1
|
Aktepe TE, Deerain JM, Hyde JL, Fritzlar S, Mead EM, Carrera Montoya J, Hachani A, Pearson JS, White PA, Mackenzie JM. Norovirus-mediated translation repression promotes macrophage cell death. PLoS Pathog 2024; 20:e1012480. [PMID: 39226332 PMCID: PMC11398682 DOI: 10.1371/journal.ppat.1012480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/13/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
Norovirus infection is characterised by a rapid onset of disease and the development of debilitating symptoms including projectile vomiting and diffuse diarrhoea. Vaccines and antivirals are sorely lacking and developments in these areas are hampered by the lack of an adequate cell culture system to investigate human norovirus replication and pathogenesis. Herein, we describe how the model norovirus, Mouse norovirus (MNV), produces a viral protein, NS3, with the functional capacity to attenuate host protein translation which invokes the activation of cell death via apoptosis. We show that this function of NS3 is conserved between human and mouse viruses and map the protein domain attributable to this function. Our study highlights a critical viral protein that mediates crucial activities during replication, potentially identifying NS3 as a worthy target for antiviral drug development.
Collapse
Affiliation(s)
- Turgut E Aktepe
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Joshua M Deerain
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jennifer L Hyde
- Department of Microbiology, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Svenja Fritzlar
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Eleanor M Mead
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Julio Carrera Montoya
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jaclyn S Pearson
- The Hudson Institute of Medical Research, Centre for Innate Immunity and Infectious Diseases, Melbourne, Australia
| | - Peter A White
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| |
Collapse
|
2
|
Wu Q, Jin Y, Li S, Guo X, Sun W, Liu J, Li Q, Niu D, Zou Y, Du X, Li Y, Zhao T, Li Z, Li X, Ren G. Oncolytic Newcastle disease virus carrying the IL24 gene exerts antitumor effects by inhibiting tumor growth and vascular sprouting. Int Immunopharmacol 2024; 136:112305. [PMID: 38823178 DOI: 10.1016/j.intimp.2024.112305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/29/2024] [Accepted: 05/16/2024] [Indexed: 06/03/2024]
Abstract
The second-leading cause of death, cancer, poses a significant threat to human life. Innovations in cancer therapies are crucial due to limitations in traditional approaches. Newcastle disease virus (NDV), a nonpathogenic oncolytic virus, exhibits multifunctional anticancer properties by selectively infecting, replicating, and eliminating tumor cells. To enhance NDV's antitumor activity, four oncolytic NDV viruses were developed, incorporating IL24 and/or GM-CSF genes at different gene loci using reverse genetics. In vitro experiments revealed that oncolytic NDV virus augmented the antitumor efficacy of the parental virus rClone30, inhibiting tumor cell proliferation, inducing tumor cell fusion, and promoting apoptosis. Moreover, NDV carrying the IL24 gene inhibited microvessel formation in CAM experiments. Evaluation in a mouse model of liver cancer confirmed the therapeutic efficacy of oncolytic NDV viral therapy. Tumors in mice treated with oncolytic NDV virus significantly decreased in size, accompanied by tumor cell detachment and apoptosis evident in pathological sections. Furthermore, oncolytic NDV virus enhanced T cell and dendritic cell production and substantially improved the survival rate of mice with hepatocellular carcinoma, with rClone30-IL24(P/M) demonstrating significant therapeutic effects. This study establishes a basis for utilizing oncolytic NDV virus as an antitumor agent in clinical practice.
Collapse
Affiliation(s)
- Qing Wu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Yuhan Jin
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Shuang Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xiaochen Guo
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Wenying Sun
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Jinmiao Liu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Qianhui Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Dun Niu
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Yimeng Zou
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xin Du
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Yanan Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Tianqi Zhao
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhitong Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xinyu Li
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Guiping Ren
- Biopharmaceutical Lab, College of Life Science, Northeast Agricultural University, Harbin 150030, China; Research Center of Genetic Engineering of Pharmaceuticals of Heilongjiang Province, Northeast Agricultural University, Harbin 150030, China; Key Laboratory of Agricultural Biological Functional Gene, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
3
|
Di T, Luo QY, Song JT, Yan XL, Zhang L, Pan WT, Guo Y, Lu FT, Sun YT, Xia ZF, Yang LQ, Qiu MZ, Yang DJ, Sun J. APG-1252 combined with Cabozantinib inhibits hepatocellular carcinoma by suppressing MEK/ERK and CREB/Bcl-xl pathways. Int Immunopharmacol 2024; 139:112615. [PMID: 39032475 DOI: 10.1016/j.intimp.2024.112615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/30/2024] [Accepted: 06/30/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND AND PURPOSE Liver cancer is the fourth leading cause of cancer-related death worldwide, with hepatocellular carcinoma (HCC) being the most common type of primary liver cancer. APG-1252 is a small molecule inhibitor targeting Bcl-2 and Bcl-xl. However, its anti-tumor effects in HCC, alone or in combination with Cabozantinib, have not been extensively studied. EXPERIMENTAL Approach: TCGA database analysis was used to analysis the gene expression levels of Bcl-2 and Bcl-xl in HCC tissues. Western blot was employed to detect the protein expression levels. And the inhibitory effects of APG-1252 and Cabozantinib on the proliferation of HCC cell lines was detected by CCK-8. The effect on the migration and invasion of HCC cells was verified by transwell assay. Huh7 xenograft model in nude mice was used to investigate the combination antitumor effect in vivo. KEY RESULTS Our study demonstrated that APG-1252 monotherapy inhibited the proliferation and migration ability of HCC cells, and induced HCC cells apoptosis. The combination of APG-1252 and Cabozantinib showed significant synergistic antitumor effects. Furthermore, the in vivo experiment demonstrated that the combination therapy exerted a synergistic effect in delaying tumor growth, notably downregulating MEK/ERK phosphorylation levels. In terms of mechanism, Cabozantinib treatment caused an increase in the phosphorylation levels of CREB and Bcl-xl proteins, while the combination with APG-1252 mitigated this effect, thereby enhanced the antitumor effect of Cabozantinib. CONCLUSION AND IMPLICATIONS Our findings suggest that APG-1252 in combination with Cabozantinib offers a more effective treatment strategy for HCC patients, warranting further clinical investigation.
Collapse
Affiliation(s)
- Tian Di
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Qiu-Yun Luo
- Department of Clinical Research, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jiang-Tao Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiang-Lei Yan
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden; Center of Molecular Medicine, Stockholm, Sweden
| | - Lin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Wen-Tao Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yu Guo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Fei-Teng Lu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yu-Ting Sun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zeng-Fei Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Qiong Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Miao-Zhen Qiu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China; Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Da-Jun Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China; Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Jian Sun
- Department of Clinical Research, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
4
|
Hengst JA, Nduwumwami AJ, Sharma A, Yun JK. Fanning the Flames of Endoplasmic Reticulum (ER) Stress: Can Sphingolipid Metabolism Be Targeted to Enhance ER Stress-Associated Immunogenic Cell Death in Cancer? Mol Pharmacol 2024; 105:155-165. [PMID: 38164594 PMCID: PMC10877730 DOI: 10.1124/molpharm.123.000786] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/03/2024] Open
Abstract
The three arms of the unfolded protein response (UPR) surveil the luminal environment of the endoplasmic reticulum (ER) and transmit information through the lipid bilayer to the cytoplasm to alert the cell of stress conditions within the ER lumen. That same lipid bilayer is the site of de novo synthesis of phospholipids and sphingolipids. Thus, it is no surprise that lipids are modulated by and are modulators of ER stress. Given that sphingolipids have both prosurvival and proapoptotic effects, they also exert opposing effects on life/death decisions in the face of prolonged ER stress detected by the UPR. In this review, we will focus on several recent studies that demonstrate how sphingolipids affect each arm of the UPR. We will also discuss the role of sphingolipids in the process of immunogenic cell death downstream of the protein kinase RNA-like endoplasmic reticulum kinase (PERK)/eukaryotic initiating factor 2α (eIF2α) arm of the UPR. Furthermore, we will discuss strategies to target the sphingolipid metabolic pathway that could potentially act synergistically with agents that induce ER stress as novel anticancer treatments. SIGNIFICANCE STATEMENT: This review provides the readers with a brief discussion of the sphingolipid metabolic pathway and the unfolded protein response. The primary focus of the review is the mechanism(s) by which sphingolipids modulate the endoplasmic reticulum (ER) stress response pathways and the critical role of sphingolipids in the process of immunogenic cell death associated with the ER stress response.
Collapse
Affiliation(s)
- Jeremy A Hengst
- Departments of Pediatrics (J.A.H.) and Pharmacology (A.S., J.K.Y.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Drug Metabolism and Pharmacokinetics, National Center for Advancing Translational Science, Rockville, Maryland (A.J.N.)
| | - Asvelt J Nduwumwami
- Departments of Pediatrics (J.A.H.) and Pharmacology (A.S., J.K.Y.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Drug Metabolism and Pharmacokinetics, National Center for Advancing Translational Science, Rockville, Maryland (A.J.N.)
| | - Arati Sharma
- Departments of Pediatrics (J.A.H.) and Pharmacology (A.S., J.K.Y.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Drug Metabolism and Pharmacokinetics, National Center for Advancing Translational Science, Rockville, Maryland (A.J.N.)
| | - Jong K Yun
- Departments of Pediatrics (J.A.H.) and Pharmacology (A.S., J.K.Y.), Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Drug Metabolism and Pharmacokinetics, National Center for Advancing Translational Science, Rockville, Maryland (A.J.N.)
| |
Collapse
|
5
|
Schwarzer A, Oliveira M, Kleppa MJ, Slattery SD, Anantha A, Cooper A, Hannink M, Schambach A, Dörrie A, Kotlyarov A, Gaestel M, Hembrough T, Levine J, Luther M, Stocum M, Stiles L, Weinstock DM, Liesa M, Kostura MJ. Targeting Aggressive B-cell Lymphomas through Pharmacological Activation of the Mitochondrial Protease OMA1. Mol Cancer Ther 2023; 22:1290-1303. [PMID: 37643767 PMCID: PMC10723637 DOI: 10.1158/1535-7163.mct-22-0718] [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: 11/22/2022] [Revised: 07/02/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
DLBCL are aggressive, rapidly proliferating tumors that critically depend on the ATF4-mediated integrated stress response (ISR) to adapt to stress caused by uncontrolled growth, such as hypoxia, amino acid deprivation, and accumulation of misfolded proteins. Here, we show that ISR hyperactivation is a targetable liability in DLBCL. We describe a novel class of compounds represented by BTM-3528 and BTM-3566, which activate the ISR through the mitochondrial protease OMA1. Treatment of tumor cells with compound leads to OMA1-dependent cleavage of DELE1 and OPA1, mitochondrial fragmentation, activation of the eIF2α-kinase HRI, cell growth arrest, and apoptosis. Activation of OMA1 by BTM-3528 and BTM-3566 is mechanistically distinct from inhibitors of mitochondrial electron transport, as the compounds induce OMA1 activity in the absence of acute changes in respiration. We further identify the mitochondrial protein FAM210B as a negative regulator of BTM-3528 and BTM-3566 activity. Overexpression of FAM210B prevents both OMA1 activation and apoptosis. Notably, FAM210B expression is nearly absent in healthy germinal center B-lymphocytes and in derived B-cell malignancies, revealing a fundamental molecular vulnerability which is targeted by BTM compounds. Both compounds induce rapid apoptosis across diverse DLBCL lines derived from activated B-cell, germinal center B-cell, and MYC-rearranged lymphomas. Once-daily oral dosing of BTM-3566 resulted in complete regression of xenografted human DLBCL SU-DHL-10 cells and complete regression in 6 of 9 DLBCL patient-derived xenografts. BTM-3566 represents a first-of-its kind approach of selectively hyperactivating the mitochondrial ISR for treating DLBCL.
Collapse
Affiliation(s)
- Adrian Schwarzer
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostaseology, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Matheus Oliveira
- Department of Medicine, Endocrinology, UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Marc-Jens Kleppa
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | - Andy Anantha
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Alan Cooper
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Mark Hannink
- Biochemistry Department, Life Sciences Center and Ellis Fischel Cancer Center, University of Missouri, Columbia, Missouri
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Anneke Dörrie
- Department of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Alexey Kotlyarov
- Department of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Matthias Gaestel
- Department of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Todd Hembrough
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Jedd Levine
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Michael Luther
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Michael Stocum
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Linsey Stiles
- Department of Medicine, Endocrinology, UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Marc Liesa
- Department of Medicine, Endocrinology, UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona, Catalonia, Spain
| | | |
Collapse
|
6
|
Tantawy SI, Sarkar A, Hubner S, Tan Z, Wierda WG, Eldeib A, Zhang S, Kornblau S, Gandhi V. Mechanisms of MCL-1 Protein Stability Induced by MCL-1 Antagonists in B-Cell Malignancies. Clin Cancer Res 2023; 29:446-457. [PMID: 36346691 PMCID: PMC9852224 DOI: 10.1158/1078-0432.ccr-22-2088] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/19/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE Several MCL-1 inhibitors (MCL-1i), including AMG-176 and AZD5991, have shown promise in preclinical studies and are being tested for the treatment of hematologic malignancies. A unique feature of these agents is induction and stability of Mcl-1 protein; however, the precise mechanism is unknown. We aim to study the mechanism of MCL-1i-induced Mcl-1 protein stability. EXPERIMENTAL DESIGN Using several B-cell leukemia and lymphoma cell lines and primary chronic lymphocytic leukemia (CLL) lymphocytes, we evaluated molecular events associated with Mcl-1 protein stability including protein half-life, reverse-phase protein array, protein-protein interaction, phosphorylation, ubiquitination, and de-ubiquitination, followed by molecular simulation and modeling. RESULTS Using both in vivo and in vitro analysis, we demonstrate that MCL-1i-induced Mcl-1 protein stability is predominantly associated with defective Mcl-1 ubiquitination and concurrent apoptosis induction in both cell lines and primary CLL subjects. These MCL1i also induced ERK-mediated Mcl-1Thr163 phosphorylation, which partially contributed to Mcl-1 stability. Disruption of Mcl-1:Noxa interaction followed by Noxa degradation, enhanced Mcl-1 de-ubiquitination by USP9x, and Mule destabilization are the major effects of these inhibitors. However, unlike other BH3 proteins, Mule:Mcl-1 interaction was unaffected by MCL-1i. WP1130, a global deubiquitinase (DUB) inhibitor, abrogated Mcl-1 induction reaffirming a critical role of DUBs in the observed Mcl-1 protein stability. Further, in vitro ubiquitination studies of Mcl-1 showed distinct difference among these inhibitors. CONCLUSIONS We conclude that MCL-1i blocked Mcl-1 ubiquitination via enhanced de-ubiquitination and dissociation of Mcl-1 from Noxa, Bak and Bax, and Mule de-stabilization. These are critical events associated with increased Mcl-1 protein stability with AMG-176 and AZD5991.
Collapse
Affiliation(s)
- Shady I. Tantawy
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas,Department of Internal Medicine, College of Medicine, Suez Canal University, Ismailia, Egypt
| | - Aloke Sarkar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan Hubner
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zhi Tan
- Center for Drug Discovery, Department of Pathology and Immunology, Department of Pharmacology and Chemical Biology, Baylor College of Medicine
| | - William G. Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abdelraouf Eldeib
- Center for Drug Discovery, Department of Pathology and Immunology, Department of Pharmacology and Chemical Biology, Baylor College of Medicine
| | - Shuxing Zhang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven Kornblau
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas,Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
7
|
Sancho M, Leiva D, Lucendo E, Orzáez M. Understanding MCL1: from cellular function and regulation to pharmacological inhibition. FEBS J 2022; 289:6209-6234. [PMID: 34310025 PMCID: PMC9787394 DOI: 10.1111/febs.16136] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/09/2021] [Accepted: 07/22/2021] [Indexed: 12/30/2022]
Abstract
Myeloid cell leukemia-1 (MCL1), an antiapoptotic member of the BCL2 family characterized by a short half-life, functions as a rapid sensor that regulates cell death and other relevant processes that include cell cycle progression and mitochondrial homeostasis. In cancer, MCL1 overexpression contributes to cell survival and resistance to diverse chemotherapeutic agents; for this reason, several MCL1 inhibitors are currently under preclinical and clinical development for cancer treatment. However, the nonapoptotic functions of MCL1 may influence their therapeutic potential. Overall, the complexity of MCL1 regulation and function represent challenges to the clinical application of MCL1 inhibitors. We now summarize the current knowledge regarding MCL1 structure, regulation, and function that could impact the clinical success of MCL1 inhibitors.
Collapse
Affiliation(s)
- Mónica Sancho
- Targeted Therapies on Cancer and Inflammation LaboratoryCentro de Investigación Príncipe FelipeValenciaSpain
| | - Diego Leiva
- Targeted Therapies on Cancer and Inflammation LaboratoryCentro de Investigación Príncipe FelipeValenciaSpain
| | - Estefanía Lucendo
- Targeted Therapies on Cancer and Inflammation LaboratoryCentro de Investigación Príncipe FelipeValenciaSpain
| | - Mar Orzáez
- Targeted Therapies on Cancer and Inflammation LaboratoryCentro de Investigación Príncipe FelipeValenciaSpain
| |
Collapse
|
8
|
Manni S, Pesavento M, Spinello Z, Saggin L, Arjomand A, Fregnani A, Quotti Tubi L, Scapinello G, Gurrieri C, Semenzato G, Trentin L, Piazza F. Protein Kinase CK2 represents a new target to boost Ibrutinib and Venetoclax induced cytotoxicity in mantle cell lymphoma. Front Cell Dev Biol 2022; 10:935023. [PMID: 36035991 PMCID: PMC9403710 DOI: 10.3389/fcell.2022.935023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an incurable B cell non-Hodgkin lymphoma, characterized by frequent relapses. In the last decade, the pro-survival pathways related to BCR signaling and Bcl-2 have been considered rational therapeutic targets in B cell derived lymphomas. The BTK inhibitor Ibrutinib and the Bcl-2 inhibitor Venetoclax are emerging as effective drugs for MCL. However, primary and acquired resistance also to these agents may occur. Protein Kinase CK2 is a S/T kinase overexpressed in many solid and blood-derived tumours. CK2 promotes cancer cell growth and clonal expansion, sustaining pivotal survival signaling cascades, such as the ones dependent on AKT, NF-κB, STAT3 and others, counteracting apoptosis through a “non-oncogene” addiction mechanism. We previously showed that CK2 is overexpressed in MCL and regulates the levels of activating phosphorylation on S529 of the NF-κB family member p65/RelA. In the present study, we investigated the effects of CK2 inactivation on MCL cell proliferation, survival and apoptosis and this kinase’s involvement in the BCR and Bcl-2 related signaling. By employing CK2 loss of function MCL cell models, we demonstrated that CK2 sustains BCR signaling (such as BTK, NF-κB and AKT) and the Bcl-2-related Mcl-1 expression. CK2 inactivation enhanced Ibrutinib and Venetoclax-induced cytotoxicity. The demonstration of a CK2-dependent upregulation of pathways that may antagonize the effect of these drugs may offer a novel strategy to overcome primary and secondary resistance.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
- *Correspondence: Sabrina Manni, ; Francesco Piazza,
| | - Maria Pesavento
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Zaira Spinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Lara Saggin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Arash Arjomand
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Anna Fregnani
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Greta Scapinello
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Carmela Gurrieri
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Livio Trentin
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
| | - Francesco Piazza
- Department of Medicine-DIMED, Hematology and Clinical Immunology Section, University of Padova, Padova, Italy
- Myeloma and Lymphoma Pathobiology Lab, Veneto Institute of Molecular Medicine, Padova, Italy
- *Correspondence: Sabrina Manni, ; Francesco Piazza,
| |
Collapse
|
9
|
Wirth M. Targeting the translational machinery to overcome apoptosis resistance in pancreatic cancer. Transl Oncol 2022; 17:101343. [PMID: 35078019 PMCID: PMC8790655 DOI: 10.1016/j.tranon.2022.101343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 01/29/2023] Open
Affiliation(s)
- Matthias Wirth
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
10
|
NOXA expression drives synthetic lethality to RUNX1 inhibition in pancreatic cancer. Proc Natl Acad Sci U S A 2022; 119:2105691119. [PMID: 35197278 PMCID: PMC8892327 DOI: 10.1073/pnas.2105691119] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2021] [Indexed: 01/18/2023] Open
Abstract
Recent evidence demonstrated the existence of molecular subtypes in pancreatic ductal adenocarcinoma (PDAC), which resist all current therapies. The paucity of therapeutic options, including a complete lack of targeted therapies, underscores the urgent and unmet medical need for the identification of targets and novel treatment strategies for PDAC. Our study unravels a function of the transcription factor RUNX1 in apoptosis regulation in PDAC. We show that pharmacological RUNX1 inhibition in PDAC is feasible and leads to NOXA-dependent apoptosis. The development of targeted therapies that influence the transcriptional landscape of PDAC might have great benefits for patients who are resistant to conventional therapies. RUNX1 inhibition as a new therapeutic intervention offers an attractive strategy for future therapies. Evasion from drug-induced apoptosis is a crucial mechanism of cancer treatment resistance. The proapoptotic protein NOXA marks an aggressive pancreatic ductal adenocarcinoma (PDAC) subtype. To identify drugs that unleash the death-inducing potential of NOXA, we performed an unbiased drug screening experiment. In NOXA-deficient isogenic cellular models, we identified an inhibitor of the transcription factor heterodimer CBFβ/RUNX1. By genetic gain and loss of function experiments, we validated that the mode of action depends on RUNX1 and NOXA. Of note is that RUNX1 expression is significantly higher in PDACs compared to normal pancreas. We show that pharmacological RUNX1 inhibition significantly blocks tumor growth in vivo and in primary patient-derived PDAC organoids. Through genome-wide analysis, we detected that RUNX1-loss reshapes the epigenetic landscape, which gains H3K27ac enrichment at the NOXA promoter. Our study demonstrates a previously unknown mechanism of NOXA-dependent cell death, which can be triggered pharmaceutically. Therefore, our data show a way to target a therapy-resistant PDAC, an unmet clinical need.
Collapse
|
11
|
Gugliuzza MV, Crist C. Muscle stem cell adaptations to cellular and environmental stress. Skelet Muscle 2022; 12:5. [PMID: 35151369 PMCID: PMC8840228 DOI: 10.1186/s13395-022-00289-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/30/2022] [Indexed: 12/21/2022] Open
Abstract
Background Lifelong regeneration of the skeletal muscle is dependent on a rare population of resident skeletal muscle stem cells, also named ‘satellite cells’ for their anatomical position on the outside of the myofibre and underneath the basal lamina. Muscle stem cells maintain prolonged quiescence, but activate the myogenic programme and the cell cycle in response to injury to expand a population of myogenic progenitors required to regenerate muscle. The skeletal muscle does not regenerate in the absence of muscle stem cells. Main body The notion that lifelong regeneration of the muscle is dependent on a rare, non-redundant population of stem cells seems contradictory to accumulating evidence that muscle stem cells have activated multiple stress response pathways. For example, muscle stem cell quiescence is mediated in part by the eIF2α arm of the integrated stress response and by negative regulators of mTORC1, two translational control pathways that downregulate protein synthesis in response to stress. Muscle stem cells also activate pathways to protect against DNA damage, heat shock, and environmental stress. Here, we review accumulating evidence that muscle stem cells encounter stress during their prolonged quiescence and their activation. While stress response pathways are classically described to be bimodal whereby a threshold dictates cell survival versus cell death responses to stress, we review evidence that muscle stem cells additionally respond to stress by spontaneous activation and fusion to myofibres. Conclusion We propose a cellular stress test model whereby the prolonged state of quiescence and the microenvironment serve as selective pressures to maintain muscle stem cell fitness, to safeguard the lifelong regeneration of the muscle. Fit muscle stem cells that maintain robust stress responses are permitted to maintain the muscle stem cell pool. Unfit muscle stem cells are depleted from the pool first by spontaneous activation, or in the case of severe stress, by activating cell death or senescence pathways.
Collapse
|
12
|
Oligodendrocytes depend on MCL-1 to prevent spontaneous apoptosis and white matter degeneration. Cell Death Dis 2021; 12:1133. [PMID: 34873168 PMCID: PMC8648801 DOI: 10.1038/s41419-021-04422-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/08/2021] [Accepted: 11/22/2021] [Indexed: 11/26/2022]
Abstract
Neurologic disorders often disproportionately affect specific brain regions, and different apoptotic mechanisms may contribute to white matter pathology in leukodystrophies or gray matter pathology in poliodystrophies. We previously showed that neural progenitors that generate cerebellar gray matter depend on the anti-apoptotic protein BCL-xL. Conditional deletion of Bcl-xL in these progenitors produces spontaneous apoptosis and cerebellar hypoplasia, while similar conditional deletion of Mcl-1 produces no phenotype. Here we show that, in contrast, postnatal oligodendrocytes depend on MCL-1. We found that brain-wide Mcl-1 deletion caused apoptosis specifically in mature oligodendrocytes while sparing astrocytes and oligodendrocyte precursors, resulting in impaired myelination and progressive white matter degeneration. Disabling apoptosis through co-deletion of Bax or Bak rescued white matter degeneration, implicating the intrinsic apoptotic pathway in Mcl-1-dependence. Bax and Bak co-deletions rescued different aspects of the Mcl-1-deleted phenotype, demonstrating their discrete roles in white matter stability. MCL-1 protein abundance was reduced in eif2b5-mutant mouse model of the leukodystrophy vanishing white matter disease (VWMD), suggesting the potential for MCL-1 deficiency to contribute to clinical neurologic disease. Our data show that oligodendrocytes require MCL-1 to suppress apoptosis, implicate MCL-1 deficiency in white matter pathology, and suggest apoptosis inhibition as a leukodystrophy therapy.
Collapse
|
13
|
Xu Y, Huangyang P, Wang Y, Xue L, Devericks E, Nguyen HG, Yu X, Oses-Prieto JA, Burlingame AL, Miglani S, Goodarzi H, Ruggero D. ERα is an RNA-binding protein sustaining tumor cell survival and drug resistance. Cell 2021; 184:5215-5229.e17. [PMID: 34559986 PMCID: PMC8547373 DOI: 10.1016/j.cell.2021.08.036] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 06/28/2021] [Accepted: 08/29/2021] [Indexed: 12/15/2022]
Abstract
Estrogen receptor α (ERα) is a hormone receptor and key driver for over 70% of breast cancers that has been studied for decades as a transcription factor. Unexpectedly, we discover that ERα is a potent non-canonical RNA-binding protein. We show that ERα RNA binding function is uncoupled from its activity to bind DNA and critical for breast cancer progression. Employing genome-wide cross-linking immunoprecipitation (CLIP) sequencing and a functional CRISPRi screen, we find that ERα-associated mRNAs sustain cancer cell fitness and elicit cellular responses to stress. Mechanistically, ERα controls different steps of RNA metabolism. In particular, we demonstrate that ERα RNA binding mediates alternative splicing of XBP1 and translation of the eIF4G2 and MCL1 mRNAs, which facilitates survival upon stress conditions and sustains tamoxifen resistance of cancer cells. ERα is therefore a multifaceted RNA-binding protein, and this activity transforms our knowledge of post-transcriptional regulation underlying cancer development and drug response.
Collapse
Affiliation(s)
- Yichen Xu
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peiwei Huangyang
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ying Wang
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lingru Xue
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Emily Devericks
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hao G Nguyen
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Xiuyan Yu
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Juan A Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sohit Miglani
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hani Goodarzi
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry & Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Davide Ruggero
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
14
|
Sharon D, Cathelin S, Mirali S, Di Trani JM, Yanofsky DJ, Keon KA, Rubinstein JL, Schimmer AD, Ketela T, Chan SM. Inhibition of mitochondrial translation overcomes venetoclax resistance in AML through activation of the integrated stress response. Sci Transl Med 2020; 11:11/516/eaax2863. [PMID: 31666400 DOI: 10.1126/scitranslmed.aax2863] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/07/2019] [Accepted: 09/27/2019] [Indexed: 12/12/2022]
Abstract
Venetoclax is a specific B cell lymphoma 2 (BCL-2) inhibitor with promising activity against acute myeloid leukemia (AML), but its clinical efficacy as a single agent or in combination with hypomethylating agents (HMAs), such as azacitidine, is hampered by intrinsic and acquired resistance. Here, we performed a genome-wide CRISPR knockout screen and found that inactivation of genes involved in mitochondrial translation restored sensitivity to venetoclax in resistant AML cells. Pharmacologic inhibition of mitochondrial protein synthesis with antibiotics that target the ribosome, including tedizolid and doxycycline, effectively overcame venetoclax resistance. Mechanistic studies showed that both tedizolid and venetoclax suppressed mitochondrial respiration, with the latter demonstrating inhibitory activity against complex I [nicotinamide adenine dinucleotide plus hydrogen (NADH) dehydrogenase] of the electron transport chain (ETC). The drugs cooperated to activate a heightened integrated stress response (ISR), which, in turn, suppressed glycolytic capacity, resulting in adenosine triphosphate (ATP) depletion and subsequent cell death. Combination treatment with tedizolid and venetoclax was superior to either agent alone in reducing leukemic burden in mice engrafted with treatment-resistant human AML. The addition of tedizolid to azacitidine and venetoclax further enhanced the killing of resistant AML cells in vitro and in vivo. Our findings demonstrate that inhibition of mitochondrial translation is an effective approach to overcoming venetoclax resistance and provide a rationale for combining tedizolid, azacitidine, and venetoclax as a triplet therapy for AML.
Collapse
Affiliation(s)
- David Sharon
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | | | - Sara Mirali
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Justin M Di Trani
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - David J Yanofsky
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Kristine A Keon
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - John L Rubinstein
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Troy Ketela
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada
| | - Steven M Chan
- Princess Margaret Cancer Centre, Toronto, Ontario M5G 1L7, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
15
|
Heimer S, Knoll G, Neubert P, Hammer KP, Wagner S, Bauer RJ, Jantsch J, Ehrenschwender M. Hypertonicity counteracts MCL-1 and renders BCL-XL a synthetic lethal target in head and neck cancer. FEBS J 2020; 288:1822-1838. [PMID: 32710568 DOI: 10.1111/febs.15492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/09/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an aggressive and difficult-to-treat cancer entity. Current therapies ultimately aim to activate the mitochondria-controlled (intrinsic) apoptosis pathway, but complex alterations in intracellular signaling cascades and the extracellular microenvironment hamper treatment response. On the one hand, proteins of the BCL-2 family set the threshold for cell death induction and prevent accidental cellular suicide. On the other hand, controlling a cell's readiness to die also determines whether malignant cells are sensitive or resistant to anticancer treatments. Here, we show that HNSCC cells upregulate the proapoptotic BH3-only protein NOXA in response to hyperosmotic stress. Induction of NOXA is sufficient to counteract the antiapoptotic properties of MCL-1 and switches HNSCC cells from dual BCL-XL/MCL-1 protection to exclusive BCL-XL addiction. Hypertonicity-induced functional loss of MCL-1 renders BCL-XL a synthetically lethal target in HNSCC, and inhibition of BCL-XL efficiently kills HNSCC cells that poorly respond to conventional therapies. We identify hypertonicity-induced upregulation of NOXA as link between osmotic pressure in the tumor environment and mitochondrial priming, which could perspectively be exploited to boost efficacy of anticancer drugs.
Collapse
Affiliation(s)
- Sina Heimer
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Gertrud Knoll
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Patrick Neubert
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Karin P Hammer
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Richard J Bauer
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany.,Department of Oral and Maxillofacial Surgery, Center for Medical Biotechnology, University Hospital Regensburg, Regensburg, Germany
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Martin Ehrenschwender
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
16
|
Knoll G, Riffelsberger P, Raats D, Kranenburg O, Ehrenschwender M. NOXA-dependent contextual synthetic lethality of BCL-XL inhibition and "osmotic reprogramming" in colorectal cancer. Cell Death Dis 2020; 11:257. [PMID: 32312973 PMCID: PMC7171071 DOI: 10.1038/s41419-020-2446-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
Abstract
A sophisticated network of BCL-2 family proteins regulates the mitochondria-associated (intrinsic) apoptosis pathway. Antiapoptotic members such as BCL-XL or MCL-1 safeguard the outer mitochondrial membrane and prevent accidental cell death in a functionally redundant and/or compensatory manner. However, BCL-XL/MCL-1-mediated “dual apoptosis protection” also impairs response of cancer cells to chemotherapy. Here, we show that hyperosmotic stress in the tumor environment abrogates dual BCL-XL/MCL-1 protection. Hypertonicity triggers upregulation of NOXA and loss of MCL-1 and thereby enforces exclusive BCL-XL addiction. Concomitant targeting of BCL-XL is sufficient to unlock the intrinsic apoptosis pathway in colorectal cancer cells. Functionally, “osmotic reprogramming” of the tumor environment grants contextual synthetic lethality to BCL-XL inhibitors in dually BCL-XL/MCL-1-protected cells. Generation of contextual synthetic lethality through modulation of the tumor environment could perspectively boost efficacy of anticancer drugs.
Collapse
Affiliation(s)
- Gertrud Knoll
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Petra Riffelsberger
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Danielle Raats
- Department of Surgical Oncology, UMC Utrecht Cancer Centre, PO Box 85500, 3506 GA, Utrecht, The Netherlands
| | - Onno Kranenburg
- Department of Surgical Oncology, UMC Utrecht Cancer Centre, PO Box 85500, 3506 GA, Utrecht, The Netherlands
| | - Martin Ehrenschwender
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
17
|
Abhari BA, McCarthy N, Agostinis P, Fulda S. NF-κB contributes to Smac mimetic-conferred protection from tunicamycin-induced apoptosis. Apoptosis 2020; 24:269-277. [PMID: 30680482 DOI: 10.1007/s10495-018-1507-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Smac mimetics that deplete cellular inhibitor of apoptosis (cIAP) proteins have been shown to activate Nuclear Factor-kappa B (NF-κB). Here, we report that Smac mimetic-mediated activation of NF-κB contributes to the rescue of cancer cells from tunicamycin (TM)-triggered apoptosis. The prototypic Smac mimetic BV6 activates non-canonical and canonical NF-κB pathways, while TM has little effect on NF-κB signaling. Importantly, ectopic expression of dominant-negative IκBα superrepressor (IκBα-SR), which inhibits canonical and non-canonical NF-κB activation, significantly reversed this BV6-imposed protection against TM. Similarly, transient or stable knockdown of NF-κB-inducing kinase, which accumulated upon exposure to BV6 alone and in combination with TM, significantly counteracted BV6-mediated inhibition of TM-induced apoptosis. Interestingly, while cIAP2 was initially degraded upon BV6 treatment, it was subsequently upregulated in an NF-κB-dependent manner, as this restoration of cIAP2 expression was abolished in IκBα-SR-overexpressing cells. Interestingly, upon exposure to TM/BV6 apoptosis was significantly increased in cIAP2 knockdown cells. Furthermore, NF-κB inhibition partially prevented BV6-stimulated expression of Mcl-1 upon TM treatment. Consistently, Mcl-1 silencing significantly inhibited BV6-mediated protection from TM-induced apoptosis. Thus, NF-κB activation by Smac mimetic contributes to Smac mimetic-mediated protection against TM-induced apoptosis.
Collapse
Affiliation(s)
- Behnaz Ahangarian Abhari
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528, Frankfurt, Germany
| | - Nicole McCarthy
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528, Frankfurt, Germany
| | - Patrizia Agostinis
- Cell Death Research and Therapy Unit, Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528, Frankfurt, Germany. .,German Cancer Consortium (DKTK), Partner Site Frankfurt, Heidelberg, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
18
|
Saga of Mcl-1: regulation from transcription to degradation. Cell Death Differ 2020; 27:405-419. [PMID: 31907390 DOI: 10.1038/s41418-019-0486-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/05/2019] [Accepted: 12/13/2019] [Indexed: 01/01/2023] Open
Abstract
The members of the Bcl-2 family are the central regulators of various cell death modalities. Some of these proteins contribute to apoptosis, while others counteract this type of programmed cell death, thus balancing cell demise and survival. A disruption of this balance leads to the development of various diseases, including cancer. Therefore, understanding the mechanisms that underlie the regulation of proteins of the Bcl-2 family is of great importance for biomedical research. Among the members of the Bcl-2 family, antiapoptotic protein Mcl-1 is characterized by a short half-life, which renders this protein highly sensitive to changes in its synthesis or degradation. Hence, the regulation of Mcl-1 is of particular scientific interest, and the study of Mcl-1 modulators could aid in the understanding of the mechanisms of disease development and the ways of their treatment. Here, we summarize the present knowledge regarding the regulation of Mcl-1, from transcription to degradation, focusing on aspects that have not yet been described in detail.
Collapse
|
19
|
Targeting KPNB1 overcomes TRAIL resistance by regulating DR5, Mcl-1 and FLIP in glioblastoma cells. Cell Death Dis 2019; 10:118. [PMID: 30742128 PMCID: PMC6370806 DOI: 10.1038/s41419-019-1383-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/16/2018] [Accepted: 01/09/2019] [Indexed: 01/08/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine with potential anticancer effect, but innate and adaptive TRAIL resistance in majority of cancers limit its clinical application. Karyopherin β1 (KPNB1) inhibition in cancer cells has been reported to abrogate the nuclear import of TRAIL receptor DR5 and facilitate its localization on the cell surface ready for TRAIL stimulation. However, our study reveals a more complicated mechanism. Genetic or pharmacological inhibition of KPNB1 potentiated TRAIL-induced apoptosis selectively in glioblastoma cells mainly by unfolded protein response (UPR). First, it augmented ATF4-mediated DR5 expression and promoted the assembly of death-inducing signaling complex (DISC). Second, it freed Bax and Bak from Mcl-1. Third, it downregulated FLIPL and FLIPS, inhibitors of caspase-8 cleavage, partly through upregulating ATF4–induced 4E-BP1 expression and disrupting the cap-dependent translation initiation. Meanwhile, KPNB1 inhibition-induced undesirable autophagy and accelerated cleaved caspase-8 clearance. Inhibition of autophagic flux maintained cleaved caspase-8 and aggravated apoptosis induced by KPNB1 inhibitor plus TRAIL, which were abolished by caspase-8 inhibitor. These results unveil new molecular mechanism for optimizing TRAIL-directed therapeutic efficacy against cancer.
Collapse
|
20
|
Florean C, Kim KR, Schnekenburger M, Kim HJ, Moriou C, Debitus C, Dicato M, Al-Mourabit A, Han BW, Diederich M. Synergistic AML Cell Death Induction by Marine Cytotoxin (+)-1( R), 6( S), 1'( R), 6'( S), 11( R), 17( S)-Fistularin-3 and Bcl-2 Inhibitor Venetoclax. Mar Drugs 2018; 16:md16120518. [PMID: 30572618 PMCID: PMC6316187 DOI: 10.3390/md16120518] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 11/23/2022] Open
Abstract
Treatment of acute myeloid leukemia (AML) patients is still hindered by resistance and relapse, resulting in an overall poor survival rate. Recently, combining specific B-cell lymphoma (Bcl)-2 inhibitors with compounds downregulating myeloid cell leukemia (Mcl)-1 has been proposed as a new effective strategy to eradicate resistant AML cells. We show here that 1(R), 6(S), 1’(R), 6’(S), 11(R), 17(S)-fistularin-3, a bromotyrosine compound of the fistularin family, isolated from the marine sponge Suberea clavata, synergizes with Bcl-2 inhibitor ABT-199 to efficiently kill Mcl-1/Bcl-2-positive AML cell lines, associated with Mcl-1 downregulation and endoplasmic reticulum stress induction. The absolute configuration of carbons 11 and 17 of the fistularin-3 stereoisomer was fully resolved in this study for the first time, showing that the fistularin we isolated from the marine sponge Subarea clavata is in fact the (+)-11(R), 17(S)-fistularin-3 stereoisomer keeping the known configuration 1(R), 6(S), 1’(R), and 6’(S) for the verongidoic acid part. Docking studies and in vitro assays confirm the potential of this family of molecules to inhibit DNA methyltransferase 1 activity.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/administration & dosage
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Line, Tumor
- Cell Survival/drug effects
- Drug Screening Assays, Antitumor
- Drug Synergism
- Endoplasmic Reticulum Stress/drug effects
- HL-60 Cells
- Humans
- Isoxazoles/administration & dosage
- Isoxazoles/chemistry
- Isoxazoles/isolation & purification
- Isoxazoles/pharmacology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Molecular Docking Simulation
- Porifera/chemistry
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Sulfonamides/administration & dosage
- Sulfonamides/pharmacology
- Tyrosine/administration & dosage
- Tyrosine/analogs & derivatives
- Tyrosine/chemistry
- Tyrosine/isolation & purification
- Tyrosine/pharmacology
- U937 Cells
Collapse
Affiliation(s)
- Cristina Florean
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg.
| | - Kyung Rok Kim
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg.
| | - Hyun-Jung Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea.
| | - Céline Moriou
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Univ. Paris-Sud, University of Paris-Saclay, 1, Avenue de la Terrasse, 91198 Gif-Sur-Yvette, France.
| | - Cécile Debitus
- LEMAR, IRD, UBO, CNRS, IFREMER, IUEM, 29280 Plouzané, France.
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg.
| | - Ali Al-Mourabit
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Univ. Paris-Sud, University of Paris-Saclay, 1, Avenue de la Terrasse, 91198 Gif-Sur-Yvette, France.
| | - Byung Woo Han
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| | - Marc Diederich
- Department of Pharmacy, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.
| |
Collapse
|
21
|
By reducing global mRNA translation in several ways, 2-deoxyglucose lowers MCL-1 protein and sensitizes hemopoietic tumor cells to BH3 mimetic ABT737. Cell Death Differ 2018; 26:1766-1781. [PMID: 30538285 PMCID: PMC6748140 DOI: 10.1038/s41418-018-0244-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/30/2018] [Accepted: 11/15/2018] [Indexed: 01/15/2023] Open
Abstract
Drugs targeting various pro-survival BCL-2 family members (‘‘BH3 mimetics’’) have efficacy in hemopoietic malignancies, but the non-targeted pro-survival family members can promote resistance. Pertinently, the sensitivity of some tumor cell lines to BH3 mimetic ABT737, which targets BCL-2, BCL-XL, and BCL-W but not MCL-1, is enhanced by 2-deoxyglucose (2DG). We found that 2DG augmented apoptosis induced by ABT737 in 3 of 8 human hemopoietic tumor cell lines, most strongly in pre-B acute lymphocytic leukemia cell line NALM-6, the focus of our mechanistic studies. Although 2DG can lower MCL-1 translation, how it does so is incompletely understood, in part because 2DG inhibits both glycolysis and protein glycosylation in the endoplasmic reticulum (ER). Its glycolysis inhibition lowered ATP and, through the AMPK/mTORC1 pathway, markedly reduced global protein synthesis, as did an ER integrated stress response. A dual reporter assay revealed that 2DG impeded not only cap-dependent translation but also elongation or cap-independent translation. MCL-1 protein fell markedly, whereas 12 other BCL-2 family members were unaffected. We ascribe the MCL-1 drop to the global fall in translation, exacerbated for mRNAs with a structured 5′ untranslated region (5′UTR) containing potential regulatory motifs like those in MCL-1 mRNA and the short half-life of MCL-1 protein. Pertinently, 2DG downregulated two other short-lived oncoproteins, MYC and MDM2. Thus, our results support MCL-1 as a critical 2DG target, but also reveal multiple effects on global translation that may well also affect its promotion of apoptosis.
Collapse
|
22
|
Arai S, Jonas O, Whitman MA, Corey E, Balk SP, Chen S. Tyrosine Kinase Inhibitors Increase MCL1 Degradation and in Combination with BCLXL/BCL2 Inhibitors Drive Prostate Cancer Apoptosis. Clin Cancer Res 2018; 24:5458-5470. [PMID: 30021909 PMCID: PMC6214713 DOI: 10.1158/1078-0432.ccr-18-0549] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/18/2018] [Accepted: 07/09/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Clinically available BH3 mimetic drugs targeting BCLXL and/or BCL2 (navitoclax and venetoclax, respectively) are effective in some hematologic malignancies, but have limited efficacy in solid tumors. This study aimed to identify combination therapies that exploit clinical BH3 mimetics for prostate cancer.Experimental Design: Prostate cancer cells or xenografts were treated with BH3 mimetics as single agents or in combination with other agents, and effects on MCL1 and apoptosis were assessed. MCL1 was also targeted directly using RNAi, CRISPR, or an MCL1-specific BH3 mimetic, S63845.Results: We initially found that MCL1 depletion or inhibition markedly sensitized prostate cancer cells to apoptosis mediated by navitoclax, but not venetoclax, in vitro and in vivo, indicating that they are primed to undergo apoptosis and protected by MCL1 and BCLXL. Small-molecule EGFR kinase inhibitors (erlotinib, lapatinib) also dramatically sensitized to navitoclax-mediated apoptosis, and this was associated with markedly increased proteasome-dependent degradation of MCL1. This increased MCL1 degradation appeared to be through a novel mechanism, as it was not dependent upon GSK3β-mediated phosphorylation and subsequent ubiquitylation by the ubiquitin ligases βTRCP and FBW7, or through other previously identified MCL1 ubiquitin ligases or deubiquitinases. Inhibitors targeting additional kinases (cabozantinib and sorafenib) similarly caused GSK3β-independent MCL1 degradation, and in combination with navitoclax drove apoptosis in vitro and in vivo Conclusions: These results show that prostate cancer cells are primed to undergo apoptosis and that cotargeting BCLXL and MCL1, directly or indirectly through agents that increase MCL1 degradation, can induce dramatic apoptotic responses. Clin Cancer Res; 24(21); 5458-70. ©2018 AACR.
Collapse
Affiliation(s)
- Seiji Arai
- Hematology-Oncology Division, Department of Medicine, and Cancer Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
- Department of Urology, Gunma University Hospital, Maebashi, Gunma, Japan
| | - Oliver Jonas
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Matthew A Whitman
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Steven P Balk
- Hematology-Oncology Division, Department of Medicine, and Cancer Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts.
| | - Sen Chen
- Hematology-Oncology Division, Department of Medicine, and Cancer Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
23
|
Tosello V, Saccomani V, Yu J, Bordin F, Amadori A, Piovan E. Calcineurin complex isolated from T-cell acute lymphoblastic leukemia (T-ALL) cells identifies new signaling pathways including mTOR/AKT/S6K whose inhibition synergize with calcineurin inhibition to promote T-ALL cell death. Oncotarget 2018; 7:45715-45729. [PMID: 27304189 PMCID: PMC5216755 DOI: 10.18632/oncotarget.9933] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 05/28/2016] [Indexed: 02/06/2023] Open
Abstract
Calcineurin (Cn) is a calcium activated protein phosphatase involved in many aspects of normal T cell physiology, however the role of Cn and/or its downstream targets in leukemogenesis are still ill-defined. In order to identify putative downstream targets/effectors involved in the pro-oncogenic activity of Cn in T-cell acute lymphoblastic leukemia (T-ALL) we used tandem affinity chromatography, followed by mass spectrometry to purify novel Cn-interacting partners. We found the Cn-interacting proteins to be part of numerous cellular signaling pathways including eIF2 signaling and mTOR signaling. Coherently, modulation of Cn activity in T-ALL cells determined alterations in the phosphorylation status of key molecules implicated in protein translation such as eIF-2α and ribosomal protein S6. Joint targeting of PI3K-mTOR, eIF-2α and 14-3-3 signaling pathways with Cn unveiled novel synergistic pro-apoptotic drug combinations. Further analysis disclosed that the synergistic interaction between PI3K-mTOR and Cn inhibitors was prevalently due to AKT inhibition. Finally, we showed that the synergistic pro-apoptotic response determined by jointly targeting AKT and Cn pathways was linked to down-modulation of key anti-apoptotic proteins including Mcl-1, Claspin and XIAP. In conclusion, we identify AKT inhibition as a novel promising drug combination to potentiate the pro-apoptotic effects of Cn inhibitors.
Collapse
Affiliation(s)
- Valeria Tosello
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto-IRCCS, Padova, 35128, Italy
| | - Valentina Saccomani
- Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Sezione di Oncologia, Universita' di Padova, Padova, 35128, Italy
| | - Jiyang Yu
- Department of Biomedical Informatics, Columbia University, New York, NY, 10032, USA.,Department of Systems Biology, Columbia University, New York, NY, 10032, USA.,Present address: Department of Precision Medicine, Oncology Research Unit, Pfizer Inc., Pearl River, NY, 10965, USA
| | - Fulvio Bordin
- Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Sezione di Oncologia, Universita' di Padova, Padova, 35128, Italy
| | - Alberto Amadori
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto-IRCCS, Padova, 35128, Italy.,Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Sezione di Oncologia, Universita' di Padova, Padova, 35128, Italy
| | - Erich Piovan
- UOC Immunologia e Diagnostica Molecolare Oncologica, Istituto Oncologico Veneto-IRCCS, Padova, 35128, Italy.,Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche, Sezione di Oncologia, Universita' di Padova, Padova, 35128, Italy
| |
Collapse
|
24
|
Young AI, Timpson P, Gallego-Ortega D, Ormandy CJ, Oakes SR. Myeloid cell leukemia 1 (MCL-1), an unexpected modulator of protein kinase signaling during invasion. Cell Adh Migr 2017; 12:513-523. [PMID: 29166822 PMCID: PMC6363037 DOI: 10.1080/19336918.2017.1393591] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Myeloid cell leukemia-1 (MCL-1), closely related to B-cell lymphoma 2 (BCL-2), has a well-established role in cell survival and has emerged as an important target for cancer therapeutics. We have demonstrated that inhibiting MCL-1 is efficacious in suppressing tumour progression in pre-clinical models of breast cancer and revealed that in addition to its role in cell survival, MCL-1 modulated cellular invasion. Utilizing a MCL-1-specific genetic antagonist, we found two possible mechanisms; firstly MCL-1 directly binds to and alters the phosphorylation of the cytoskeletal remodeling protein, Cofilin, a protein important for cytoskeletal remodeling during invasion, and secondly MCL-1 modulates the levels SRC family kinases (SFKs) and their targets. These data provide evidence that MCL-1 activities are not limited to endpoints of extracellular and intracellular signaling culminating in cell survival as previously thought, but can directly modulate the output of SRC family kinases signaling during cellular invasion. Here we review the pleotropic roles of MCL-1 and discuss the implications of this newly discovered effect on protein kinase signaling for the development of cancer therapeutics.
Collapse
Affiliation(s)
- Adelaide Ij Young
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia
| | - Paul Timpson
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia.,b St. Vincent's Clinical School, UNSW Medicine , Victoria Street, Darlinghurst , NSW , Australia
| | - David Gallego-Ortega
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia.,b St. Vincent's Clinical School, UNSW Medicine , Victoria Street, Darlinghurst , NSW , Australia
| | - Christopher J Ormandy
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia.,b St. Vincent's Clinical School, UNSW Medicine , Victoria Street, Darlinghurst , NSW , Australia
| | - Samantha R Oakes
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia.,b St. Vincent's Clinical School, UNSW Medicine , Victoria Street, Darlinghurst , NSW , Australia
| |
Collapse
|
25
|
Protein Kinase R Mediates the Inflammatory Response Induced by Hyperosmotic Stress. Mol Cell Biol 2017; 37:MCB.00521-16. [PMID: 27920257 DOI: 10.1128/mcb.00521-16] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/01/2016] [Indexed: 12/25/2022] Open
Abstract
High extracellular osmolarity results in a switch from an adaptive to an inflammatory gene expression program. We show that hyperosmotic stress activates the protein kinase R (PKR) independently of its RNA-binding domain. In turn, PKR stimulates nuclear accumulation of nuclear factor κB (NF-κB) p65 species phosphorylated at serine-536, which is paralleled by the induction of a subset of inflammatory NF-κB p65-responsive genes, including inducible nitric oxide synthase (iNOS), interleukin-6 (IL-6), and IL-1β. The PKR-mediated hyperinduction of iNOS decreases cell survival in mouse embryonic fibroblasts via mechanisms involving nitric oxide (NO) synthesis and posttranslational modification of proteins. Moreover, we demonstrate that the PKR inhibitor C16 ameliorates both iNOS amplification and disease-induced phenotypic breakdown of the intestinal epithelial barrier caused by an increase in extracellular osmolarity induced by dextran sodium sulfate (DSS) in vivo Collectively, these findings indicate that PKR activation is an essential part of the molecular switch from adaptation to inflammation in response to hyperosmotic stress.
Collapse
|
26
|
Bommer UA. The Translational Controlled Tumour Protein TCTP: Biological Functions and Regulation. Results Probl Cell Differ 2017; 64:69-126. [PMID: 29149404 DOI: 10.1007/978-3-319-67591-6_4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The Translational Controlled Tumour Protein TCTP (gene symbol TPT1, also called P21, P23, Q23, fortilin or histamine-releasing factor, HRF) is a highly conserved protein present in essentially all eukaryotic organisms and involved in many fundamental cell biological and disease processes. It was first discovered about 35 years ago, and it took an extended period of time for its multiple functions to be revealed, and even today we do not yet fully understand all the details. Having witnessed most of this history, in this chapter, I give a brief overview and review the current knowledge on the structure, biological functions, disease involvements and cellular regulation of this protein.TCTP is able to interact with a large number of other proteins and is therefore involved in many core cell biological processes, predominantly in the response to cellular stresses, such as oxidative stress, heat shock, genotoxic stress, imbalance of ion metabolism as well as other conditions. Mechanistically, TCTP acts as an anti-apoptotic protein, and it is involved in DNA-damage repair and in cellular autophagy. Thus, broadly speaking, TCTP can be considered a cytoprotective protein. In addition, TCTP facilitates cell division through stabilising the mitotic spindle and cell growth through modulating growth signalling pathways and through its interaction with the proteosynthetic machinery of the cell. Due to its activities, both as an anti-apoptotic protein and in promoting cell growth and division, TCTP is also essential in the early development of both animals and plants.Apart from its involvement in various biological processes at the cellular level, TCTP can also act as an extracellular protein and as such has been involved in modulating whole-body defence processes, namely in the mammalian immune system. Extracellular TCTP, typically in its dimerised form, is able to induce the release of cytokines and other signalling molecules from various types of immune cells. There are also several examples, where TCTP was shown to be involved in antiviral/antibacterial defence in lower animals. In plants, the protein appears to have a protective effect against phytotoxic stresses, such as flooding, draught, too high or low temperature, salt stress or exposure to heavy metals. The finding for the latter stress condition is corroborated by earlier reports that TCTP levels are considerably up-regulated upon exposure of earthworms to high levels of heavy metals.Given the involvement of TCTP in many biological processes aimed at maintaining cellular or whole-body homeostasis, it is not surprising that dysregulation of TCTP levels may promote a range of disease processes, foremost cancer. Indeed a large body of evidence now supports a role of TCTP in at least the most predominant types of human cancers. Typically, this can be ascribed to both the anti-apoptotic activity of the protein and to its function in promoting cell growth and division. However, TCTP also appears to be involved in the later stages of cancer progression, such as invasion and metastasis. Hence, high TCTP levels in tumour tissues are often associated with a poor patient outcome. Due to its multiple roles in cancer progression, TCTP has been proposed as a potential target for the development of new anti-cancer strategies in recent pilot studies. Apart from its role in cancer, TCTP dysregulation has been reported to contribute to certain processes in the development of diabetes, as well as in diseases associated with the cardiovascular system.Since cellular TCTP levels are highly regulated, e.g. in response to cell stress or to growth signalling, and because deregulation of this protein contributes to many disease processes, a detailed understanding of regulatory processes that impinge on TCTP levels is required. The last section of this chapter summarises our current knowledge on the mechanisms that may be involved in the regulation of TCTP levels. Essentially, expression of the TPT1 gene is regulated at both the transcriptional and the translational level, the latter being particularly advantageous when a rapid adjustment of cellular TCTP levels is required, for example in cell stress responses. Other regulatory mechanisms, such as protein stability regulation, may also contribute to the regulation of overall TCTP levels.
Collapse
Affiliation(s)
- Ulrich-Axel Bommer
- School of Medicine, Graduate Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
27
|
Liu H, Chen J, Jiang X, Wang T, Xie X, Hu H, Yu F, Wang X, Fan H. Apoptotic signal pathways and regulatory mechanisms of cancer cells induced by IL-24. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s11859-016-1205-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
28
|
Gomez-Bougie P, Halliez M, Moreau P, Pellat-Deceunynck C, Amiot M. Repression of Mcl-1 and disruption of the Mcl-1/Bak interaction in myeloma cells couple ER stress to mitochondrial apoptosis. Cancer Lett 2016; 383:204-211. [PMID: 27697610 DOI: 10.1016/j.canlet.2016.09.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 09/22/2016] [Accepted: 09/22/2016] [Indexed: 12/22/2022]
Abstract
As myeloma cells actively produce and secrete immunoglobulins, they are prone to ER stress, which if unresolved leads to apoptosis. We found that myeloma cell death induced by the ER stressor Thapsigargin was highly variable, ranging from 2 to 89%. Induction of ATF4 and CHOP was observed in myeloma cells under Thapsigargin independently of cell death. The decrease in Mcl-1 was associated with protein translation inhibition and identified as a crucial factor in Thapsigargin sensitivity, since it was the only Bcl-2 family protein differentially modified between sensitive and resistant myeloma cells. Bak but not Bax was found to contribute to Thapsigargin-induced apoptosis. Appropriately, a basal Mcl-1/Bak interaction was demonstrated in Thapsigargin-sensitive cells. Of note, the only pro-apoptotic protein freed from Mcl-1 under Thapsigargin was Bak, whereas Mcl-1/Noxa or Mcl-1/Bim complexes were simultaneously increased. Thus, the disruption of the basal Mcl-1/Bak complex in Thapsigargin-sensitive cells seemed to be an essential event in cell death induction, probably favored by the induced Noxa and Bim BH3-only proteins. These findings underscore the implication of the Mcl-1/Bak axis in myeloma cell death triggered by Thapsigargin.
Collapse
Affiliation(s)
- Patricia Gomez-Bougie
- CRCNA, INSERM, CNRS, CHU, Université d'Angers, Université de Nantes, F-44000, Nantes, France.
| | - Maxime Halliez
- CRCNA, INSERM, CNRS, CHU, Université d'Angers, Université de Nantes, F-44000, Nantes, France
| | - Philippe Moreau
- CRCNA, INSERM, CNRS, CHU, Université d'Angers, Université de Nantes, F-44000, Nantes, France
| | | | - Martine Amiot
- CRCNA, INSERM, CNRS, CHU, Université d'Angers, Université de Nantes, F-44000, Nantes, France
| |
Collapse
|
29
|
Yu Q, Liu ZY, Chen Q, Lin JS. Mcl-1 as a potential therapeutic target for human hepatocelluar carcinoma. ACTA ACUST UNITED AC 2016; 36:494-500. [DOI: 10.1007/s11596-016-1614-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/06/2016] [Indexed: 02/08/2023]
|
30
|
Cui J, Placzek WJ. PTBP1 modulation of MCL1 expression regulates cellular apoptosis induced by antitubulin chemotherapeutics. Cell Death Differ 2016; 23:1681-90. [PMID: 27367564 DOI: 10.1038/cdd.2016.60] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 04/30/2016] [Accepted: 05/30/2016] [Indexed: 01/01/2023] Open
Abstract
Myeloid cell leukemia sequence 1 (MCL1), an anti-apoptotic BCL2 family protein, is a key regulator of intrinsic apoptosis. Normal cells require strict control over MCL1 expression with aberrant MCL1 expression linked to the emergence of various diseases and chemoresistance. Previous studies have detailed how MCL1 expression is regulated by multiple mechanisms both transcriptionally and translationally. However, characterization of the post-transcriptional regulators of MCL1 mRNA is limited. Polypyrimidine tract binding protein 1 (PTBP1) is a known regulator of post-transcriptional gene expression that can control mRNA splicing, translation, stability and localization. Here we demonstrate that PTBP1 binds to MCL1 mRNA and that knockdown of PTBP1 upregulates MCL1 expression in cancer cells by stabilizing MCL1 mRNA and increasing MCL1 mRNA accumulation in cytoplasm. Further, we show that depletion of PTBP1 protects cancer cells from antitubulin agent-induced apoptosis in a MCL1-dependent manner. Taken together, our findings suggest that PTBP1 is a novel regulator of MCL1 mRNA by which it controls apoptotic response to antitubulin chemotherapeutics.
Collapse
Affiliation(s)
- J Cui
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - W J Placzek
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
31
|
Burger JA, Landau DA, Taylor-Weiner A, Bozic I, Zhang H, Sarosiek K, Wang L, Stewart C, Fan J, Hoellenriegel J, Sivina M, Dubuc AM, Fraser C, Han Y, Li S, Livak KJ, Zou L, Wan Y, Konoplev S, Sougnez C, Brown JR, Abruzzo LV, Carter SL, Keating MJ, Davids MS, Wierda WG, Cibulskis K, Zenz T, Werner L, Cin PD, Kharchencko P, Neuberg D, Kantarjian H, Lander E, Gabriel S, O'Brien S, Letai A, Weitz DA, Nowak MA, Getz G, Wu CJ. Clonal evolution in patients with chronic lymphocytic leukaemia developing resistance to BTK inhibition. Nat Commun 2016; 7:11589. [PMID: 27199251 PMCID: PMC4876453 DOI: 10.1038/ncomms11589] [Citation(s) in RCA: 259] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 04/12/2016] [Indexed: 02/06/2023] Open
Abstract
Resistance to the Bruton's tyrosine kinase (BTK) inhibitor ibrutinib has been attributed solely to mutations in BTK and related pathway molecules. Using whole-exome and deep-targeted sequencing, we dissect evolution of ibrutinib resistance in serial samples from five chronic lymphocytic leukaemia patients. In two patients, we detect BTK-C481S mutation or multiple PLCG2 mutations. The other three patients exhibit an expansion of clones harbouring del(8p) with additional driver mutations (EP300, MLL2 and EIF2A), with one patient developing trans-differentiation into CD19-negative histiocytic sarcoma. Using droplet-microfluidic technology and growth kinetic analyses, we demonstrate the presence of ibrutinib-resistant subclones and estimate subclone size before treatment initiation. Haploinsufficiency of TRAIL-R, a consequence of del(8p), results in TRAIL insensitivity, which may contribute to ibrutinib resistance. These findings demonstrate that the ibrutinib therapy favours selection and expansion of rare subclones already present before ibrutinib treatment, and provide insight into the heterogeneity of genetic changes associated with ibrutinib resistance.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- Adult
- Agammaglobulinaemia Tyrosine Kinase
- Aged, 80 and over
- Apoptosis
- Cell Transdifferentiation
- Clonal Evolution
- Drug Resistance, Neoplasm/genetics
- Female
- Histiocytic Sarcoma/etiology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Male
- Middle Aged
- Mutation
- Neoplasm Recurrence, Local/genetics
- Piperidines
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/genetics
- Pyrazoles/pharmacology
- Pyrazoles/therapeutic use
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Selection, Genetic
Collapse
Affiliation(s)
- Jan A. Burger
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Dan A. Landau
- Broad Institute, Cambridge, Massachusetts 02142, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York 10065, USA
- New York Genome Center, New York, New York 10013, USA
| | | | - Ivana Bozic
- Department of Mathematics, Program for Evolutionary Dynamics, Harvard University, Cambridge, Massachusetts 02138, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Huidan Zhang
- Department of Physics, School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, China Medical University, Shenyang 110001, China
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110001, China
| | - Kristopher Sarosiek
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Lili Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Chip Stewart
- Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Jean Fan
- Center for Biomedical Informatics, Harvard Medical School, Boston Massachusetts 02115, USA
| | - Julia Hoellenriegel
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Mariela Sivina
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Adrian M. Dubuc
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Cameron Fraser
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Yulong Han
- Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shuqiang Li
- Fluidigm Corporation, South San Francisco, California 94080, USA
| | - Kenneth J. Livak
- Fluidigm Corporation, South San Francisco, California 94080, USA
| | - Lihua Zou
- Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Youzhong Wan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Sergej Konoplev
- Department of Hematopathology, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | - Jennifer R. Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Lynne V. Abruzzo
- Department of Hematopathology, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | - Michael J. Keating
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - William G. Wierda
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | - Thorsten Zenz
- National Center for Tumors and German Cancer Research Center (DKFZ), 69121 Heidelberg, Germany
| | - Lillian Werner
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Paola Dal Cin
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Peter Kharchencko
- Center for Biomedical Informatics, Harvard Medical School, Boston Massachusetts 02115, USA
| | - Donna Neuberg
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Hagop Kantarjian
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Eric Lander
- Broad Institute, Cambridge, Massachusetts 02142, USA
| | | | - Susan O'Brien
- Department of Leukemia, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - David A. Weitz
- Department of Physics, School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Martin A. Nowak
- Department of Mathematics, Program for Evolutionary Dynamics, Harvard University, Cambridge, Massachusetts 02138, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Gad Getz
- Broad Institute, Cambridge, Massachusetts 02142, USA
| | - Catherine J. Wu
- Broad Institute, Cambridge, Massachusetts 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
32
|
Zhong L, Wang L, Xu L, Liu Q, Jiang L, Zhi Y, Lu W, Zhou P. The cytotoxic effect of the NOS-mediated oxidative stress in MCF-7 cells after PbCl₂ exposure. ENVIRONMENTAL TOXICOLOGY 2016; 31:601-608. [PMID: 25410796 DOI: 10.1002/tox.22073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 10/28/2014] [Accepted: 10/31/2014] [Indexed: 06/04/2023]
Abstract
The potential Pb-induced cytotoxicity in various tissues and biological systems has been reported. Some evidences also indicate that the Pb-caused cytotoxicity may be associated with the nitric oxide synthase (NOS). However, there remains uncertainty about the role of the NOS signaling pathway during the Pb-induced cytotoxicity. In this report, we provide data showing that PbCl2 treatment depresses the expressions of the three distinct NOS isoforms: neuronal nitric oxide synthase (nNOS), endothelial NOS (eNOS), and inducible NOS (iNOS) on both transcriptional and translational levels in MCF-7 cells. The down-regulation of NOSs expressions by PbCl2 exposure leads to reduced NOS activity and nitric oxide (NO) production. Meanwhile, the intracellular reactive oxygen species (ROS) level is elevated after PbCl2 exposure, which leads to the alpha subunit of eukaryotic initiation factor 2 (elF2α) phosphorylation. The reduction effects of the free radical scavenger N-acetyl-L-cysteine or the NOS substrate l-arginine on the Pb-induced ROS generation suggest that the NOS signaling pathway plays a key role in the Pb-induced oxidative stress, which further results in the elF2α phosphorylation and cytotoxicity.
Collapse
Affiliation(s)
- Lingying Zhong
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- Bor S. Luh Food Safety Research Center, Shanghai, 200240, People's Republic of China
| | - Lumei Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- Bor S. Luh Food Safety Research Center, Shanghai, 200240, People's Republic of China
- Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai, 200240, People's Republic of China
| | - Lurong Xu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- Bor S. Luh Food Safety Research Center, Shanghai, 200240, People's Republic of China
- Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai, 200240, People's Republic of China
| | - Qunlu Liu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- Bor S. Luh Food Safety Research Center, Shanghai, 200240, People's Republic of China
- Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai, 200240, People's Republic of China
| | - Linlei Jiang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- Bor S. Luh Food Safety Research Center, Shanghai, 200240, People's Republic of China
- Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai, 200240, People's Republic of China
- Shanghai Food Safety Engineering Research Center, Shanghai, 200240, People's Republic of China
| | - Yuee Zhi
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- Bor S. Luh Food Safety Research Center, Shanghai, 200240, People's Republic of China
- Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai, 200240, People's Republic of China
| | - Wei Lu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- Bor S. Luh Food Safety Research Center, Shanghai, 200240, People's Republic of China
- Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai, 200240, People's Republic of China
| | - Pei Zhou
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
- Bor S. Luh Food Safety Research Center, Shanghai, 200240, People's Republic of China
- Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai, 200240, People's Republic of China
| |
Collapse
|
33
|
Hong MN, Nam KY, Kim KK, Kim SY, Kim I. The small molecule '1-(4-biphenylylcarbonyl)-4-(5-bromo-2-methoxybenzyl) piperazine oxalate' and its derivatives regulate global protein synthesis by inactivating eukaryotic translation initiation factor 2-alpha. Cell Stress Chaperones 2016; 21:485-97. [PMID: 26873011 PMCID: PMC4837177 DOI: 10.1007/s12192-016-0677-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 01/29/2016] [Accepted: 01/30/2016] [Indexed: 10/22/2022] Open
Abstract
By environmental stresses, cells can initiate a signaling pathway in which eukaryotic translation initiation factor 2-alpha (eIF2-α) is involved to regulate the response. Phosphorylation of eIF2-α results in the reduction of overall protein neogenesis, which allows cells to conserve resources and to reprogram energy usage for effective stress control. To investigate the role of eIF2-α in cell stress responses, we conducted a viability-based compound screen under endoplasmic reticulum (ER) stress condition, and identified 1-(4-biphenylylcarbonyl)-4-(5-bromo-2-methoxybenzyl) piperazine oxalate (AMC-01) and its derivatives as eIF2-α-inactivating chemical. Molecular characterization of this signaling pathway revealed that AMC-01 induced inactivation of eIF2-α by phosphorylating serine residue 51 in a dose- and time-dependent manner, while the negative control compounds did not affect eIF2-α phosphorylation. In contrast with ER stress induction by thapsigargin, phosphorylation of eIF2-α persisted for the duration of incubation with AMC-01. By pathway analysis, AMC-01 clearly induced the activation of protein kinase RNA-activated (PKR) kinase and nuclear factor-κB (NF-κB), whereas it did not modulate the activity of PERK or heme-regulated inhibitor (HRI). Finally, we could detect a lower protein translation rate in cells incubated with AMC-01, establishing AMC-01 as a potent chemical probe that can regulate eIF2-α activity. We suggest from these data that AMC-01 and its derivative compounds can be used as chemical probes in future studies of the role of eIF2-α in protein synthesis-related cell physiology.
Collapse
Affiliation(s)
- Mi-Na Hong
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, Convergence Medicine Research Building, 43 gil Olympicro, Pungnapdong, Songpagu, Seoul, 138-736, Republic of Korea
| | - Ky-Youb Nam
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, Convergence Medicine Research Building, 43 gil Olympicro, Pungnapdong, Songpagu, Seoul, 138-736, Republic of Korea
| | - Kyung Kon Kim
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, Convergence Medicine Research Building, 43 gil Olympicro, Pungnapdong, Songpagu, Seoul, 138-736, Republic of Korea
- Department of Convergence Medicine, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - So-Young Kim
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, Convergence Medicine Research Building, 43 gil Olympicro, Pungnapdong, Songpagu, Seoul, 138-736, Republic of Korea
| | - InKi Kim
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, Convergence Medicine Research Building, 43 gil Olympicro, Pungnapdong, Songpagu, Seoul, 138-736, Republic of Korea.
- Department of Convergence Medicine, College of Medicine, University of Ulsan, Seoul, Republic of Korea.
| |
Collapse
|
34
|
De Falco F, Sabatini R, Del Papa B, Falzetti F, Di Ianni M, Sportoletti P, Baldoni S, Screpanti I, Marconi P, Rosati E. Notch signaling sustains the expression of Mcl-1 and the activity of eIF4E to promote cell survival in CLL. Oncotarget 2016; 6:16559-72. [PMID: 26041884 PMCID: PMC4599289 DOI: 10.18632/oncotarget.4116] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/23/2015] [Indexed: 01/19/2023] Open
Abstract
In chronic lymphocytic leukemia (CLL), Notch1 and Notch2 signaling is constitutively activated and contributes to apoptosis resistance. We show that genetic inhibition of either Notch1 or Notch2, through small-interfering RNA, increases apoptosis of CLL cells and is associated with decreased levels of the anti-apoptotic protein Mcl-1. Thus, Notch signaling promotes CLL cell survival at least in part by sustaining Mcl-1 expression. In CLL cells, an enhanced Notch activation also contributes to the increase in Mcl-1 expression and cell survival induced by IL-4. Mcl-1 downregulation by Notch targeting is not due to reduced transcription or degradation by caspases, but in part, to increased degradation by the proteasome. Mcl-1 downregulation by Notch targeting is also accompanied by reduced phosphorylation of eukaryotic translation initiation factor 4E (eIF4E), suggesting that this protein is another target of Notch signaling in CLL cells. Overall, we show that Notch signaling sustains CLL cell survival by promoting Mcl-1 expression and eIF4E activity, and given the oncogenic role of these factors, we underscore the therapeutic potential of Notch inhibition in CLL.
Collapse
Affiliation(s)
- Filomena De Falco
- Department of Experimental Medicine, Biosciences and Medical Embryology Section, University of Perugia, Perugia, Italy
| | - Rita Sabatini
- Department of Experimental Medicine, Biosciences and Medical Embryology Section, University of Perugia, Perugia, Italy
| | - Beatrice Del Papa
- Department of Medicine, Hematology and Clinical Immunology Section, University of Perugia, Perugia, Italy
| | - Franca Falzetti
- Department of Medicine, Hematology and Clinical Immunology Section, University of Perugia, Perugia, Italy
| | - Mauro Di Ianni
- Department of Life, Health and Environmental Sciences, Hematology Section, University of L'Aquila, L'Aquila, Italy
| | - Paolo Sportoletti
- Department of Medicine, Hematology and Clinical Immunology Section, University of Perugia, Perugia, Italy
| | - Stefano Baldoni
- Department of Medicine, Hematology and Clinical Immunology Section, University of Perugia, Perugia, Italy
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Pierfrancesco Marconi
- Department of Experimental Medicine, Biosciences and Medical Embryology Section, University of Perugia, Perugia, Italy
| | - Emanuela Rosati
- Department of Experimental Medicine, Biosciences and Medical Embryology Section, University of Perugia, Perugia, Italy
| |
Collapse
|
35
|
Effects of induced pluripotent stem cells-derived conditioned medium on the proliferation and anti-apoptosis of human adipose-derived stem cells. Mol Cell Biochem 2016; 413:69-85. [DOI: 10.1007/s11010-015-2640-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/23/2015] [Indexed: 01/09/2023]
|
36
|
EIF2A-dependent translational arrest protects leukemia cells from the energetic stress induced by NAMPT inhibition. BMC Cancer 2015; 15:855. [PMID: 26542945 PMCID: PMC4636066 DOI: 10.1186/s12885-015-1845-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/23/2015] [Indexed: 01/04/2023] Open
Abstract
Background Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in NAD+ biosynthesis from nicotinamide, is one of the major factors regulating cancer cells metabolism and is considered a promising target for treating cancer. The prototypical NAMPT inhibitor FK866 effectively lowers NAD+ levels in cancer cells, reducing the activity of NAD+-dependent enzymes, lowering intracellular ATP, and promoting cell death. Results We show that FK866 induces a translational arrest in leukemia cells through inhibition of MTOR/4EBP1 signaling and of the initiation factors EIF4E and EIF2A. Specifically, treatment with FK866 is shown to induce 5′AMP-activated protein kinase (AMPK) activation, which, together with EIF2A phosphorylation, is responsible for the inhibition of protein synthesis. Notably, such an effect was also observed in patients’ derived primary leukemia cells including T-cell Acute Lymphoblastic Leukemia. Jurkat cells in which AMPK or LKB1 expression was silenced or in which a non-phosphorylatable EIF2A mutant was ectopically expressed showed enhanced sensitivity to the NAMPT inhibitor, confirming a key role for the LKB1-AMPK-EIF2A axis in cell fate determination in response to energetic stress via NAD+ depletion. Conclusions We identified EIF2A phosphorylation as a novel early molecular event occurring in response to NAMPT inhibition and mediating protein synthesis arrest. In addition, our data suggest that tumors exhibiting an impaired LBK1- AMPK- EIF2A response may be especially susceptible to NAMPT inhibitors and thus become an elective indication for this type of agents. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1845-1) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
Interleukin-24 inhibits influenza A virus replication in vitro through induction of toll-like receptor 3 dependent apoptosis. Antiviral Res 2015; 123:93-104. [DOI: 10.1016/j.antiviral.2015.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/31/2015] [Accepted: 09/06/2015] [Indexed: 11/22/2022]
|
38
|
Yu X, Xia W, Li S, Blumenfeld J, Zhang B, Yang J, Miao J, Gu ZJ. Antitumor effect and underlying mechanism of RGD-modified adenovirus mediated IL-24 expression on myeloid leukemia cells. Int Immunopharmacol 2015. [DOI: 10.1016/j.intimp.2015.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
39
|
Leclerc GJ, DeSalvo J, Du J, Gao N, Leclerc GM, Lehrman MA, Lampidis TJ, Barredo JC. Mcl-1 downregulation leads to the heightened sensitivity exhibited by BCR-ABL positive ALL to induction of energy and ER-stress. Leuk Res 2015; 39:S0145-2126(15)30360-X. [PMID: 26346348 PMCID: PMC4783293 DOI: 10.1016/j.leukres.2015.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/21/2015] [Accepted: 08/15/2015] [Indexed: 12/31/2022]
Abstract
BCR-ABL positive (+) acute lymphoblastic leukemia (ALL) accounts for ∼30% of cases of ALL. We recently demonstrated that 2-deoxy-d-glucose (2-DG), a dual energy (glycolysis inhibition) and ER-stress (N-linked-glycosylation inhibition) inducer, leads to cell death in ALL via ER-stress/UPR-mediated apoptosis. Among ALL subtypes, BCR-ABL+ ALL cells exhibited the highest sensitivity to 2-DG suggesting BCR-ABL expression may be linked to this increased vulnerability. To confirm the role of BCR-ABL, we constructed a NALM6/BCR-ABL stable cell line and found significant increase in 2-DG-induced apoptosis compared to control. We found that Mcl-1 was downregulated by agents inducing ER-stress and Mcl-1 levels correlated with ALL sensitivity. In addition, we showed that Mcl-1 expression is positively regulated by the MEK/ERK pathway, dependent on BCR-ABL, and further downregulated by combining ER-stressors with TKIs. We determined that energy/ER stressors led to translational repression of Mcl-1 via the AMPK/mTOR and UPR/PERK/eIF2α pathways. Taken together, our data indicate that BCR-ABL+ ALL exhibits heightened sensitivity to induction of energy and ER-stress through inhibition of the MEK/ERK pathway, and translational repression of Mcl-1 expression via AMPK/mTOR and UPR/PERK/eIF2α pathways. This study supports further consideration of strategies combining energy/ER-stress inducers with BCR-ABL TKIs for future clinical translation in BCR-ABL+ ALL patients.
Collapse
Affiliation(s)
- Guy J Leclerc
- Department of Pediatrics, Division of Hematology and Oncology, University of Miami Miller School of Medicine, Miami, FL 33101, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Joanna DeSalvo
- Department of Pediatrics, Division of Hematology and Oncology, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Jianfeng Du
- Department of Pediatrics, Division of Hematology and Oncology, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Ningguo Gao
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Gilles M Leclerc
- Department of Pediatrics, Division of Hematology and Oncology, University of Miami Miller School of Medicine, Miami, FL 33101, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Mark A Lehrman
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Theodore J Lampidis
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33101, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Julio C Barredo
- Department of Pediatrics, Division of Hematology and Oncology, University of Miami Miller School of Medicine, Miami, FL 33101, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33101, USA; Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33101, USA; Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33101, USA.
| |
Collapse
|
40
|
Bommer UA, Iadevaia V, Chen J, Knoch B, Engel M, Proud CG. Growth-factor dependent expression of the translationally controlled tumour protein TCTP is regulated through the PI3-K/Akt/mTORC1 signalling pathway. Cell Signal 2015; 27:1557-68. [PMID: 25936523 DOI: 10.1016/j.cellsig.2015.04.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/21/2015] [Accepted: 04/25/2015] [Indexed: 11/21/2022]
Abstract
Translationally controlled tumour protein TCTP (gene symbol: TPT1) is a highly-conserved, cyto-protective protein implicated in many physiological and disease processes, in particular cancer, where it is associated with poor patient outcomes. To understand the mechanisms underlying the accumulation of high TCTP levels in cancer cells, we studied the signalling pathways that control translation of TCTP mRNA, which contains a 5'-terminal oligopyrimidine tract (5'-TOP). In HT29 colon cancer cells and in HeLa cells, serum increases the expression of TCTP two- and four-fold, respectively, and this is inhibited by rapamycin or mTOR kinase inhibitors. Polysome profiling and mRNA quantification indicate that these effects occur at the level of mRNA translation. Blocking this pathway upstream of mTOR complex 1 (mTORC1) by inhibiting Akt also prevented increases in TCTP levels in both HeLa and HT29 colon cancer cells, whereas knockout of TSC2, a negative regulator of mTORC1, led to derepression of TCTP synthesis under serum starvation. Overexpression of eIF4E enhanced the polysomal association of the TCTP mRNA, although it did not protect its translation from inhibition by rapamycin. Conversely, expression of a constitutively-active mutant of the eIF4E inhibitor 4E-BP1, which is normally inactivated by mTORC1, inhibited TCTP mRNA translation in HEK293 cells. Our results demonstrate that TCTP mRNA translation is regulated by signalling through the PI3-K/Akt/mTORC1 pathway. This explains why TCTP levels are frequently increased in cancers, since mTORC1 signalling is hyperactive in ~80% of tumours.
Collapse
Affiliation(s)
- Ulrich-Axel Bommer
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2522 NSW, Australia; Graduate School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong 2522 NSW, Australia.
| | | | - Jiezhong Chen
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2522 NSW, Australia
| | - Bianca Knoch
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2522 NSW, Australia
| | - Martin Engel
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2522 NSW, Australia
| | | |
Collapse
|
41
|
Busacca S, Law EWP, Powley IR, Proia DA, Sequeira M, Le Quesne J, Klabatsa A, Edwards JM, Matchett KB, Luo JL, Pringle JH, El-Tanani M, MacFarlane M, Fennell DA. Resistance to HSP90 inhibition involving loss of MCL1 addiction. Oncogene 2015; 35:1483-92. [PMID: 26096930 PMCID: PMC4819782 DOI: 10.1038/onc.2015.213] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 03/02/2015] [Accepted: 03/14/2015] [Indexed: 12/21/2022]
Abstract
Inhibition of the chaperone heat-shock protein 90 (HSP90) induces apoptosis, and it is a promising anti-cancer strategy. The mechanisms underpinning apoptosis activation following HSP90 inhibition and how they are modified during acquired drug resistance are unknown. We show for the first time that, to induce apoptosis, HSP90 inhibition requires the cooperation of multi BH3-only proteins (BID, BIK, PUMA) and the reciprocal suppression of the pro-survival BCL-2 family member MCL1, which occurs via inhibition of STAT5A. A subset of tumour cell lines exhibit dependence on MCL1 expression for survival and this dependence is also associated with tumour response to HSP90 inhibition. In the acquired resistance setting, MCL1 suppression in response to HSP90 inhibitors is maintained; however, a switch in MCL1 dependence occurs. This can be exploited by the BH3 peptidomimetic ABT737, through non-BCL-2-dependent synthetic lethality.
Collapse
Affiliation(s)
- S Busacca
- Department of Cancer Studies, Cancer Research UK Leicester Centre, University of Leicester, Leicester, UK
| | - E W P Law
- Department of Cancer Studies, Cancer Research UK Leicester Centre, University of Leicester, Leicester, UK
| | | | - D A Proia
- Synta Pharmaceuticals Corp., Lexington, MA, USA
| | - M Sequeira
- Synta Pharmaceuticals Corp., Lexington, MA, USA
| | - J Le Quesne
- Department of Cancer Studies, Cancer Research UK Leicester Centre, University of Leicester, Leicester, UK.,MRC Toxicology Unit, Leicester, UK
| | - A Klabatsa
- Division of Cancer Studies, King's College London, London, UK
| | | | - K B Matchett
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - J L Luo
- Department of Cancer Studies, Cancer Research UK Leicester Centre, University of Leicester, Leicester, UK
| | - J H Pringle
- Department of Cancer Studies, Cancer Research UK Leicester Centre, University of Leicester, Leicester, UK
| | - M El-Tanani
- Institute of Cancer Therapeutics, School of Life Sciences, University of Bradford, Bradford, UK
| | | | - D A Fennell
- Department of Cancer Studies, Cancer Research UK Leicester Centre, University of Leicester, Leicester, UK
| |
Collapse
|
42
|
Auner HW, Cenci S. Recent advances and future directions in targeting the secretory apparatus in multiple myeloma. Br J Haematol 2015; 168:14-25. [PMID: 25296649 DOI: 10.1111/bjh.13172] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Multiple myeloma is a genetically heterogeneous tumour of transformed plasma cells, terminally differentiated effectors of the B cell lineage specialized in producing large amounts of immunoglobulins. The uniquely well-developed secretory apparatus that equips normal and transformed plasma cells with the capacity for high-level protein secretion constitutes a distinctive therapeutic target. In this review we discuss how fundamental cellular processes, such as the unfolded protein response (UPR), endoplasmic reticulum (ER)-associated degradation and autophagy, maintain intracellular protein homeostasis (proteostasis) and regulate plasma cell ontogeny and malignancy. We summarize our current understanding of the cellular effects of proteasome inhibitors and the molecular bases of resistance to them. Furthermore, we discuss how improvements in our understanding of the secretory apparatus and of the complex interactions between intracellular protein synthesis and degradation pathways can disclose novel drug targets for multiple myeloma, defining a paradigm of general interest for cancer biology and disorders of altered proteostasis.
Collapse
Affiliation(s)
- Holger W Auner
- Department of Medicine, Centre for Haematology, Imperial College London, London, UK
| | | |
Collapse
|
43
|
The role of NOS-mediated ROS accumulation in an early phase Cu-induced acute cytotoxicity in MCF-7 cells. Biometals 2014; 28:113-22. [PMID: 25403658 DOI: 10.1007/s10534-014-9807-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/29/2014] [Indexed: 10/24/2022]
Abstract
Copper (Cu) ion is essential for the biological systems, however, high level of CuCl2 exposure causes detrimental effects, which leads to cell apoptosis. Nitric oxide (NO) is an efficient cell signal messenger, which plays an important role in cell apoptosis. However, the potential mechanism of an early phase Cu-induced acute cytotoxicity through the nitric oxide synthase (NOS) signaling pathway and its interaction has not been studied. In this report, we provide data showing that high level of CuCl2 could rapidly decrease the NO production with the release of Ca(2+) and Zn(2+), and then modulate the transcriptional and translational expression of NOSs in MCF-7 cells. The reactive oxygen species (ROS) level in cells was increased after high level of CuCl2 exposure, which led to the alpha subunit of eukaryotic initiation factor 2 phosphorylation. By using the free radical scavenger N-acetyl-L-cysteine or the NOS substrate L-arginine, it demonstrated that NOS played a critical role on the Cu-induced ROS generation, which further led to the oxidative stress and cell apoptosis. These results suggested that Cu-induced apoptosis was associated with the oxidative stress, and through the NOS-mediated signaling pathway.
Collapse
|
44
|
Zhong L, Wang L, Xu L, Liu Q, Jiang L, Zhi Y, Lu W, Zhou P. The role of nitric oxide synthase signaling pathway in the Zn-induced cellular responses in MCF-7 cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 38:783-791. [PMID: 25305739 DOI: 10.1016/j.etap.2014.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/13/2014] [Accepted: 09/21/2014] [Indexed: 06/04/2023]
Abstract
Trace amount zinc plays key roles in biological systems, while in excessive amount it causes toxic effects. Evidence shows that there exists a crosstalk between NO and Zn apoptotic signal transduction pathway. However, the potential mechanism of Zn-induced cellular responses through the NOS signaling pathway has not been determined yet. In this research, trace amount ZnCl2 (1nM) could induce the NO production, however it appears that this influence does not extend to genetic level in MCF-7 cells. Whereas, excess ZnCl2 (100μM, 1mM) could lead to a decreased NO production first with the release of Ca(2+), and then induce the NO production with the transcriptional and translational activation of NOSs. The ROS generation was also induced by excess ZnCl2, causing the elF2α phosphorylation. The alleviation effect of N-acetyl-l-cysteine or l-arginine on the Zn-induced ROS generation and apoptosis suggested that Zn-induced apoptosis was associated with the NOS-mediated oxidative stress.
Collapse
Affiliation(s)
- Lingying Zhong
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China; School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Bor S. Luh Food Safety Research Center, Shanghai 200240, PR China
| | - Lumei Wang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai 200240, PR China; Bor S. Luh Food Safety Research Center, Shanghai 200240, PR China
| | - Lurong Xu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai 200240, PR China; Bor S. Luh Food Safety Research Center, Shanghai 200240, PR China
| | - Qunlu Liu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai 200240, PR China; Bor S. Luh Food Safety Research Center, Shanghai 200240, PR China
| | - Linlei Jiang
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai 200240, PR China; Bor S. Luh Food Safety Research Center, Shanghai 200240, PR China; Shanghai Food Safety Engineering Research Center, Shanghai 200240, PR China
| | - Yuee Zhi
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai 200240, PR China; Bor S. Luh Food Safety Research Center, Shanghai 200240, PR China
| | - Wei Lu
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai 200240, PR China; Bor S. Luh Food Safety Research Center, Shanghai 200240, PR China.
| | - Pei Zhou
- School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China; Key Laboratory of Urban Agriculture (South) of Ministry of Agriculture, Shanghai 200240, PR China; Bor S. Luh Food Safety Research Center, Shanghai 200240, PR China.
| |
Collapse
|
45
|
Rainbolt TK, Saunders JM, Wiseman RL. Stress-responsive regulation of mitochondria through the ER unfolded protein response. Trends Endocrinol Metab 2014; 25:528-37. [PMID: 25048297 DOI: 10.1016/j.tem.2014.06.007] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 12/31/2022]
Abstract
The endoplasmic reticulum (ER) and mitochondria form physical interactions involved in the regulation of biologic functions including mitochondrial bioenergetics and apoptotic signaling. To coordinate these functions during stress, cells must coregulate ER and mitochondria through stress-responsive signaling pathways such as the ER unfolded protein response (UPR). Although the UPR is traditionally viewed as a signaling pathway responsible for regulating ER proteostasis, it is becoming increasingly clear that the protein kinase RNA (PKR)-like endoplasmic reticulum kinase (PERK) signaling pathway within the UPR can also regulate mitochondria proteostasis and function in response to pathologic insults that induce ER stress. Here, we discuss the contributions of PERK in coordinating ER-mitochondrial activities and describe the mechanisms by which PERK adapts mitochondrial proteostasis and function in response to ER stress.
Collapse
Affiliation(s)
- T Kelly Rainbolt
- Department of Molecular and Experimental Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jaclyn M Saunders
- Department of Molecular and Experimental Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Luke Wiseman
- Department of Molecular and Experimental Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
46
|
Mcl-1 ubiquitination: unique regulation of an essential survival protein. Cells 2014; 3:418-37. [PMID: 24814761 PMCID: PMC4092850 DOI: 10.3390/cells3020418] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 04/16/2014] [Accepted: 04/29/2014] [Indexed: 01/26/2023] Open
Abstract
Mcl-1 is an anti-apoptotic protein of the Bcl-2 family that is essential for the survival of multiple cell lineages and that is highly amplified in human cancer. Under physiological conditions, Mcl-1 expression is tightly regulated at multiple levels, involving transcriptional, post-transcriptional and post-translational processes. Ubiquitination of Mcl-1, that targets it for proteasomal degradation, allows for rapid elimination of the protein and triggering of cell death, in response to various cellular events. In the last decade, a number of studies have elucidated different pathways controlling Mcl-1 ubiquitination and degradation. Four different E3 ubiquitin-ligases (e.g., Mule, SCFβ-TrCP, SCFFbw7 and Trim17) and one deubiquitinase (e.g., USP9X), that respectively mediate and oppose Mcl-1 ubiquitination, have been formerly identified. The interaction between Mule and Mcl-1 can be modulated by other Bcl-2 family proteins, while recognition of Mcl-1 by the other E3 ubiquitin-ligases and deubiquitinase is influenced by phosphorylation of specific residues in Mcl-1. The protein kinases and E3 ubiquitin-ligases that are involved in the regulation of Mcl-1 stability vary depending on the cellular context, highlighting the complexity and pivotal role of Mcl-1 regulation. In this review, we attempt to recapitulate progress in understanding Mcl-1 regulation by the ubiquitin-proteasome system.
Collapse
|
47
|
MDA-7/IL-24: multifunctional cancer killing cytokine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 818:127-53. [PMID: 25001534 DOI: 10.1007/978-1-4471-6458-6_6] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
First identified almost two decades ago as a novel gene differentially expressed in human melanoma cells induced to terminally differentiate, MDA-7/IL-24 has since shown great potential as an anti-cancer gene. MDA-7/IL24, a secreted protein of the IL-10 family, functions as a cytokine at normal physiological levels and is expressed in tissues of the immune system. At supra-physiological levels, MDA-7/IL-24 plays a prominent role in inhibiting tumor growth, invasion, metastasis and angiogenesis and was recently shown to target tumor stem/initiating cells for death. Much of the attention focused on MDA-7/IL-24 originated from the fact that it can selectively induce cell death in cancer cells without affecting normal cells. Thus, this gene originally shown to be associated with melanoma cell differentiation has now proven to be a multi-functional protein affecting a broad array of cancers. Moreover, MDA-7/IL-24 has proven efficacious in a Phase I/II clinical trial in humans with multiple advanced cancers. As research in the field progresses, we will unravel more of the functions of MDA-7/IL-24 and define novel ways to utilize MDA-7/IL-24 in the treatment of cancer.
Collapse
|
48
|
Fernald K, Kurokawa M. Evading apoptosis in cancer. Trends Cell Biol 2013; 23:620-33. [PMID: 23958396 DOI: 10.1016/j.tcb.2013.07.006] [Citation(s) in RCA: 390] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/16/2013] [Accepted: 07/17/2013] [Indexed: 12/12/2022]
Abstract
Carcinogenesis is a mechanistically complex and variable process with a plethora of underlying genetic causes. Cancer development comprises a multitude of steps that occur progressively starting with initial driver mutations leading to tumorigenesis and, ultimately, metastasis. During these transitions, cancer cells accumulate a series of genetic alterations that confer on the cells an unwarranted survival and proliferative advantage. During the course of development, however, cancer cells also encounter a physiologically ubiquitous cellular program that aims to eliminate damaged or abnormal cells: apoptosis. Thus, it is essential that cancer cells acquire instruments to circumvent programmed cell death. Here we discuss emerging evidence indicating how cancer cells adopt various strategies to override apoptosis, including amplifying the antiapoptotic machinery, downregulating the proapoptotic program, or both.
Collapse
Affiliation(s)
- Kaleigh Fernald
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | | |
Collapse
|
49
|
Juin P, Geneste O, Gautier F, Depil S, Campone M. Decoding and unlocking the BCL-2 dependency of cancer cells. Nat Rev Cancer 2013; 13:455-65. [PMID: 23783119 DOI: 10.1038/nrc3538] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer cells are subject to many apoptotic stimuli that would kill them were it not for compensatory prosurvival alterations. BCL-2-like (BCL-2L) proteins contribute to such aberrant behaviour by engaging a network of interactions that is potent at promoting survival but that is also fragile: inhibition of a restricted number of interactions may suffice to trigger cancer cell death. Currently available and novel compounds that inhibit these interactions could be efficient therapeutic agents if this phenotype of BCL-2L dependence was better understood at a molecular, cellular and systems level and if it could be diagnosed by relevant biomarkers.
Collapse
Affiliation(s)
- Philippe Juin
- Team 8 Cell survival and tumor escape in breast cancer, UMR 892 INSERM / 6299 CNRS / Université de Nantes, Institut de Recherche Thérapeutique de l'Université de Nantes, 8 quai Moncousu, BP 70721, 44007 Nantes Cedex, 1 France.
| | | | | | | | | |
Collapse
|
50
|
Lee SK, Kim YS. Phosphorylation of eIF2α attenuates statin-induced apoptosis by inhibiting the stabilization and translocation of p53 to the mitochondria. Int J Oncol 2013; 42:810-6. [PMID: 23354132 PMCID: PMC3597453 DOI: 10.3892/ijo.2013.1792] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 12/17/2012] [Indexed: 12/26/2022] Open
Abstract
Statins are effective cholesterol-lowering drugs that exert pleiotropic effects, including cytotoxicity to cancer cells. We previously reported that simvastatin triggered the mitochondrial apoptotic pathway in MethA fibrosarcoma cells, which was accompanied by the translocation of stabilized p53 to the mitochondria. In this study, we investigated whether statins induce the endoplasmic reticulum (ER) stress response and the mechanisms by which this response is linked to the stabilization of p53 and its translocation to the mitochondria. Statins induced typical ER stress-related proteins, such as BiP/78 kDa glucose-regulated protein (Grp78) and CCAAT/ enhancer-binding protein homologous protein (CHOP), as well as the phosphorylation of protein kinase RNA-like endoplasmic reticulum kinase (PERK), eIF2α and JNK. The statin-induced phosphorylation of eIF2α and JNK was inhibited by supplementation with components of the mevalonate pathway, such as mevalonate, farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP). Salubrinal, an inhibitor of the dephosphorylation of eIF2α, suppressed the loss of mitochondrial membrane potential and the translocation of stabilized p53 and Bax to the mitochondria; however, SP600125, a JNK kinase inhibitor, did not exert this effect. Furthermore, the eIF2α knockdown sensitized cells to simvastatin-induced apoptosis and the overexpression of a non-phosphorylatable eIF2α-mutant [serine 51(Ser51)/alanine] enhanced the stabilization of p53 and its translocation to the mitochondria in response to simvastatin treatment. Taken together, these data indicate that eIF2α phosphorylation in the context of the ER stress response plays a role in cell survival by counteracting the p53-mediated mitochondrial apoptosis in response to statins.
Collapse
Affiliation(s)
- Sang Kyu Lee
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 305-764, Republic of Korea
| | | |
Collapse
|