show that eIF4A1 and eIF4E pro- 5 University of Massachusetts, Worcester, Massachusetts. Onami team, The mote melanoma proliferation through shared, network-level 6 Systems Biology Institute, Tokyo, Japan. Laboratory for Developmental regulation of the cell cycle, and moreover, that eIF4F-dependent Dynamics, RIKEN Quantitative Biology Center, Hyogo, Japan. cell-cycle proteins are significant combinatorial predictors of Note: Supplementary data for this article are available at Cancer Research patient survival. Finally, we demonstrate that intrinsic features Online (http://cancerres.aacrjournals.org/). of mRNA-coding sequence (CDS) and 30UTR, but not 50UTR, Corresponding Author: Carl D. Novina, Dana-Farber Cancer Institute, Dana predict shared targets of eIF4A1 and eIF4E. These data demon- 1420B, 450 Brookline Avenue, Boston, MA 02215. Phone: 617-582-7961; Fax: 617- strate that eIF4A1 and eIF4E have distinct molecular conse- 582-7962; E-mail: [email protected] quences in the same cancer model, which implies that targeting doi: 10.1158/0008-5472.CAN-16-1298 eIF4F with different subunit-specific inhibitors will influence on- Ó2016 American Association for Cancer Research. target and off-target therapeutic outcomes. www.aacrjournals.org OF1 Downloaded from cancerres.aacrjournals.org on September 27, 2021. © 2016 American Association for Cancer Research. Published OnlineFirst November 22, 2016; DOI: 10.1158/0008-5472.CAN-16-1298 Joyce et al. Materials and Methods Quantitative mass spectrometry Knockdowns were performed in triplicate in 10-cm dishes and Cell culture cells were collected 72 hours post-transfection. Samples were WM858, WM46, and A375 were cultured in high-glucose processed and analyzed for MS3 mass spectrometry as described DMEM (Cellgro) supplemented with 10% FBS (Life Technol- previously (23). Briefly, cells were lysed, proteins were digested ogies). WM46 was transduced with lentivirus containing empty using LysC and trypsin, and peptides were labeled with Tandem SparQ IRES RFP vector (QM531A-2, System Biosciences), or Mass Tag 10-plex reagents and fractionated. Data were collected vector containing eIF4A1 (4096621, Open Biosystems) or on an Orbitrap Fusion mass spectrometer operating in MS3 eIF4E cDNA (5295521, Open Biosystems). RFP-positive cells mode using synchronous precursor selection for MS2 to MS3 in the middle quantiles were selected by flow cytometry. The fragmentation, and forward and reverse sequences were searched A375 melanoma cell line was a gift from Dr. David Fisher against a Uniprot human database (February 2014) using the (Massachusetts General Hospital, Boston, MA), and WM46 and SEQUEST algorithm. Additional processing steps included pro- WM858 were a gift from Dr. Levi Garraway (Dana-Farber tein assembly and quantification from peptides, and calculation Cancer Institute, Boston, MA), all obtained prior to 2012. All of Benjamini–Hochberg false discovery rates (FDR). Protein levels cell lines were authenticated via short tandem repeat profiling were normalized to the percent signal in each sample relative to at the American Tissue Culture Repository on May 19, 2016. the total signal across all multiplexed samples. Differentially expressed proteins had a fold change |1.25| in all three biolog- Transient knockdown ical replicates and FDR < 0.1. A375 and WM858 cells were transfected with Allstars Negative Control siRNA (Qiagen) or gene-specific siRNA at 7.5 nmol/L using Pathway and network analysis RNAiMAX (Life Technologies). Gene-specific siRNA sequences Gene set enrichment analysis (GSEA) was performed using were 50-GGACCAGATCTATGACATATT-30 and 50-GCATGGAGA- GSEA software (http://software.broadinstitute.org/gsea/index. TATGGACCAATT-30 for eIF4A1 (custom design, Qiagen) and jspGene). Differentially expressed proteins for each treatment 50-AAGAGCGGCTCCACCACTAAA-30 and 50-CAAAGCTTTGCTA- were ranked on the basis of their average fold-change across CAAATTTA-30 for eIF4E (Hs_EIF4E_1 and Hs_EIF4E_8, Qiagen). triplicates and analyzed against Reactome pathways (http:// siRNA-like off-target effects were assessed by siRNA-check (http:// www.reactome.org/pathways) using the GSEA Preranked tool. projects.insilico.us/SpliceCenter/siRNACheck). miRNA-like off- We applied a classic enrichment statistic, filtered gene sets with target effects were assessed by scanning 30UTRs for perfect matches >500 or <15 hits, and applied an FDR threshold 0.1. To visualize to the 8-mer seed region of each oligonucleotide (underlined enrichment of common pathways, we ranked all genes based on above). their average fold-change across all knockdown treatments and performed GSEA Preranked as above. The normalized enrichment Proliferation assays score calculates the degree to which a gene set is over-represented A total of 2,500 cells per well were seeded into 96-well plates at the extremes of a ranked list, accounting for size of the gene set. and, where appropriate, transfected with siRNA. After 24 hours, To predict functional outcomes of differentially expressed genes, cell culture medium was replaced with 10 mL Wst-1 (Pierce) in we used the Diseases and Functions tool in Ingenuity Pathway 100 mL complete medium.
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]
  • 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]
  • 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]
  • 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]
  • UNDERSTANDING the ROLE of EIF4A in GENE REGULATION in HEALTH and DISEASE THOMAS EDWARD WEBB B.Sc
    UNDERSTANDING THE ROLE OF EIF4A IN GENE REGULATION IN HEALTH AND DISEASE THOMAS EDWARD WEBB B.Sc. (Hons, Warwick) M.Sc. (Oxon) Thesis submitted to The University of Nottingham for the degree of Doctor of Philosophy (July 2012) 1 Abstract eIF4A Eukaryotic initiation factor 4A (eIF4A) is an ATP-dependent RNA helicase responsible for unwinding the secondary structure of mRNAs. In humans, eIF4A exists as three separate paralogs: eIF4AI and eIF4AII possess a high degree of homology while eIF4AIII is distinct. Knockdown of eIF4AII had no effect on the expression of a reporter construct containing a structured RNA hairpin. Knockdown of eIF4AI and treatment with hippuristanol (an eIF4A inhibitor) caused a dramatic reduction in the hairpin-mediated gene. This reporter system was developed as part of this project to act as a screen for eIF4A activity along with an in vitro screening approach. PDCD4 The activity of eIF4A is suppressed in vivo by the tumour suppressor PDCD4. The fact that loss of PDCD4 function increases the severity of DNA damage is probably attributable its eIF4A-suppressive activity. Alzheimer's Disease Based on previous microarray data, it was supposed that eIF4A inhibition may be therapeutically beneficial in the treatment of Alzheimer's disease. As part of this project, it was demonstrated that eIF4A suppression significantly reduced the expression of reporter genes preceded by the 5’ UTRs of genes predicted to play harmful roles in Alzheimer’s disease. The expression of reporter genes preceded by the 5’ UTR sequences of genes predicted to be beneficial in Alzheimer's were not affected by this suppression.
    [Show full text]
  • Lysine Demethylase KDM4A Associates with Translation Machinery and Regulates Protein Synthesis
    Published OnlineFirst January 6, 2015; DOI: 10.1158/2159-8290.CD-14-1326 RESEARCH BRIEF Lysine Demethylase KDM4A Associates with Translation Machinery and Regulates Protein Synthesis Capucine Van Rechem 1 , Joshua C. Black 1 , Myriam Boukhali 1 , Martin J. Aryee 2 , Susanne Gräslund 3 , Wilhelm Haas 1 , Cyril H. Benes 1 , and Johnathan R. Whetstine 1 ABSTRACT Chromatin-modifying enzymes are predominantly nuclear; however, these factors are also localized to the cytoplasm, and very little is known about their role in this compartment. In this report, we reveal a non–chromatin-linked role for the lysine-specifi c demethylase KDM4A. We demonstrate that KDM4A interacts with the translation initiation complex and affects the distribution of translation initiation factors within polysome fractions. Furthermore, KDM4A depletion reduced protein synthesis and enhanced the protein synthesis suppression observed with mTOR inhibi- tors, which paralleled an increased sensitivity to these drugs. Finally, we demonstrate that JIB-04, a JmjC demethylase inhibitor, suppresses translation initiation and enhances mTOR inhibitor sensitivity. These data highlight an unexpected cytoplasmic role for KDM4A in regulating protein synthesis and suggest novel potential therapeutic applications for this class of enzyme. SIGNIFICANCE: This report documents an unexpected cytoplasmic role for the lysine demethylase KDM4A. We demonstrate that KDM4A interacts with the translation initiation machinery, regulates protein synthesis and, upon coinhibition with mTOR inhibitors, enhances the translation suppression and cell sensitivity to these therapeutics. Cancer Discov; 5(3); 255–63. ©2015 AACR. See related commentary by Rothbart et al., p. 228. See related article by Van Rechem et al., p. 245. INTRODUCTION (KDM), respectively, are affecting the associated targets or their downstream processes.
    [Show full text]
  • The Role of Eif5a Hypusination in Mediating Oncogenic Mtor Signaling
    The role of eIF5A hypusination in mediating oncogenic mTOR signaling Yutian Cai Faculty of Medicine Department of Biochemistry McGill University, Montreal, Quebec, Canada December 2016 A thesis submitted to McGill University in partial fulfillment of the requirements of the degree of Master of Science ©Yutian Cai 2016 TABLE OF CONTENTS TABLE OF CONTENTS ........................................................................................ 2 TABLE OF FIGURES ............................................................................................ 4 PREFACE.............................................................................................................. 5 ACKNOWLEDGEMENTS ...................................................................................... 6 ABSTRACT ........................................................................................................... 7 RÉSUMÉ ............................................................................................................... 9 LIST OF ABBREVIATIONS ................................................................................. 11 CHAPTER 1: INTRODUCTION ........................................................................... 14 1.1 mRNA TRANSLATION ......................................................................... 14 1.1.1 TRANSLATION INITIATION....................................................................... 15 1.1.2 TRANSLATIONAL EFFICIENCY ............................................................... 20 1.1.3 TRANSLATION AND CANCER
    [Show full text]
  • A Comparative Study of Small Molecules Targeting Eif4a
    Downloaded from rnajournal.cshlp.org on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press A Comparative Study of Small Molecules Targeting eIF4A Sai Kiran Naineni1,*, Rayelle Itoua Maïga1,*, Regina Cencic1, Andrea A. Putnam2, Luis A. Amador3, Abimael D. Rodriguez3, Eckhard Jankowsky2, and Jerry Pelletier1,4,5 1Department of Biochemistry, McGill University, 2Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH; 3University of Puerto Rico, Faculty of Natural Sciences, Dept of Chemistry, San Juan, Puerto Rico, 4Department of Oncology, 5Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, Québec, Canada, H3G 1Y6 Running Title: A comparative study of eIF4A inhibitors Key words: Translation initiation, eIF4A, RNA helicase, eIF4F, hippuristanol *Equal contribution by both authors To whom correspondence should be addressed: Jerry Pelletier, Department of Biochemistry, McGill University, Montreal, Quebec, H3G 1Y6, Canada. Tel: 1-514-398-2323; Fax: 514-398-2965; Email: [email protected] 1 Downloaded from rnajournal.cshlp.org on October 2, 2021 - Published by Cold Spring Harbor Laboratory Press ABSTRACT The PI3K/Akt/mTOR kinase pathway is extensively deregulated in human cancers. One critical node under regulation of this signaling axis is eukaryotic initiation factor (eIF) 4F, a complex involved in the control of translation initiation rates. eIF4F-dependent addictions arise during tumor initiation and maintenance due to increased eIF4F activity – generally in response to elevated PI3K/Akt/mTOR signaling flux. There is thus much interest in exploring eIF4F as a small molecule target for the development of new anti-cancer drugs. The DEAD-box RNA helicase, eIF4A, is an essential subunit of eIF4F and several potent small molecules (rocaglates, hippuristanol, pateamine A) affecting its activity have been identified and shown to demonstrate anti-cancer activity in vitro and in vivo in pre-clinical models.
    [Show full text]
  • Phosphoglycerate Dehydrogenase Promotes Pancreatic Cancer
    Ma et al. Journal of Experimental & Clinical Cancer Research (2019) 38:66 https://doi.org/10.1186/s13046-019-1053-y RESEARCH Open Access Phosphoglycerate dehydrogenase promotes pancreatic cancer development by interacting with eIF4A1 and eIF4E Xuhui Ma1,2,3, Boya Li1,2,3, Jie Liu1,2,3, Yan Fu1,2,3 and Yongzhang Luo1,2,3* Abstract Background: Pancreatic cancer is one of the most malignant cancers. The overall 5-year survival rate of its patients is 8%, the lowest among major cancer types. It is very urgent to study the development mechanisms of this cancer and provide potential targets for therapeutics design. Glucose, one of the most essential nutrients, is highly exploited for aerobic glycolysis in tumor cells to provide building blocks. However, the glucose consumption manner in pancreatic cancer cells is unclear. And the mechanism of the substantial metabolic pathway promoting pancreatic cancer development is also unrevealed. 13 Methods: C6 glucose was used to trace the glucose carbon flux and detected by mass spectrum. The expressions of PHGDH were determined in cells and pancreatic adenocarcinomas. Knockdown and overexpression were performed to investigate the roles of PHGDH on pancreatic cancer cell proliferation, colony formation and tumor growth. The mechanisms of PHGDH promoting pancreatic cancer development were studied by identifying the interacting proteins and detecting the regulatory functions on translation initiations. Results: Pancreatic cancer cells PANC-1 consumed large amounts of glucose in the serine and glycine de novo synthesis. Phosphoglycerate dehydrogenase (PHGDH) highly expressed and controlled this pathway. Knockdown of PHGDH significantly attenuated the tumor growth and prolonged the survival of tumor bearing mice.
    [Show full text]
  • General and Target-Specific Dexd/H RNA Helicases in Eukaryotic Translation Initiation
    International Journal of Molecular Sciences Review General and Target-Specific DExD/H RNA Helicases in Eukaryotic Translation Initiation Leo Shen 1 and Jerry Pelletier 1,2,3,* 1 Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada; [email protected] 2 Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada 3 Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, QC H3A 1A3, Canada * Correspondence: [email protected]; Tel.: +1-514-398-2323 Received: 5 June 2020; Accepted: 18 June 2020; Published: 20 June 2020 Abstract: DExD (DDX)- and DExH (DHX)-box RNA helicases, named after their Asp-Glu-x-Asp/His motifs, are integral to almost all RNA metabolic processes in eukaryotic cells. They play myriad roles in processes ranging from transcription and mRNA-protein complex remodeling, to RNA decay and translation. This last facet, translation, is an intricate process that involves DDX/DHX helicases and presents a regulatory node that is highly targetable. Studies aimed at better understanding this family of conserved proteins have revealed insights into their structures, catalytic mechanisms, and biological roles. They have also led to the development of chemical modulators that seek to exploit their essential roles in diseases. Herein, we review the most recent insights on several general and target-specific DDX/DHX helicases in eukaryotic translation initiation. Keywords: helicase; translation; initiation; DDX; DHX; eIF4A1; DDX3X; DHX9; DHX29; DHX36 1. Introduction RNA helicases play a role in all facets of RNA metabolism, ranging from transcription and translation, to processing and decay. These highly conserved enzymes use ATP to bind, unwind, and disrupt RNA structures and RNA-protein complexes [1].
    [Show full text]