1
|
dos Santos JV, Medina JM, Dias Teixeira KL, Agostinho DMJ, Chorev M, Diotallevi A, Galluzzi L, Aktas BH, Gazos Lopes U. Activity of the Di-Substituted Urea-Derived Compound I-17 in Leishmania In Vitro Infections. Pathogens 2024; 13:104. [PMID: 38392842 PMCID: PMC10893125 DOI: 10.3390/pathogens13020104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/25/2024] Open
Abstract
Protein synthesis has been a very rich target for developing drugs to control prokaryotic and eukaryotic pathogens. Despite the development of new drug formulations, treating human cutaneous and visceral Leishmaniasis still needs significant improvements due to the considerable side effects and low adherence associated with the current treatment regimen. In this work, we show that the di-substituted urea-derived compounds I-17 and 3m are effective in inhibiting the promastigote growth of different Leishmania species and reducing the macrophage intracellular load of amastigotes of the Leishmania (L.) amazonensis and L. major species, in addition to exhibiting low macrophage cytotoxicity. We also show a potential immunomodulatory effect of I-17 and 3m in infected macrophages, which exhibited increased expression of inducible Nitric Oxide Synthase (NOS2) and production of Nitric Oxide (NO). Our data indicate that I-17, 3m, and their analogs may be helpful in developing new drugs for treating leishmaniasis.
Collapse
Affiliation(s)
- José Vitorino dos Santos
- Laboratory of Molecular Parasitology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.V.d.S.); (J.M.M.); (D.M.J.A.)
| | - Jorge Mansur Medina
- Laboratory of Molecular Parasitology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.V.d.S.); (J.M.M.); (D.M.J.A.)
| | | | - Daniel Marcos Julio Agostinho
- Laboratory of Molecular Parasitology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.V.d.S.); (J.M.M.); (D.M.J.A.)
| | - Michael Chorev
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Aurora Diotallevi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (A.D.)
| | - Luca Galluzzi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (A.D.)
| | - Bertal Huseyin Aktas
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Ulisses Gazos Lopes
- Laboratory of Molecular Parasitology, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil; (J.V.d.S.); (J.M.M.); (D.M.J.A.)
| |
Collapse
|
2
|
Yang H, Chen T, Denoyelle S, Chen L, Fan J, Zhang Y, Halperin JA, Chorev M, Aktas BH. Role of symmetry in 3,3-diphenyl-1,3-dihydroindol-2-one derivatives as inhibitors of translation initiation. Bioorg Med Chem Lett 2023; 80:129119. [PMID: 36581302 PMCID: PMC9922553 DOI: 10.1016/j.bmcl.2022.129119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/13/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022]
Abstract
The ternary complex (eIF2·GTP·Met-tRNAiMet) and the eIF4F complex assembly are two major regulatory steps in the eukaryotic translation initiation. Inhibition of the ternary complex assembly is therefore a promising target for the development of novel anti-cancer therapeutics. Building on the finding that clotrimazole (CLT), a molecular probe that depletes intracellular Ca2+ stores and subsequently induce eIF2α phosphorylation, inhibit translation initiation, and reduce preferentially the expression of oncoproteins over "housekeeping" ones,1-3 we undertook structure activity relationship (SAR) studies that identified 3,3-diarylindoline-2-one #1181 as an interesting scaffold. Compound #1181 also induce phosphorylation of eIF2α thereby reducing the availability of the ternary complex, which leads to inhibition of translation initiation.4 Our subsequent efforts focused on understanding SAR iterative lead optimization to enhance potency and improve bioavailability. Herein, we report a complementing study focusing on heavily substituted symmetric and asymmetric 3,3-(o,m-disubstituted)diarylindoline-2-ones. These compounds were evaluated by the dual luciferase reporter ternary complex assay that recapitualates phosphorylation of eIF2α in a quantitative manner. We also evaluated all compounds by sulforhodamine B assay, which measures the overall effect of compounds on cell proliferations and/or viability.
Collapse
Affiliation(s)
- Hongwei Yang
- Brigham and Women's Hospital, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA.
| | - Ting Chen
- Brigham and Women's Hospital, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA
| | - Séverine Denoyelle
- Brigham and Women's Hospital, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA
| | - Limo Chen
- Brigham and Women's Hospital, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA
| | - Jing Fan
- Brigham and Women's Hospital, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA
| | - Yingzhen Zhang
- Brigham and Women's Hospital, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA
| | - José A Halperin
- Brigham and Women's Hospital, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA; Harvard Medical School, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA
| | - Michael Chorev
- Brigham and Women's Hospital, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA; Harvard Medical School, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA.
| | - Bertal H Aktas
- Brigham and Women's Hospital, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA; Harvard Medical School, Division of Hematology, 4 Balckfan Circle. HIM 7, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Jiang SL, Mo JL, Peng J, Lei L, Yin JY, Zhou HH, Liu ZQ, Hong WX. Targeting translation regulators improves cancer therapy. Genomics 2020; 113:1247-1256. [PMID: 33189778 DOI: 10.1016/j.ygeno.2020.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/14/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
Deregulation of protein synthesis may be involved in multiple aspects of cancer, such as gene expression, signal transduction and drive specific cell biological responses, resulting in promoting cancer growth, invasion and metastasis. Study the molecular mechanisms about translational control may help us to find more effective anti-cancer drugs and develop novel therapeutic opportunities. Recently, the researchers had focused on targeting translational machinery to overcome cancer, and various small molecular inhibitors targeting translation factors or pathways have been tested in clinical trials and exhibited improving outcomes in several cancer types. There is no doubt that an insight into the class of translation regulation protein would provide new target for pharmacologic intervention and further provide opportunities to develop novel anti-tumor therapeutic interventions. In this review, we summarized the developments of translational control in cancer survival and progression et al, and highlighted the therapeutic approach targeted translation regulation to overcome the cancer.
Collapse
Affiliation(s)
- Shi-Long Jiang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China
| | - Jun-Luan Mo
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China; Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen 518020, PR China
| | - Ji Peng
- Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen 518020, PR China
| | - Lin Lei
- Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen 518020, PR China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Engineering Research Center for applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, PR China.
| | - Wen-Xu Hong
- Shenzhen Center for Chronic Disease Control and Prevention, Shenzhen 518020, PR China.
| |
Collapse
|
4
|
Wu Chuang A, Kepp O, Kroemer G, Bezu L. Endoplasmic reticulum stress in the cellular release of damage-associated molecular patterns. BIOLOGY OF THE ENDOPLASMIC RETICULUM 2020; 350:1-28. [DOI: 10.1016/bs.ircmb.2019.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
5
|
Zhang Q, Du R, Reis Monteiro Dos Santos GR, Yefidoff-Freedman R, Bohm A, Halperin J, Chorev M, Aktas BH. New activators of eIF2α Kinase Heme-Regulated Inhibitor (HRI) with improved biophysical properties. Eur J Med Chem 2019; 187:111973. [PMID: 31881453 DOI: 10.1016/j.ejmech.2019.111973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 12/13/2019] [Accepted: 12/13/2019] [Indexed: 01/21/2023]
Abstract
Heme-regulated inhibitor (HRI), a eukaryotic translation initiation factor 2 alpha (eIF2α) kinase, is critically important for coupling protein synthesis to heme availability in reticulocytes and adaptation to various environmental stressors in all cells. HRI modifies the severity of several hemoglobin misfolding disorders including β-thalassemia. Small molecule activators of HRI are essential for studying normal- and patho-biology of this kinase as well as for the treatment of various human disorders for which activation of HRI or phosphorylation of eIF2α may be beneficial. We previously reported development of 1-((1,4-trans)-4-aryloxycyclohexyl)-3-arylureas (cHAUs) as specific HRI activators and demonstrated their potential as molecular probes for studying HRI biology and as lead compounds for treatment of various human disorders. To develop more druglike cHAUs for in vivo studies and drug development and to expand the chemical space, we undertook bioassay guided structure-activity relationship studies replacing cyclohexyl ring with various 4-6-membered rings and explored further substitutions on the N-phenyl ring. We tested all analogs in the surrogate eIF2α phosphorylation and cell proliferation assays, and a subset of analogs in secondary mechanistic assays that included endogenous eIF2α phosphorylation and expression of C/EBP homologous protein (CHOP), a downstream effector. Finally, we determined specificity of these compounds for HRI by testing their anti-proliferative activity in cells transfected with siRNA targeting HRI or mock. These compounds have significantly improved cLogPs with no loss of potencies, making them excellent candidates for lead optimization for development of investigational new drugs that potently and specifically activate HRI.
Collapse
Affiliation(s)
- Qingwen Zhang
- Division of Medicinal and Process Chemistry, Shanghai Institute of Pharmaceutical Industry, Pudong, Shanghai, 201203, China; Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Ronghui Du
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA; Medicine School of Nanjing University, Nanjing, Jiangsu, 210093, China
| | | | - Revital Yefidoff-Freedman
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew Bohm
- Tufts University Medical School, Boston, MA, 02117, USA
| | - Jose Halperin
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Chorev
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Bertal H Aktas
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
6
|
The Best for the Most Important: Maintaining a Pristine Proteome in Stem and Progenitor Cells. Stem Cells Int 2019; 2019:1608787. [PMID: 31191665 PMCID: PMC6525796 DOI: 10.1155/2019/1608787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 03/05/2019] [Indexed: 12/19/2022] Open
Abstract
Pluripotent stem cells give rise to reproductively enabled offsprings by generating progressively lineage-restricted multipotent stem cells that would differentiate into lineage-committed stem and progenitor cells. These lineage-committed stem and progenitor cells give rise to all adult tissues and organs. Adult stem and progenitor cells are generated as part of the developmental program and play critical roles in tissue and organ maintenance and/or regeneration. The ability of pluripotent stem cells to self-renew, maintain pluripotency, and differentiate into a multicellular organism is highly dependent on sensing and integrating extracellular and extraorganismal cues. Proteins perform and integrate almost all cellular functions including signal transduction, regulation of gene expression, metabolism, and cell division and death. Therefore, maintenance of an appropriate mix of correctly folded proteins, a pristine proteome, is essential for proper stem cell function. The stem cells' proteome must be pristine because unfolded, misfolded, or otherwise damaged proteins would interfere with unlimited self-renewal, maintenance of pluripotency, differentiation into downstream lineages, and consequently with the development of properly functioning tissue and organs. Understanding how various stem cells generate and maintain a pristine proteome is therefore essential for exploiting their potential in regenerative medicine and possibly for the discovery of novel approaches for maintaining, propagating, and differentiating pluripotent, multipotent, and adult stem cells as well as induced pluripotent stem cells. In this review, we will summarize cellular networks used by various stem cells for generation and maintenance of a pristine proteome. We will also explore the coordination of these networks with one another and their integration with the gene regulatory and signaling networks.
Collapse
|
7
|
Clotrimazole inhibits the Wnt/β-catenin pathway by activating two eIF2α kinases: The heme-regulated translational inhibitor and the double-stranded RNA-induced protein kinase. Biochem Biophys Res Commun 2018; 506:183-188. [PMID: 30342850 DOI: 10.1016/j.bbrc.2018.10.053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/07/2018] [Indexed: 01/08/2023]
Abstract
The Wnt/β-catenin signaling pathway controls cell proliferation and differentiation, and therefore, when this pathway is excessively activated, it causes tumorigenesis. Our chemical suppressor screening in zebrafish embryos identified antifungal azoles including clotrimazole, miconazole, and itraconazole, as Wnt/β-catenin signaling inhibitors. Here we show the mechanism underlying the Wnt/β-catenin pathway inhibition by antifungal azoles. Clotrimazole reduced β-catenin revels in a proteasome-independent fashion. By gene knockdown of two translational regulators, heme-regulated translational inhibitor and double-stranded RNA-induced protein kinase, we show that they mediate the clotrimazole-induced inhibition of the Wnt/β-catenin pathway. Thus, clotrimazole inhibits the Wnt/β-catenin pathway by decreasing β-catenin protein levels through translational regulation. Antifungal azoles represent genuine candidate compounds for anticancer drugs or chemopreventive agents that reduce adenomatous polyps.
Collapse
|
8
|
Opposite Regulation of CHOP and GRP78 and Synergistic Apoptosis Induction by Selenium Yeast and Fish Oil via AMPK Activation in Lung Adenocarcinoma Cells. Nutrients 2018; 10:nu10101458. [PMID: 30297634 PMCID: PMC6213479 DOI: 10.3390/nu10101458] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/18/2018] [Accepted: 09/26/2018] [Indexed: 11/17/2022] Open
Abstract
Selenium has been intensively studied for the use of cancer prevention and treatment. However, the clinical effects are still plausible. To enhance its efficacy, a combinational study of selenium yeast (SY) and fish oil (FO) was performed in A549, CL1-0, H1299, HCC827 lung adenocarcinoma (LADC) cells to investigate the enhancement in apoptosis induction and underlying mechanism. By sulforhodamine B staining, Western blot and flow cytometric assays, we found a synergism between SY and FO in growth inhibition and apoptosis induction of LADC cells. In contrast, the fetal lung fibroblast cells (MRC-5) were unsusceptible to this combination effect. FO synergized SY-induced apoptosis of A549 cells, accompanied with synergistic activation of AMP-activated protein kinase (AMPK) and reduction of Cyclooxygenase (COX)-2 and β-catenin. Particularly, combining with FO not only enhanced the SY-elevated proapoptotic endoplasmic reticulum (ER) stress marker CCAAT/enhancer-binding protein homologous protein (CHOP), but also reduced the cytoprotective glucose regulated protein of molecular weight 78 kDa (GRP78). Consequently, the CHOP downstream targets such as phospho-JNK and death receptor 5 were also elevated, along with the cleavage of caspase-8, -3, and the ER stress-related caspase-4. Accordingly, inhibition of AMPK by compound C diminished the synergistic apoptosis induction, and elevated CHOP/GRP78 ratio by SY combined with FO. The AMPK-dependent synergism suggests the combination of SY and FO for chemoprevention and integrative treatment of LADC.
Collapse
|
9
|
Burwick N, Aktas BH. The eIF2-alpha kinase HRI: a potential target beyond the red blood cell. Expert Opin Ther Targets 2017; 21:1171-1177. [PMID: 29063813 DOI: 10.1080/14728222.2017.1397133] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The eIF2α kinase heme-regulated inhibitor (HRI) is one of four well-described kinases that phosphorylate eIF2α in response to various cell stressors, resulting in reduced ternary complex formation and attenuation of mRNA translation. Although HRI is well known for its role as a heme sensor in erythroid progenitors, pharmacologic activation of HRI has been demonstrated to have anti-cancer activity across a wide range of tumor sub-types. Here, the potential of HRI activators as novel cancer therapeutics is explored. Areas covered: We provide an introduction to eIF2 signaling pathways in general, and specifically review data on the eIF2α kinase HRI in erythroid and non-erythroid cells. We review aspects of targeting eIF2 signaling in cancer and highlight promising data using HRI activators against tumor cells. Expert opinion: Pharmacologic activation of HRI inhibits tumor growth as a single agent without appreciable toxicity in vivo. The ability of HRI activators to provide direct and sustained eIF2α phosphorylation without inducing oxidative stress or broad eIF2α kinase activation may be especially advantageous for tolerability. Combination therapy with established therapeutics may further augment anti-cancer activity to overcome disease resistance.
Collapse
Affiliation(s)
- Nicholas Burwick
- a Division of hematology , VA Puget Sound Health Care System , Seattle , WA , USA.,b Division of Hematology , University of Washington School of Medicine , Seattle WA , USA
| | - Bertal H Aktas
- c Department of Medicine , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| |
Collapse
|
10
|
Yefidoff-Freedman R, Fan J, Yan L, Zhang Q, Dos Santos GRR, Rana S, Contreras JI, Sahoo R, Wan D, Young J, Dias Teixeira KL, Morisseau C, Halperin J, Hammock B, Natarajan A, Wang P, Chorev M, Aktas BH. Development of 1-((1,4-trans)-4-Aryloxycyclohexyl)-3-arylurea Activators of Heme-Regulated Inhibitor as Selective Activators of the Eukaryotic Initiation Factor 2 Alpha (eIF2α) Phosphorylation Arm of the Integrated Endoplasmic Reticulum Stress Response. J Med Chem 2017; 60:5392-5406. [PMID: 28590739 DOI: 10.1021/acs.jmedchem.7b00059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Heme-regulated inhibitor (HRI), an eukaryotic translation initiation factor 2 alpha (eIF2α) kinase, plays critical roles in cell proliferation, differentiation, adaptation to stress, and hemoglobin disorders. HRI phosphorylates eIF2α, which couples cellular signals, including endoplasmic reticulum (ER) stress, to translation. We previously identified 1,3-diarylureas and 1-((1,4-trans)-4-aryloxycyclohexyl)-3-arylureas (cHAUs) as specific activators of HRI that trigger the eIF2α phosphorylation arm of ER stress response as molecular probes for studying HRI biology and its potential as a druggable target. To develop drug-like cHAUs needed for in vivo studies, we undertook bioassay-guided structure-activity relationship studies and tested them in the surrogate eIF2α phosphorylation and cell proliferation assays. We further evaluated some of these cHAUs in endogenous eIF2α phosphorylation and in the expression of the transcription factor C/EBP homologous protein (CHOP) and its mRNA, demonstrating significantly improved solubility and/or potencies. These cHAUs are excellent candidates for lead optimization for development of investigational new drugs that potently and specifically activate HRI.
Collapse
Affiliation(s)
- Revital Yefidoff-Freedman
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States
| | - Jing Fan
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States.,Department of Orthopedics, Jiangsu Province Hospital of TCM, Nanjing University of Chinese Medicine , 155 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Lu Yan
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States
| | - Qingwen Zhang
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States.,Division of Medicinal and Process Chemistry, Shanghai Institute of Pharmaceutical Industry , 1111 Zhongshan North One Road, Hongkou District, Shanghai 200437, China
| | - Guillermo Rodrigo Reis Dos Santos
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States
| | - Sandeep Rana
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Jacob I Contreras
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Rupam Sahoo
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States
| | - Debin Wan
- Department of Entomology and Nematology, University of California Davis Comprehensive Cancer Center, University of California , One Shields Avenue, Davis, California 95616, United States
| | - Jun Young
- Department of Entomology and Nematology, University of California Davis Comprehensive Cancer Center, University of California , One Shields Avenue, Davis, California 95616, United States
| | - Karina Luiza Dias Teixeira
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States
| | - Christophe Morisseau
- Department of Entomology and Nematology, University of California Davis Comprehensive Cancer Center, University of California , One Shields Avenue, Davis, California 95616, United States
| | - Jose Halperin
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States
| | - Bruce Hammock
- Department of Entomology and Nematology, University of California Davis Comprehensive Cancer Center, University of California , One Shields Avenue, Davis, California 95616, United States
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Peimin Wang
- Department of Orthopedics, Jiangsu Province Hospital of TCM, Nanjing University of Chinese Medicine , 155 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Michael Chorev
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States
| | - Bertal H Aktas
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States
| |
Collapse
|
11
|
Persaud L, Zhong X, Alvarado G, Do W, Dejoie J, Zybtseva A, Aktas BH, Sauane M. eIF2α Phosphorylation Mediates IL24-Induced Apoptosis through Inhibition of Translation. Mol Cancer Res 2017; 15:1117-1124. [PMID: 28461326 DOI: 10.1158/1541-7786.mcr-16-0454] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 04/01/2017] [Accepted: 04/26/2017] [Indexed: 11/16/2022]
Abstract
IL24 is an immunomodulatory cytokine that also displays broad cancer-specific suppressor effects. The tumor-suppressor activities of IL24 include inhibition of angiogenesis, sensitization to chemotherapy, and cancer-specific apoptosis. Supra-physiologic activation and/or overexpression of translation initiation factors are implicated in the initiation and progression of cancer animal models as well as a subset of human cancers. Activation and/or overexpression of translation initiation factors correlate with aggressiveness of cancer and poor prognosis. Two rate-limiting translation initiation complexes, the ternary complex and the eIF4F complex, are regulated by eIF2α and 4E-BP1 phosphorylation, respectively. The work reported here provides direct evidence that IL24 induces inhibition of translation initiation leading to apoptosis in squamous cell carcinoma. A dominant constitutively active mutant of eIF2α, which is resistant to phosphorylation, was used to determine the involvement of eIF2α in IL24-induced apoptosis. Treatment with IL24 resulted in inhibition of protein synthesis, expression of downstream biomarkers of ternary complex depletion such as CHOP, and induction of apoptosis in cancer cells. The constitutively active nonphosphorylatable mutant of eIF2α, eIF2α-S51A, reversed both the IL24-mediated translational block and IL24-induced apoptosis. Intriguingly, IL24 treatment also caused hypophosphorylation of 4E-BP1, which binds to eIF4E with high affinity, thus preventing its association with eIF4G and therefore preventing elF4F complex assembly.Implications: These results demonstrate a previously unrecognized role of IL24 in inhibition of translation, mediated through both phosphorylation of eIF2α and dephosphorylation of 4E-BP1, and provide the first direct evidence for translation control of gene-specific expression by IL24. Mol Cancer Res; 15(8); 1117-24. ©2017 AACR.
Collapse
Affiliation(s)
- Leah Persaud
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, Bronx, New York.,Department of Biology, the Graduate Center, City University of New York, New York, New York
| | - Xuelin Zhong
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, Bronx, New York.,Department of Biology, the Graduate Center, City University of New York, New York, New York
| | - Giselle Alvarado
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, Bronx, New York
| | - Winchie Do
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, Bronx, New York
| | - Jordan Dejoie
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, Bronx, New York
| | - Anna Zybtseva
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, Bronx, New York
| | - Bertal Huseyin Aktas
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Harvard Medical School, Laboratory for Translational Research, Cambridge, Massachusetts
| | - Moira Sauane
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, Bronx, New York. .,Department of Biology, the Graduate Center, City University of New York, New York, New York
| |
Collapse
|
12
|
Guo L, Chi Y, Xue J, Ma L, Shao Z, Wu J. Phosphorylated eIF2α predicts disease-free survival in triple-negative breast cancer patients. Sci Rep 2017; 7:44674. [PMID: 28294178 PMCID: PMC5353635 DOI: 10.1038/srep44674] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022] Open
Abstract
Phosphorylated eukaryotic translation initiation factor 2α (p-eIF2α), which functions as a marker of endoplasmic reticulum stress, has been reported to be associated with patient prognosis in various cancers. However, little is known about the prognostic value of p-eIF2α in breast cancer, particularly in different breast cancer subtypes. An immunohistochemistry screen for p-eIF2α was performed using a tissue microarray containing 233 tumors and paired peritumoral tissues from female patients diagnosed with breast cancer. The staining results were scored semiquantitatively, and the p-eIF2α expression level in breast cancer and its potential prognostic value were investigated. In this retrospective cohort study, we found that p-eIF2α levels were significantly upregulated in breast cancer (P < 0.001). p-eIF2α level was negatively correlated with lymph node status (P = 0.039). Survival analysis by Kaplan–Meier estimation and Cox regression showed that p-eIF2α level was correlated with better disease free survival (P = 0.026) and served as an independent prognostic factor (P = 0.046) in patients with triple-negative breast cancer. Our study revealed that p-eIF2α was upregulated in breast cancer and represented a novel predictor of prognosis in patients with triple-negative subtype.
Collapse
Affiliation(s)
- Liang Guo
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
| | - Yayun Chi
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
| | - Jingyan Xue
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
| | - Linxiaoxi Ma
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
| | - Zhiming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
| | - Jiong Wu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai 200032, PR China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, PR China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
13
|
Xu D, Song R, Wang G, Jeyabal PVS, Weiskoff AM, Ding K, Shi ZZ. Obg-like ATPase 1 regulates global protein serine/threonine phosphorylation in cancer cells by suppressing the GSK3β-inhibitor 2-PP1 positive feedback loop. Oncotarget 2016; 7:3427-39. [PMID: 26655089 PMCID: PMC4823117 DOI: 10.18632/oncotarget.6496] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/21/2015] [Indexed: 11/29/2022] Open
Abstract
OLA1 is an Obg family P-loop NTPase that possesses both GTP- and ATP-hydrolyzing activities. Here we report that OLA1 is a GSK3β interacting protein, and through its ATPase activity, inhibits the GSK3β-mediated activation of protein serine/threonine phosphatase 1 (PP1). It is hypothesized that GSK3β phosphorylates inhibitor 2 (I-2) of PP1 at Thr-72 and activates the PP1 · I-2 complex, which in turn dephosphorylates and stimulates GSK3β, thus forming a positive feedback loop. We revealed that the positive feedback loop is normally suppressed by OLA1, and becomes over-activated under OLA1 deficiency, resulting in increased cellular PP1 activity and dephosphorylation of multiple Ser/Thr phosphoproteins, and more strikingly, decreased global protein threonine phosphorylation. Furthermore, using xenograft models of colon cancer (H116) and ovarian cancer (SKOV3), we established a correlation among downregulation of OLA1, over-activation of the positive feedback loop as indicated by under-phosphorylation of I-2, and more aggressive tumor growth. This study provides the first evidence for the existence of a GSK3β-I-2-PP1 positive feedback loop in human cancer cells, and identifies OLA1 as an endogenous suppressor of this signaling motif.
Collapse
Affiliation(s)
- Dong Xu
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Renduo Song
- Department of Translational Imaging, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Guohui Wang
- Department of Translational Imaging, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Prince V S Jeyabal
- Department of Translational Imaging, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Amanda M Weiskoff
- Department of Translational Imaging, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Kefeng Ding
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Zheng-Zheng Shi
- Department of Translational Imaging, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
14
|
Yalon M, Tuval-Kochen L, Castel D, Moshe I, Mazal I, Cohen O, Avivi C, Rosenblatt K, Aviel-Ronen S, Schiby G, Yahalom J, Amariglio N, Pfeffer R, Lawrence Y, Toren A, Rechavi G, Paglin S. Overcoming Resistance of Cancer Cells to PARP-1 Inhibitors with Three Different Drug Combinations. PLoS One 2016; 11:e0155711. [PMID: 27196668 PMCID: PMC4873128 DOI: 10.1371/journal.pone.0155711] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 05/03/2016] [Indexed: 01/08/2023] Open
Abstract
Inhibitors of poly[ADP-ribose] polymerase 1 (PARPis) show promise for treatment of cancers which lack capacity for homologous recombination repair (HRR). However, new therapeutic strategies are required in order to overcome innate and acquired resistance to these drugs and thus expand the array of cancers that could benefit from them. We show that human cancer cell lines which respond poorly to ABT-888 (a PARPi), become sensitive to it when co-treated with vorinostat (a histone deacetylase inhibitor (HDACi)). Vorinostat also sensitized PARPis insensitive cancer cell lines to 6-thioguanine (6-TG)–a drug that targets PARPis sensitive cells. The sensitizing effect of vorinostat was associated with increased phosphorylation of eukaryotic initiation factor (eIF) 2α which in and of itself increases the sensitivity of cancer cells to ABT-888. Importantly, these drug combinations did not affect survival of normal fibroblasts and breast cells, and significantly increased the inhibition of xenograft tumor growth relative to each drug alone, without affecting the mice weight or their liver and kidney function. Our results show that combination of vorinostat and ABT-888 could potentially prove useful for treatment of cancer with innate resistance to PARPis due to active HRR machinery, while the combination of vorinostat and 6-TG could potentially overcome innate or acquired resistance to PARPis due to secondary or reversal BRCA mutations, to decreased PARP-1 level or to increased expression of multiple drug resistant proteins. Importantly, drugs which increase phosphorylation of eIF2α may mimic the sensitizing effect of vorinostat on cellular response to PARPis or to 6-TG, without activating all of its downstream effectors.
Collapse
Affiliation(s)
- Michal Yalon
- Department of Pediatric Hematology-Oncology, Safra Children's Hospital, Sheba Medical Center, Ramat Gan 52621, Israel
- Cancer Research Center, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Liron Tuval-Kochen
- Cancer Research Center, Sheba Medical Center, Ramat-Gan 52621, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - David Castel
- Neufeld Cardiac Research Institute, Sheba Medical Center, Ramat Gan 52621, Israel
| | - Itai Moshe
- Department of Pediatric Hematology-Oncology, Safra Children's Hospital, Sheba Medical Center, Ramat Gan 52621, Israel
| | - Inbal Mazal
- Cancer Research Center, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Osher Cohen
- Department of Surgery, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Camila Avivi
- Department of Pathology, Sheba Medical Center, Ramat-Gan 52621, Israel
| | | | - Sarit Aviel-Ronen
- Department of Pathology, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Ginette Schiby
- Department of Pathology, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Joachim Yahalom
- Department of Radiation Oncology, Memorial Sloan Kettering, New York 10021, United States of America
| | - Ninette Amariglio
- Cancer Research Center, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Raphael Pfeffer
- Cancer Research Center, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Yaacov Lawrence
- Cancer Research Center, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Amos Toren
- Department of Pediatric Hematology-Oncology, Safra Children's Hospital, Sheba Medical Center, Ramat Gan 52621, Israel
- Cancer Research Center, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Gideon Rechavi
- Cancer Research Center, Sheba Medical Center, Ramat-Gan 52621, Israel
| | - Shoshana Paglin
- Cancer Research Center, Sheba Medical Center, Ramat-Gan 52621, Israel
- * E-mail:
| |
Collapse
|
15
|
Gao S, Zhou F, Zhao C, Ma Z, Jia R, Liang S, Zhang M, Zhu X, Zhang P, Wang L, Su F, Zhao J, Liu G, Peng B, Feng X. Gastric cardia adenocarcinoma microRNA profiling in Chinese patients. Tumour Biol 2016; 37:9411-22. [PMID: 26781873 DOI: 10.1007/s13277-016-4824-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/08/2016] [Indexed: 12/13/2022] Open
Abstract
Gastric cardia adenocarcinoma (GCA), which occurs at the gastroesophageal boundary, is one of the most malignant types of cancer. Over the past 30 years, the incidence of GCA has increased by approximately sevenfold, which has a more substantial increase than that of many other malignancies. However, as previous studies mainly focus on non-cardia gastric cancer, until now, the mechanisms behind GCA remain largely unknown. MicroRNAs (miRNAs) have been shown to play pivotal roles in carcinogenesis. To gain insight into the molecular mechanisms regulated by miRNAs in GCA development, we investigated miRNA expression profiles using 81 pairs of primary GCAs and corresponding non-tumorigenic tissues. First, 21 pairs of samples were used for microarray analysis, and then another 60 pairs of samples were used for further analysis. Our results showed that 464 miRNAs (237 upregulated, 227 downregulated, false discovery rate FDR <0.05) were differently expressed between GCA and non-tumor tissues. Pearson test and pathway analysis revealed that these dysregulated miRNA correlated coding RNAs may have effects on several cancer-related pathways. Four miRNAs (miR-1244, miR-135b-5p, miR-3196, and miR-628-3p) were found to be associated with GCA differentiation. One miRNA, miR-196a-5p, was found to be associated with age of GCA onset. Further, survival analysis showed that the expression level of miR-135b-5p was associated with GCA survival. Taken together, our study first provided the genome-wide expression profiles of miRNA in GCA and will be good help for further functional studies.
Collapse
Affiliation(s)
- Shegan Gao
- Henan Key Laboratory of Cancer Epigenetic; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003, Luoyang, 471003, China
| | - Fuyou Zhou
- Department of Oncology, Anyang People's Hospital, Anyang, 471500, China
| | - Chen Zhao
- Fudan-Zhangjiang Center for Clinical Genomics, Zuchongzhi Road 899, Shanghai, 201203, China
| | - Zhikun Ma
- Henan Key Laboratory of Cancer Epigenetic; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003, Luoyang, 471003, China
| | - Ruinuo Jia
- Henan Key Laboratory of Cancer Epigenetic; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003, Luoyang, 471003, China
| | - Shuo Liang
- Henan Key Laboratory of Cancer Epigenetic; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003, Luoyang, 471003, China
| | - Mengxi Zhang
- Henan Key Laboratory of Cancer Epigenetic; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003, Luoyang, 471003, China
| | - Xiaojuan Zhu
- Henan Key Laboratory of Cancer Epigenetic; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003, Luoyang, 471003, China
| | - Pengfei Zhang
- Henan Key Laboratory of Cancer Epigenetic; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003, Luoyang, 471003, China
| | - Lu Wang
- Zhangjiang Center for Translational Medicine, Zuchongzhi Road 899, Shanghai, 201203, China
| | - Feng Su
- Zhangjiang Center for Translational Medicine, Zuchongzhi Road 899, Shanghai, 201203, China
| | - Jiangman Zhao
- Zhangjiang Center for Translational Medicine, Zuchongzhi Road 899, Shanghai, 201203, China
| | - Gang Liu
- Henan Key Laboratory of Cancer Epigenetic; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003, Luoyang, 471003, China
| | - Bo Peng
- Zhangjiang Center for Translational Medicine, Zuchongzhi Road 899, Shanghai, 201203, China.
| | - Xiaoshan Feng
- Henan Key Laboratory of Cancer Epigenetic; Cancer Institute, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China, 471003, Luoyang, 471003, China. .,Henan University of Science and Technology, Jing hua Road 24, Luoyang, 471500, China.
| |
Collapse
|
16
|
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
|
17
|
Aktas BH, Bordelois P, Peker S, Merajver S, Halperin JA. Depletion of eIF2·GTP·Met-tRNAi translation initiation complex up-regulates BRCA1 expression in vitro and in vivo. Oncotarget 2016; 6:6902-14. [PMID: 25762631 PMCID: PMC4466658 DOI: 10.18632/oncotarget.3125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/09/2015] [Indexed: 01/27/2023] Open
Abstract
Most sporadic breast and ovarian cancers express low levels of the breast cancer susceptibility gene, BRCA1. The BRCA1 gene produces two transcripts, mRNAa and mRNAb. mRNAb, present in breast cancer but not in normal mammary epithelial cells, contains three upstream open reading frames (uORFs) in its 5′UTR and is translationally repressed. Comparable tandem uORFs are characteristically seen in mRNAs whose translational efficiency paradoxically increases when the overall translation rate is decreased due to phosphorylation of eukaryotic translation initiation factor 2 α (eIF2α). Here we show fish oil derived eicosopanthenoic acid (EPA) that induces eIF2α phosphorylation translationally up-regulates the expression of BRCA1 in human breast cancer cells. We demonstrate further that a diet rich in EPA strongly induces expression of BRCA1 in human breast cancer xenografts.
Collapse
Affiliation(s)
- Bertal H Aktas
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | - Selen Peker
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA.,Ankara University Biotechnology Institute, Ankara, Turkey
| | - Sophia Merajver
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jose A Halperin
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
An inhibitor of HIV-1 protease modulates constitutive eIF2α dephosphorylation to trigger a specific integrated stress response. Proc Natl Acad Sci U S A 2015; 113:E117-26. [PMID: 26715744 DOI: 10.1073/pnas.1514076113] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inhibitors of the HIV aspartyl protease [HIV protease inhibitors (HIV-PIs)] are the cornerstone of treatment for HIV. Beyond their well-defined antiretroviral activity, these drugs have additional effects that modulate cell viability and homeostasis. However, little is known about the virus-independent pathways engaged by these molecules. Here we show that the HIV-PI Nelfinavir decreases translation rates and promotes a transcriptional program characteristic of the integrated stress response (ISR). Mice treated with Nelfinavir display hallmarks of this stress response in the liver, including α subunit of translation initiation factor 2 (eIF2α) phosphorylation, activating transcription factor-4 (ATF4) induction, and increased expression of known downstream targets. Mechanistically, Nelfinavir-mediated ISR bypassed direct activation of the eIF2α stress kinases and instead relied on the inhibition of the constitutive eIF2α dephosphorylation and down-regulation of the phophatase cofactor CReP (Constitutive Repressor of eIF2α Phosphorylation; also known as PPP1R15B). These findings demonstrate that the modulation of eIF2α-specific phosphatase cofactor activity can be a rheostat of cellular homeostasis that initiates a functional ISR and suggest that the HIV-PIs could be repositioned as therapeutics in human diseases to modulate translation rates and stress responses.
Collapse
|
19
|
OLA1 regulates protein synthesis and integrated stress response by inhibiting eIF2 ternary complex formation. Sci Rep 2015; 5:13241. [PMID: 26283179 PMCID: PMC4539610 DOI: 10.1038/srep13241] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 07/10/2015] [Indexed: 01/06/2023] Open
Abstract
Translation is a fundamental cellular process, and its dysregulation can contribute to human diseases such as cancer. During translation initiation the eukaryotic initiation factor 2 (eIF2) forms a ternary complex (TC) with GTP and the initiator methionyl-tRNA (tRNAi), mediating ribosomal recruitment of tRNAi. Limiting TC availability is a central mechanism for triggering the integrated stress response (ISR), which suppresses global translation in response to various cellular stresses, but induces specific proteins such as ATF4. This study shows that OLA1, a member of the ancient Obg family of GTPases, is an eIF2-regulatory protein that inhibits protein synthesis and promotes ISR by binding eIF2, hydrolyzing GTP, and interfering with TC formation. OLA1 thus represents a novel mechanism of translational control affecting de novo TC formation, different from the traditional model in which phosphorylation of eIF2α blocks the regeneration of TC. Depletion of OLA1 caused a hypoactive ISR and greater survival in stressed cells. In vivo, OLA1-knockdown rendered cancer cells deficient in ISR and the downstream proapoptotic effector, CHOP, promoting tumor growth and metastasis. Our work suggests that OLA1 is a novel translational GTPase and plays a suppressive role in translation and cell survival, as well as cancer growth and progression.
Collapse
|
20
|
Mbatia HW, Ramalingam S, Ramamurthy VP, Martin MS, Kwegyir-Afful AK, Njar VCO. Novel C-4 heteroaryl 13-cis-retinamide Mnk/AR degrading agents inhibit cell proliferation and migration and induce apoptosis in human breast and prostate cancer cells and suppress growth of MDA-MB-231 human breast and CWR22Rv1 human prostate tumor xenografts in mice. J Med Chem 2015; 58:1900-14. [PMID: 25634130 DOI: 10.1021/jm501792c] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The synthesis and in vitro and in vivo antibreast and antiprostate cancers activities of novel C-4 heteroaryl 13-cis-retinamides that modulate Mnk-eIF4E and AR signaling are discussed. Modifications of the C-4 heteroaryl substituents reveal that the 1H-imidazole is essential for high anticancer activity. The most potent compounds against a variety of human breast and prostate cancer (BC/PC) cell lines were compounds 16 (VNHM-1-66), 20 (VNHM-1-81), and 22 (VNHM-1-73). In these cell lines, the compounds induce Mnk1/2 degradation to substantially suppress eIF4E phosphorylation. In PC cells, the compounds induce degradation of both full-length androgen receptor (fAR) and splice variant AR (AR-V7) to inhibit AR transcriptional activity. More importantly, VNHM-1-81 has strong in vivo antibreast and antiprostate cancer activities, while VNHM-1-73 exhibited strong in vivo antibreast cancer activity, with no apparent host toxicity. Clearly, these lead compounds are strong candidates for development for the treatments of human breast and prostate cancers.
Collapse
Affiliation(s)
- Hannah W Mbatia
- Department of Pharmacology, ‡Center for Biomolecular Therapeutics, and §Marlene Stewart Greenebaum Cancer Center, University of Maryland School of Medicine , 685 West Baltimore Street, Baltimore, Maryland 21201-1559, United States
| | | | | | | | | | | |
Collapse
|
21
|
Shi X, Sun M, Wu Y, Yao Y, Liu H, Wu G, Yuan D, Song Y. Post-transcriptional regulation of long noncoding RNAs in cancer. Tumour Biol 2015; 36:503-13. [PMID: 25618601 DOI: 10.1007/s13277-015-3106-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/12/2015] [Indexed: 12/15/2022] Open
Abstract
It is a great surprise that the genomes of mammals and other eukaryotes harbor many thousands of long noncoding RNAs (lncRNAs). Although these long noncoding transcripts were once considered to be simply transcriptional noise or cloning artifacts, multiple studies have suggested that lncRNAs are emerging as new players in diverse human diseases, especially in cancer, and that the molecular mechanisms of lncRNAs need to be elucidated. More recently, evidence has begun to accumulate describing the complex post-transcriptional regulation in which lncRNAs are involved. It was reported that lncRNAs can be implicated in degradation, translation, pre-messenger RNA (mRNA) splicing, and protein activities and even as microRNAs (miRNAs) sponges in both a sequence-dependent and sequence-independent manner. In this review, we present an updated vision of lncRNAs and summarize the mechanism of post-transcriptional regulation by lncRNAs, providing new insight into the functional cellular roles that they may play in human diseases, with a particular focus on cancers.
Collapse
Affiliation(s)
- Xuefei Shi
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu Province, China,
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Translational regulator eIF2α in tumor. Tumour Biol 2014; 35:6255-64. [DOI: 10.1007/s13277-014-1789-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/21/2014] [Indexed: 11/29/2022] Open
|
23
|
Abstract
Although clotrimazole was first used against fungal infections, a body of research was later developed indicating that this drug has anticancer properties as well. The mechanism of action is based on the inhibition of mitochondrial-bound glycolytic enzymes and calmodulin, which starves cancer cells of energy. Clotrimazole and its derivatives have been shown to decrease rates of cancer cell proliferation, induce G1 phase arrest, and promote pro-apoptotic factors, which lead to cell death.
Collapse
Affiliation(s)
- S Kadavakollu
- Department of Natural Sciences, Western New Mexico University, Silver City, NM, 88061, USA
| | - C Stailey
- Department of Natural Sciences, Western New Mexico University, Silver City, NM, 88061, USA
| | - C S Kunapareddy
- Department of Natural Sciences, Western New Mexico University, Silver City, NM, 88061, USA
| | - S White
- Department of Natural Sciences, Western New Mexico University, Silver City, NM, 88061, USA
| |
Collapse
|
24
|
Chen T, Takrouri K, Hee-Hwang S, Rana S, Yefidoff-Freedman R, Halperin J, Natarajan A, Morisseau C, Hammock B, Chorev M, Aktas BH. Explorations of substituted urea functionality for the discovery of new activators of the heme-regulated inhibitor kinase. J Med Chem 2013; 56:9457-70. [PMID: 24261904 DOI: 10.1021/jm400793v] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Heme-regulated inhibitor kinase (HRI), a eukaryotic translation initiation factor 2 alpha (eIF2α) kinase, plays critical roles in cell proliferation, differentiation, and adaptation to cytoplasmic stress. HRI is also a critical modifier of hemoglobin disorders such as β-thalassemia. We previously identified N,N'-diarylureas as potent activators of HRI suitable for studying the biology of this important kinase. To expand the repertoire of chemotypes that activate HRI, we screened a ∼1900 member N,N'-disubstituted urea library in the surrogate eIF2α phosphorylation assay, identifying N-aryl,N'-cyclohexylphenoxyurea as a promising scaffold. We validated hit compounds as a bona fide HRI activators in secondary assays and explored the contributions of substitutions on the N-aryl and N'-cyclohexylphenoxy groups to their activity by studying focused libraries of complementing analogues. We tested these N-aryl,N'-cyclohexylphenoxyureas in the surrogate eIF2α phosphorylation and cell proliferation assays, demonstrating significantly improved bioactivities and specificities. We consider these compounds to represent lead candidates for the development of potent and specific HRI activators.
Collapse
Affiliation(s)
- Ting Chen
- Hematology Laboratory for Translational Research, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , 75 Francis Street, Boston, Massachusetts 02115, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|