1
|
Li M. Half century of cancer research: In honor of Dr.rer.nat Manfred Schwab, the former Editor-in-Chief of Cancer Letters. Cancer Lett 2024; 585:216722. [PMID: 38341126 DOI: 10.1016/j.canlet.2024.216722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Affiliation(s)
- Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
2
|
Peramangalam PS, Surapally S, Veltri AJ, Zheng S, Burns R, Zhu N, Rao S, Muller-Tidow C, Bushweller JH, Pulikkan JA. N-MYC regulates cell survival via eIF4G1 in inv(16) acute myeloid leukemia. SCIENCE ADVANCES 2024; 10:eadh8493. [PMID: 38416825 PMCID: PMC10901375 DOI: 10.1126/sciadv.adh8493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 01/24/2024] [Indexed: 03/01/2024]
Abstract
N-MYC (encoded by MYCN) is a critical regulator of hematopoietic stem cell function. While the role of N-MYC deregulation is well established in neuroblastoma, the importance of N-MYC deregulation in leukemogenesis remains elusive. Here, we demonstrate that N-MYC is overexpressed in acute myeloid leukemia (AML) cells with chromosome inversion inv(16) and contributes to the survival and maintenance of inv(16) leukemia. We identified a previously unknown MYCN enhancer, active in multiple AML subtypes, essential for MYCN mRNA levels and survival in inv(16) AML cells. We also identified eukaryotic translation initiation factor 4 gamma 1 (eIF4G1) as a key N-MYC target that sustains leukemic survival in inv(16) AML cells. The oncogenic role of eIF4G1 in AML has not been reported before. Our results reveal a mechanism whereby N-MYC drives a leukemic transcriptional program and provides a rationale for the therapeutic targeting of the N-MYC/eIF4G1 axis in myeloid leukemia.
Collapse
Affiliation(s)
| | - Sridevi Surapally
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Anthony J. Veltri
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Shikan Zheng
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Robert Burns
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Nan Zhu
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sridhar Rao
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics, Division of Hematology, Oncology, and Transplantation, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Carsten Muller-Tidow
- Department of Medicine, Hematology, Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - John H. Bushweller
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - John A. Pulikkan
- Program in Stem Cell Biology and Hematopoiesis, Versiti Blood Research Institute, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
3
|
Schoof M, Godbole S, Albert TK, Dottermusch M, Walter C, Ballast A, Qin N, Olivera MB, Göbel C, Neyazi S, Holdhof D, Kresbach C, Peter LS, Epplen GD, Thaden V, Spohn M, Blattner-Johnson M, Modemann F, Mynarek M, Rutkowski S, Sill M, Varghese J, Afflerbach AK, Eckhardt A, Münter D, Verma A, Struve N, Jones DTW, Remke M, Neumann JE, Kerl K, Schüller U. Mouse models of pediatric high-grade gliomas with MYCN amplification reveal intratumoral heterogeneity and lineage signatures. Nat Commun 2023; 14:7717. [PMID: 38001143 PMCID: PMC10673884 DOI: 10.1038/s41467-023-43564-w] [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: 04/02/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Pediatric high-grade gliomas of the subclass MYCN (HGG-MYCN) are highly aggressive tumors frequently carrying MYCN amplifications, TP53 mutations, or both alterations. Due to their rarity, such tumors have only recently been identified as a distinct entity, and biological as well as clinical characteristics have not been addressed specifically. To gain insights into tumorigenesis and molecular profiles of these tumors, and to ultimately suggest alternative treatment options, we generated a genetically engineered mouse model by breeding hGFAP-cre::Trp53Fl/Fl::lsl-MYCN mice. All mice developed aggressive forebrain tumors early in their lifetime that mimic human HGG-MYCN regarding histology, DNA methylation, and gene expression. Single-cell RNA sequencing revealed a high intratumoral heterogeneity with neuronal and oligodendroglial lineage signatures. High-throughput drug screening using both mouse and human tumor cells finally indicated high efficacy of Doxorubicin, Irinotecan, and Etoposide as possible therapy options that children with HGG-MYCN might benefit from.
Collapse
Affiliation(s)
- Melanie Schoof
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Shweta Godbole
- Center for Molecular Neurobiology (ZMNH), University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas K Albert
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Matthias Dottermusch
- Center for Molecular Neurobiology (ZMNH), University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
- Institute of Neuropathology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Muenster, Muenster, Germany
| | - Annika Ballast
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Nan Qin
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Institute of Neuropathology, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- High-Throughput Drug Screening Core Facility, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Marlena Baca Olivera
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Institute of Neuropathology, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- High-Throughput Drug Screening Core Facility, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Carolin Göbel
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Sina Neyazi
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Dörthe Holdhof
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Catena Kresbach
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
- Institute of Neuropathology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4 University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Levke-Sophie Peter
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Gefion Dorothea Epplen
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Thaden
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Spohn
- Research Institute Children's Cancer Center, Hamburg, Germany
| | - Mirjam Blattner-Johnson
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Franziska Modemann
- Mildred Scheel Cancer Career Center HaTriCS4 University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Oncology, Hematology and Bone marrow transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Mynarek
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4 University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Rutkowski
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Sill
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Muenster, Muenster, Germany
| | - Ann-Kristin Afflerbach
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Alicia Eckhardt
- Research Institute Children's Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
- Department of Radiotherapy & Radiation Oncology, Hubertus Wald Tumorzentrum-University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Münter
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Archana Verma
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Nina Struve
- Mildred Scheel Cancer Career Center HaTriCS4 University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Radiotherapy & Radiation Oncology, Hubertus Wald Tumorzentrum-University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc Remke
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Institute of Neuropathology, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- High-Throughput Drug Screening Core Facility, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Julia E Neumann
- Center for Molecular Neurobiology (ZMNH), University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
- Institute of Neuropathology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Kornelius Kerl
- Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - Ulrich Schüller
- Research Institute Children's Cancer Center, Hamburg, Germany.
- Department of Pediatric Hematology and Oncology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany.
- Institute of Neuropathology, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
4
|
Jha RK, Kouzine F, Levens D. MYC function and regulation in physiological perspective. Front Cell Dev Biol 2023; 11:1268275. [PMID: 37941901 PMCID: PMC10627926 DOI: 10.3389/fcell.2023.1268275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/12/2023] [Indexed: 11/10/2023] Open
Abstract
MYC, a key member of the Myc-proto-oncogene family, is a universal transcription amplifier that regulates almost every physiological process in a cell including cell cycle, proliferation, metabolism, differentiation, and apoptosis. MYC interacts with several cofactors, chromatin modifiers, and regulators to direct gene expression. MYC levels are tightly regulated, and deregulation of MYC has been associated with numerous diseases including cancer. Understanding the comprehensive biology of MYC under physiological conditions is an utmost necessity to demark biological functions of MYC from its pathological functions. Here we review the recent advances in biological mechanisms, functions, and regulation of MYC. We also emphasize the role of MYC as a global transcription amplifier.
Collapse
Affiliation(s)
| | | | - David Levens
- Gene Regulation Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD, United States
| |
Collapse
|
5
|
Deng Z, Richardson DR. The Myc Family and the Metastasis Suppressor NDRG1: Targeting Key Molecular Interactions with Innovative Therapeutics. Pharmacol Rev 2023; 75:1007-1035. [PMID: 37280098 DOI: 10.1124/pharmrev.122.000795] [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: 12/06/2022] [Revised: 03/07/2023] [Accepted: 05/01/2023] [Indexed: 06/08/2023] Open
Abstract
Cancer is a leading cause of death worldwide, resulting in ∼10 million deaths in 2020. Major oncogenic effectors are the Myc proto-oncogene family, which consists of three members including c-Myc, N-Myc, and L-Myc. As a pertinent example of the role of the Myc family in tumorigenesis, amplification of MYCN in childhood neuroblastoma strongly correlates with poor patient prognosis. Complexes between Myc oncoproteins and their partners such as hypoxia-inducible factor-1α and Myc-associated protein X (MAX) result in proliferation arrest and pro-proliferative effects, respectively. Interactions with other proteins are also important for N-Myc activity. For instance, the enhancer of zest homolog 2 (EZH2) binds directly to N-Myc to stabilize it by acting as a competitor against the ubiquitin ligase, SCFFBXW7, which prevents proteasomal degradation. Heat shock protein 90 may also be involved in N-Myc stabilization since it binds to EZH2 and prevents its degradation. N-Myc downstream-regulated gene 1 (NDRG1) is downregulated by N-Myc and participates in the regulation of cellular proliferation via associating with other proteins, such as glycogen synthase kinase-3β and low-density lipoprotein receptor-related protein 6. These molecular interactions provide a better understanding of the biologic roles of N-Myc and NDRG1, which can be potentially used as therapeutic targets. In addition to directly targeting these proteins, disrupting their key interactions may also be a promising strategy for anti-cancer drug development. This review examines the interactions between the Myc proteins and other molecules, with a special focus on the relationship between N-Myc and NDRG1 and possible therapeutic interventions. SIGNIFICANCE STATEMENT: Neuroblastoma is one of the most common childhood solid tumors, with a dismal five-year survival rate. This problem makes it imperative to discover new and more effective therapeutics. The molecular interactions between major oncogenic drivers of the Myc family and other key proteins; for example, the metastasis suppressor, NDRG1, may potentially be used as targets for anti-neuroblastoma drug development. In addition to directly targeting these proteins, disrupting their key molecular interactions may also be promising for drug discovery.
Collapse
Affiliation(s)
- Zhao Deng
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia (Z.D., D.R.R.), and Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan (D.R.R.)
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Australia (Z.D., D.R.R.), and Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan (D.R.R.)
| |
Collapse
|
6
|
Li J, Peng J, Wu L, Shen X, Zhen X, Zhang Y, Ma H, Xu Y, Xiong Q, Zhu Q, Zhang P. The deubiquitinase USP28 maintains the expression of the transcription factor MYCN and is essential in neuroblastoma cells. J Biol Chem 2023; 299:104856. [PMID: 37230388 PMCID: PMC10404617 DOI: 10.1016/j.jbc.2023.104856] [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: 12/27/2022] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Neuroblastoma (NB) is one of the most common extracranial solid tumors in children. MYCN gene amplification is highly associated with poor prognosis in high-risk NB patients. In non-MYCN-amplified high-risk NB patients, the expression of c-MYC (MYCC) and its target genes is highly elevated. USP28 as a deubiquitinase is known to regulate the stability of MYCC. We show here USP28 also regulates the stability of MYCN. Genetic depletion or pharmacologic inhibition of the deubiquitinase strongly destabilizes MYCN and stops the growth of NB cells that overexpress MYCN. In addition, MYCC could be similarly destabilized in non-MYCN NB cells by compromising USP28 function. Our results strongly suggest USP28 as a therapeutic target for NB with or without MYCN amplification/overexpression.
Collapse
Affiliation(s)
- Junjun Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Peng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lingzhi Wu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiang Shen
- Chaser Therapeutics Inc., Hangzhou, Zhejiang, China
| | - Xinghua Zhen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yimao Zhang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Huailu Ma
- Institute of Translational Medicine, Zhejiang University Medical School, Hangzhou, Zhejiang, China
| | - Yongfeng Xu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Qunli Xiong
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Zhu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China.
| | - Pumin Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Institute of Translational Medicine, Zhejiang University Medical School, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Sun L, Li X, Tu L, Stucky A, Huang C, Chen X, Cai J, Li SC. RNA-Sequencing Combined With Genome-Wide Allele-Specific Expression Patterning Identifies ZNF44 Variants as a Potential New Driver Gene for Pediatric Neuroblastoma. Cancer Control 2023; 30:10732748231175017. [PMID: 37161925 DOI: 10.1177/10732748231175017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
INTRODUCTION Neuroblastoma (NB) is one of the children's most common solid tumors, accounting for approximately 8% of pediatric malignancies and 15% of childhood cancer deaths. Somatic mutations in several genes, such as ALK, have been associated with NB progression and can facilitate the discovery of novel therapeutic strategies. However, the differential expression of mutated and wild-type alleles on the transcriptome level is poorly studied. METHODS This study analyzed 219 whole-exome sequencing datasets with somatic mutations detected by MuTect from paired normal and tumor samples. RESULTS We prioritized mutations in 8 candidate genes (RIMS4, RUSC2, ALK, MYCN, PTPN11, ALOX12B, ZNF44, and CNGB1) as potential driver mutations. We further confirmed the presence of allele-specific expression of the somatic mutations in NB with integrated analysis of 127 RNA-seq samples (of which 85 also had DNA-seq data available), including MYCN, ALK, and PTPN11. The allele-specific expression of mutations suggests that the same somatic mutation may have different effects on the clinical outcomes of tumors. CONCLUSION Our study suggests 2 novel variants of ZNF44 as a novel candidate driver gene for NB.
Collapse
Affiliation(s)
- Lan Sun
- Department of Oncology,Bishan Hospital of Chongqing Medical University, The People's Hospital of Bishan District, Chongqing, China
| | - Xiaoqing Li
- Department of Oncology,Bishan Hospital of Chongqing Medical University, The People's Hospital of Bishan District, Chongqing, China
| | - Lingli Tu
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andres Stucky
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chuan Huang
- Department of Oncology,Bishan Hospital of Chongqing Medical University, The People's Hospital of Bishan District, Chongqing, China
| | - Xuelian Chen
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jin Cai
- Department of Oral and Maxillofacial Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Shengwen Calvin Li
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children's Research Institute, Children's Hospital of Orange County (CHOC), Orange, CA, USA
- Department of Neurology, University of California, Irvine School of Medicine, Orange, CA, USA
| |
Collapse
|
8
|
Jiang Y. Contribution of Microhomology to Genome Instability: Connection between DNA Repair and Replication Stress. Int J Mol Sci 2022; 23:12937. [PMID: 36361724 PMCID: PMC9657218 DOI: 10.3390/ijms232112937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/23/2022] [Accepted: 10/23/2022] [Indexed: 11/23/2023] Open
Abstract
Microhomology-mediated end joining (MMEJ) is a highly mutagenic pathway to repair double-strand breaks (DSBs). MMEJ was thought to be a backup pathway of homologous recombination (HR) and canonical nonhomologous end joining (C-NHEJ). However, it attracts more attention in cancer research due to its special function of microhomology in many different aspects of cancer. In particular, it is initiated with DNA end resection and upregulated in homologous recombination-deficient cancers. In this review, I summarize the following: (1) the recent findings and contributions of MMEJ to genome instability, including phenotypes relevant to MMEJ; (2) the interaction between MMEJ and other DNA repair pathways; (3) the proposed mechanistic model of MMEJ in DNA DSB repair and a new connection with microhomology-mediated break-induced replication (MMBIR); and (4) the potential clinical application by targeting MMEJ based on synthetic lethality for cancer therapy.
Collapse
Affiliation(s)
- Yuning Jiang
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA 22903, USA
| |
Collapse
|
9
|
Pharmacophore-Model-Based Virtual-Screening Approaches Identified Novel Natural Molecular Candidates for Treating Human Neuroblastoma. Curr Issues Mol Biol 2022; 44:4838-4858. [PMID: 36286044 PMCID: PMC9600652 DOI: 10.3390/cimb44100329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/30/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
The mortality of cancer patients with neuroblastoma is increasing due to the limited availability of specific treatment options. Few drug candidates for combating neuroblastoma have been developed, and identifying novel therapeutic candidates against the disease is an urgent issue. It has been found that muc-N protein is amplified in one-third of human neuroblastomas and expressed as an attractive drug target against the disease. The myc-N protein interferes with the bromodomain and extraterminal (BET) family proteins. Pharmacologically inhibition of the protein potently depletes MYCN in neuroblastoma cells. BET inhibitors target MYCN transcription and show therapeutic efficacy against neuroblastoma. Therefore, the study aimed to identify potential inhibitors against the BET family protein, specifically Brd4 (brodamine-containing protein 4), to hinder the activity of neuroblastoma cells. To identify effective molecular candidates against the disease, a structure-based pharmacophore model was created for the binding site of the Brd4 protein. The pharmacophore model generated from the protein Brd4 was validated to screen potential natural active compounds. The compounds identified through the pharmacophore-model-based virtual-screening process were further screened through molecular docking, ADME (absorption, distribution, metabolism, and excretion), toxicity, and molecular dynamics (MD) simulation approach. The pharmacophore-model-based screening process initially identified 136 compounds, further evaluated based on molecular docking, ADME analysis, and toxicity approaches, identifying four compounds with good binding affinity and lower side effects. The stability of the selected compounds was also confirmed by dynamic simulation and molecular mechanics with generalized Born and surface area solvation (MM-GBSA) methods. Finally, the study identified four natural lead compounds, ZINC2509501, ZINC2566088, ZINC1615112, and ZINC4104882, that will potentially inhibit the activity of the desired protein and help to fight against neuroblastoma and related diseases. However, further evaluations through in vitro and in vivo assays are suggested to identify their efficacy against the desired protein and disease.
Collapse
|
10
|
Decaesteker B, Durinck K, Van Roy N, De Wilde B, Van Neste C, Van Haver S, Roberts S, De Preter K, Vermeirssen V, Speleman F. From DNA Copy Number Gains and Tumor Dependencies to Novel Therapeutic Targets for High-Risk Neuroblastoma. J Pers Med 2021; 11:1286. [PMID: 34945759 PMCID: PMC8707517 DOI: 10.3390/jpm11121286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma is a pediatric tumor arising from the sympatho-adrenal lineage and a worldwide leading cause of childhood cancer-related deaths. About half of high-risk patients die from the disease while survivors suffer from multiple therapy-related side-effects. While neuroblastomas present with a low mutational burden, focal and large segmental DNA copy number aberrations are highly recurrent and associated with poor survival. It can be assumed that the affected chromosomal regions contain critical genes implicated in neuroblastoma biology and behavior. More specifically, evidence has emerged that several of these genes are implicated in tumor dependencies thus potentially providing novel therapeutic entry points. In this review, we briefly review the current status of recurrent DNA copy number aberrations in neuroblastoma and provide an overview of the genes affected by these genomic variants for which a direct role in neuroblastoma has been established. Several of these genes are implicated in networks that positively regulate MYCN expression or stability as well as cell cycle control and apoptosis. Finally, we summarize alternative approaches to identify and prioritize candidate copy-number driven dependency genes for neuroblastoma offering novel therapeutic opportunities.
Collapse
Grants
- P30 CA008748 NCI NIH HHS
- G087221N, G.0507.12, G049720N,12U4718N, 11C3921N, 11J8313N, 12B5313N, 1514215N, 1197617N,1238420N, 12Q8322N, 3F018519, 12N6917N Fund for Scientific Research Flanders
- 2018-087, 2018-125, 2020-112 Belgian Foundation against Cancer
Collapse
Affiliation(s)
- Bieke Decaesteker
- Department for Biomolecular Medicine, Ghent University, Medical Research Building (MRB1), Corneel Heymanslaan 10, B-9000 Ghent, Belgium; (B.D.); (K.D.); (N.V.R.); (B.D.W.); (C.V.N.); (S.V.H.); (K.D.P.); (V.V.)
| | - Kaat Durinck
- Department for Biomolecular Medicine, Ghent University, Medical Research Building (MRB1), Corneel Heymanslaan 10, B-9000 Ghent, Belgium; (B.D.); (K.D.); (N.V.R.); (B.D.W.); (C.V.N.); (S.V.H.); (K.D.P.); (V.V.)
| | - Nadine Van Roy
- Department for Biomolecular Medicine, Ghent University, Medical Research Building (MRB1), Corneel Heymanslaan 10, B-9000 Ghent, Belgium; (B.D.); (K.D.); (N.V.R.); (B.D.W.); (C.V.N.); (S.V.H.); (K.D.P.); (V.V.)
| | - Bram De Wilde
- Department for Biomolecular Medicine, Ghent University, Medical Research Building (MRB1), Corneel Heymanslaan 10, B-9000 Ghent, Belgium; (B.D.); (K.D.); (N.V.R.); (B.D.W.); (C.V.N.); (S.V.H.); (K.D.P.); (V.V.)
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Corneel Heymanslaan 10, B-9000 Ghent, Belgium
| | - Christophe Van Neste
- Department for Biomolecular Medicine, Ghent University, Medical Research Building (MRB1), Corneel Heymanslaan 10, B-9000 Ghent, Belgium; (B.D.); (K.D.); (N.V.R.); (B.D.W.); (C.V.N.); (S.V.H.); (K.D.P.); (V.V.)
| | - Stéphane Van Haver
- Department for Biomolecular Medicine, Ghent University, Medical Research Building (MRB1), Corneel Heymanslaan 10, B-9000 Ghent, Belgium; (B.D.); (K.D.); (N.V.R.); (B.D.W.); (C.V.N.); (S.V.H.); (K.D.P.); (V.V.)
| | - Stephen Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Katleen De Preter
- Department for Biomolecular Medicine, Ghent University, Medical Research Building (MRB1), Corneel Heymanslaan 10, B-9000 Ghent, Belgium; (B.D.); (K.D.); (N.V.R.); (B.D.W.); (C.V.N.); (S.V.H.); (K.D.P.); (V.V.)
| | - Vanessa Vermeirssen
- Department for Biomolecular Medicine, Ghent University, Medical Research Building (MRB1), Corneel Heymanslaan 10, B-9000 Ghent, Belgium; (B.D.); (K.D.); (N.V.R.); (B.D.W.); (C.V.N.); (S.V.H.); (K.D.P.); (V.V.)
- Department of Biomedical Molecular Biology, Ghent University, Technologiepark 71, B-9052 Zwijnaarde, Belgium
| | - Frank Speleman
- Department for Biomolecular Medicine, Ghent University, Medical Research Building (MRB1), Corneel Heymanslaan 10, B-9000 Ghent, Belgium; (B.D.); (K.D.); (N.V.R.); (B.D.W.); (C.V.N.); (S.V.H.); (K.D.P.); (V.V.)
| |
Collapse
|
11
|
Defining Pathological Activities of ALK in Neuroblastoma, a Neural Crest-Derived Cancer. Int J Mol Sci 2021; 22:ijms222111718. [PMID: 34769149 PMCID: PMC8584162 DOI: 10.3390/ijms222111718] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma is a common extracranial solid tumour of childhood, responsible for 15% of cancer-related deaths in children. Prognoses vary from spontaneous remission to aggressive disease with extensive metastases, where treatment is challenging. Tumours are thought to arise from sympathoadrenal progenitor cells, which derive from an embryonic cell population called neural crest cells that give rise to diverse cell types, such as facial bone and cartilage, pigmented cells, and neurons. Tumours are found associated with mature derivatives of neural crest, such as the adrenal medulla or paraspinal ganglia. Sympathoadrenal progenitor cells express anaplastic lymphoma kinase (ALK), which encodes a tyrosine kinase receptor that is the most frequently mutated gene in neuroblastoma. Activating mutations in the kinase domain are common in both sporadic and familial cases. The oncogenic role of ALK has been extensively studied, but little is known about its physiological role. Recent studies have implicated ALK in neural crest migration and sympathetic neurogenesis. However, very few downstream targets of ALK have been identified. Here, we describe pathological activation of ALK in the neural crest, which promotes proliferation and migration, while preventing differentiation, thus inducing the onset of neuroblastoma. Understanding the effects of ALK activity on neural crest cells will help find new targets for neuroblastoma treatment.
Collapse
|
12
|
Qin XY, Gailhouste L. Non-Genomic Control of Dynamic MYCN Gene Expression in Liver Cancer. Front Oncol 2021; 10:618515. [PMID: 33937011 PMCID: PMC8085327 DOI: 10.3389/fonc.2020.618515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/23/2020] [Indexed: 11/13/2022] Open
Abstract
Upregulated MYCN gene expression is restricted to specialized cell populations such as EpCAM+ cancer stem cells in liver cancer, regardless of DNA amplification and mutation. Here, we reviewed the role of MYCN gene expression in liver homeostasis, regeneration, and tumorigenesis, and discussed the potential non-genomic mechanisms involved in controlling MYCN gene expression in liver cancer, with a focus on inflammation-mediated signal transduction and microRNA-associated post-transcriptional regulation. We concluded that dynamic MYCN gene expression is an integrated consequence of multiple signals in the tumor microenvironment, including tumor growth-promoting signals, lipid desaturation-mediated endoplasmic reticulum stress adaptation signals, and tumor suppressive miRNAs, making it a potential predictive biomarker of tumor stemness and plasticity. Therefore, understanding and tracing the dynamic changes and functions of MYCN gene expression will shed light on the origin of liver tumorigenesis at the cellular level and the development of novel therapeutic and diagnostic strategies for liver cancer treatment.
Collapse
Affiliation(s)
- Xian-Yang Qin
- Liver Cancer Prevention Research Unit, RIKEN Cluster for Pioneering Research, Wako, Japan
| | - Luc Gailhouste
- Liver Cancer Prevention Research Unit, RIKEN Cluster for Pioneering Research, Wako, Japan
| |
Collapse
|
13
|
Liu Z, Chen SS, Clarke S, Veschi V, Thiele CJ. Targeting MYCN in Pediatric and Adult Cancers. Front Oncol 2021; 10:623679. [PMID: 33628735 PMCID: PMC7898977 DOI: 10.3389/fonc.2020.623679] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/14/2020] [Indexed: 12/18/2022] Open
Abstract
The deregulation of the MYC family of oncogenes, including c-MYC, MYCN and MYCL occurs in many types of cancers, and is frequently associated with a poor prognosis. The majority of functional studies have focused on c-MYC due to its broad expression profile in human cancers. The existence of highly conserved functional domains between MYCN and c-MYC suggests that MYCN participates in similar activities. MYC encodes a basic helix-loop-helix-leucine zipper (bHLH-LZ) transcription factor (TF) whose central oncogenic role in many human cancers makes it a highly desirable therapeutic target. Historically, as a TF, MYC has been regarded as “undruggable”. Thus, recent efforts focus on investigating methods to indirectly target MYC to achieve anti-tumor effects. This review will primarily summarize the recent progress in understanding the function of MYCN. It will explore efforts at targeting MYCN, including strategies aimed at suppression of MYCN transcription, destabilization of MYCN protein, inhibition of MYCN transcriptional activity, repression of MYCN targets and utilization of MYCN overexpression dependent synthetic lethality.
Collapse
Affiliation(s)
- Zhihui Liu
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Samuel S Chen
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Saki Clarke
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Veronica Veschi
- Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Carol J Thiele
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
14
|
Borenäs M, Umapathy G, Lai W, Lind DE, Witek B, Guan J, Mendoza‐Garcia P, Masudi T, Claeys A, Chuang T, El Wakil A, Arefin B, Fransson S, Koster J, Johansson M, Gaarder J, Van den Eynden J, Hallberg B, Palmer RH. ALK ligand ALKAL2 potentiates MYCN-driven neuroblastoma in the absence of ALK mutation. EMBO J 2021; 40:e105784. [PMID: 33411331 PMCID: PMC7849294 DOI: 10.15252/embj.2020105784] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/19/2020] [Accepted: 10/23/2020] [Indexed: 12/18/2022] Open
Abstract
High-risk neuroblastoma (NB) is responsible for a disproportionate number of childhood deaths due to cancer. One indicator of high-risk NB is amplification of the neural MYC (MYCN) oncogene, which is currently therapeutically intractable. Identification of anaplastic lymphoma kinase (ALK) as an NB oncogene raised the possibility of using ALK tyrosine kinase inhibitors (TKIs) in treatment of patients with activating ALK mutations. 8-10% of primary NB patients are ALK-positive, a figure that increases in the relapsed population. ALK is activated by the ALKAL2 ligand located on chromosome 2p, along with ALK and MYCN, in the "2p-gain" region associated with NB. Dysregulation of ALK ligand in NB has not been addressed, although one of the first oncogenes described was v-sis that shares > 90% homology with PDGF. Therefore, we tested whether ALKAL2 ligand could potentiate NB progression in the absence of ALK mutation. We show that ALKAL2 overexpression in mice drives ALK TKI-sensitive NB in the absence of ALK mutation, suggesting that additional NB patients, such as those exhibiting 2p-gain, may benefit from ALK TKI-based therapeutic intervention.
Collapse
Affiliation(s)
- Marcus Borenäs
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Ganesh Umapathy
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Wei‐Yun Lai
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Dan E Lind
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Barbara Witek
- Department of Molecular BiologyUmeå UniversityUmeåSweden
| | - Jikui Guan
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Children's Hospital Affiliated to Zhengzhou UniversityZhengzhouChina
| | - Patricia Mendoza‐Garcia
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Tafheem Masudi
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Arne Claeys
- Department of Human Structure and Repair, Anatomy and Embryology UnitGhent UniversityGhentBelgium
| | - Tzu‐Po Chuang
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Abeer El Wakil
- Department of Molecular BiologyUmeå UniversityUmeåSweden
- Present address:
Department of Biological SciencesAlexandria UniversityAlexandriaEgypt
| | - Badrul Arefin
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Susanne Fransson
- Laboratory MedicineInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Jan Koster
- Department of OncogenomicsAcademic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Mathias Johansson
- Clinical GenomicsScience for life laboratoryUniversity of GothenburgGothenburgSweden
| | - Jennie Gaarder
- Laboratory MedicineInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Jimmy Van den Eynden
- Department of Human Structure and Repair, Anatomy and Embryology UnitGhent UniversityGhentBelgium
| | - Bengt Hallberg
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Ruth H Palmer
- Department of Medical Biochemistry and Cell BiologyInstitute of BiomedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
15
|
Arlt B, Zasada C, Baum K, Wuenschel J, Mastrobuoni G, Lodrini M, Astrahantseff K, Winkler A, Schulte JH, Finkler S, Forbes M, Hundsdoerfer P, Guergen D, Hoffmann J, Wolf J, Eggert A, Kempa S, Deubzer HE. Inhibiting phosphoglycerate dehydrogenase counteracts chemotherapeutic efficacy against MYCN-amplified neuroblastoma. Int J Cancer 2020; 148:1219-1232. [PMID: 33284994 DOI: 10.1002/ijc.33423] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/03/2020] [Accepted: 11/11/2020] [Indexed: 01/12/2023]
Abstract
Here we sought metabolic alterations specifically associated with MYCN amplification as nodes to indirectly target the MYCN oncogene. Liquid chromatography-mass spectrometry-based proteomics identified seven proteins consistently correlated with MYCN in proteomes from 49 neuroblastoma biopsies and 13 cell lines. Among these was phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme in de novo serine synthesis. MYCN associated with two regions in the PHGDH promoter, supporting transcriptional PHGDH regulation by MYCN. Pulsed stable isotope-resolved metabolomics utilizing 13 C-glucose labeling demonstrated higher de novo serine synthesis in MYCN-amplified cells compared to cells with diploid MYCN. An independence of MYCN-amplified cells from exogenous serine and glycine was demonstrated by serine and glycine starvation, which attenuated nucleotide pools and proliferation only in cells with diploid MYCN but did not diminish these endpoints in MYCN-amplified cells. Proliferation was attenuated in MYCN-amplified cells by CRISPR/Cas9-mediated PHGDH knockout or treatment with PHGDH small molecule inhibitors without affecting cell viability. PHGDH inhibitors administered as single-agent therapy to NOG mice harboring patient-derived MYCN-amplified neuroblastoma xenografts slowed tumor growth. However, combining a PHGDH inhibitor with the standard-of-care chemotherapy drug, cisplatin, revealed antagonism of chemotherapy efficacy in vivo. Emergence of chemotherapy resistance was confirmed in the genetic PHGDH knockout model in vitro. Altogether, PHGDH knockout or inhibition by small molecules consistently slows proliferation, but stops short of killing the cells, which then establish resistance to classical chemotherapy. Although PHGDH inhibition with small molecules has produced encouraging results in other preclinical cancer models, this approach has limited attractiveness for patients with neuroblastoma.
Collapse
Affiliation(s)
- Birte Arlt
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Anna-Louisa-Karsch-Straβe 2, 10178, Berlin, Germany.,Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Christin Zasada
- Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Katharina Baum
- Mathematical Modelling of Cellular Processes, Max-Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Straβe 10, 13125, Berlin, Germany
| | - Jasmin Wuenschel
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Guido Mastrobuoni
- Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Marco Lodrini
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Kathy Astrahantseff
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Annika Winkler
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Anna-Louisa-Karsch-Straβe 2, 10178, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Finkler
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Martin Forbes
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany.,Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Patrick Hundsdoerfer
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Department of Pediatric Oncology, Helios Klinikum Berlin Buch, Schwanebecker Chaussee 50, 13125, Berlin, Germany
| | - Dennis Guergen
- Experimental Pharmacology and Oncology Berlin-Buch GmbH (EPO), Robert-Rössle-Straβe 10, 13125, Berlin, Germany
| | - Jens Hoffmann
- Experimental Pharmacology and Oncology Berlin-Buch GmbH (EPO), Robert-Rössle-Straβe 10, 13125, Berlin, Germany
| | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max-Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Robert-Rössle-Straβe 10, 13125, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Anna-Louisa-Karsch-Straβe 2, 10178, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Kempa
- Integrative Proteomics and Metabolomics, Berlin Institute for Medical Systems Biology at the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Hannoversche Str. 28, 10115, Berlin, Germany
| | - Hedwig E Deubzer
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Lindenberger Weg 80, 13125, Berlin, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Anna-Louisa-Karsch-Straβe 2, 10178, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
16
|
Valter K, Maximchik P, Abdrakhmanov A, Senichkin V, Zhivotovsky B, Gogvadze V. Distinct effects of etoposide on glutamine-addicted neuroblastoma. Cell Mol Life Sci 2020; 77:1197-1207. [PMID: 31392350 PMCID: PMC7109159 DOI: 10.1007/s00018-019-03232-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022]
Abstract
The majority of anticancer drugs are DNA-damaging agents, and whether or not they may directly target mitochondria remains unclear. In addition, tumors such as neuroblastoma exhibit addiction to glutamine in spite of it being a nonessential amino acid. Our aim was to evaluate the direct effect of widely used anticancer drugs on mitochondrial activity in combination with glutamine withdrawal, and possible apoptotic effects of such interaction. Our results revealed that etoposide inhibits mitochondrial respiratory chain Complex I causing the leakage of electrons and the superoxide radical formation. However, it was not sufficient to induce apoptosis, and apoptotic manifestation was detectable only alongside the withdrawal of glutamine, a precursor for antioxidant glutathione. Thus, the simultaneous depletion of glutathione and destabilization of mitochondria by ROS can compromise the barrier properties of the mitochondrial membrane, leading to cytochrome c release and the activation of the mitochondrial apoptotic pathway. Thus, the depletion of antioxidants or the inhibition of the pathways responsible for cellular antioxidant response can enhance mitochondrial targeting and strengthen antitumor therapy.
Collapse
Affiliation(s)
- Kadri Valter
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 171 77, Stockholm, Sweden
| | - Polina Maximchik
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Alibek Abdrakhmanov
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Viacheslav Senichkin
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Boris Zhivotovsky
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 171 77, Stockholm, Sweden
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Vladimir Gogvadze
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 171 77, Stockholm, Sweden.
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia.
| |
Collapse
|
17
|
Massó-Vallés D, Beaulieu ME, Soucek L. MYC, MYCL, and MYCN as therapeutic targets in lung cancer. Expert Opin Ther Targets 2020; 24:101-114. [PMID: 32003251 DOI: 10.1080/14728222.2020.1723548] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Lung cancer is the leading cause of cancer-related mortality globally. Despite recent advances with personalized therapies and immunotherapy, the prognosis remains dire and recurrence is frequent. Myc is an oncogene deregulated in human cancers, including lung cancer, where it supports tumorigenic processes and progression. Elevated Myc levels have also been associated with resistance to therapy.Areas covered: This article summarizes the genomic and transcriptomic studies that compile evidence for (i) MYC, MYCN, and MYCL amplification and overexpression in lung cancer patients, and (ii) their prognostic significance. We collected the most recent literature regarding the development of Myc inhibitors where the emphasis is on those inhibitors tested in lung cancer experimental models and their potential for future clinical application.Expert opinion: The targeting of Myc in lung cancer is potentially an unprecedented opportunity for inhibiting a key player in tumor progression and maintenance and therapeutic resistance. Myc inhibitory strategies are on the path to their clinical application but further work is necessary for the assessment of their use in combination with standard treatment approaches. Given the role of Myc in immune suppression, a significant opportunity may exist in the combination of Myc inhibitors with immunotherapies.
Collapse
Affiliation(s)
| | | | - Laura Soucek
- Peptomyc S.L., Edifici Cellex, Hospital Vall d'Hebron, Barcelona, Spain.,Edifici Cellex, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,Institució Catalana De Recerca I Estudis Avançats (ICREA), Barcelona, Spain.,Department of Biochemistry and Molecular Biology, Universitat Autònoma De Barcelona, Bellaterra, Spain
| |
Collapse
|
18
|
Chava S, Reynolds CP, Pathania AS, Gorantla S, Poluektova LY, Coulter DW, Gupta SC, Pandey MK, Challagundla KB. miR-15a-5p, miR-15b-5p, and miR-16-5p inhibit tumor progression by directly targeting MYCN in neuroblastoma. Mol Oncol 2019; 14:180-196. [PMID: 31637848 PMCID: PMC6944109 DOI: 10.1002/1878-0261.12588] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 09/17/2019] [Accepted: 10/21/2019] [Indexed: 01/15/2023] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid malignancy in children. Despite current aggressive treatment regimens, the prognosis for high-risk NB patients remains poor, with the survival of less than 40%. Amplification/stabilization of MYCN oncogene, in NB is associated with a high risk of recurrence. Thus, there is an urgent need for novel therapeutics. The deregulated expression of microRNA (miR) is reported in NB; nonetheless, its effect on MYCN regulation is poorly understood. First, we identified that miR-15a-5p, miR-15b-5p, and miR-16-5p (hereafter miR-15a, miR-15b or miR-16) were down-regulated in patient-derived xenografts (PDX) with high MYCN expression. MiR targeting sequences on MYCN mRNA were predicted using online databases such as TargetScan and miR database. The R2 database, containing 105 NB patients, showed an inverse correlation between MYCN mRNA and deleted in lymphocytic leukemia (DLEU) 2, a host gene of miR-15. Moreover, overexpression of miR-15a, miR-15b or miR-16 significantly reduced the levels of MYCN mRNA and N-Myc protein. Conversely, inhibiting miR dramatically enhanced MYCN mRNA and N-Myc protein levels, as well as increasing mRNA half-life in NB cells. By performing immunoprecipitation assays of argonaute-2 (Ago2), a core component of the RNA-induced silencing complex, we showed that miR-15a, miR-15b and miR-16 interact with MYCN mRNA. Luciferase reporter assays showed that miR-15a, miR-15b and miR-16 bind with 3'UTR of MYCN mRNA, resulting in MYCN suppression. Moreover, induced expression of miR-15a, miR-15b and miR-16 significantly reduced the proliferation, migration, and invasion of NB cells. Finally, transplanting miR-15a-, miR-15b- and miR-16-expressing NB cells into NSG mice repressed tumor formation and MYCN expression. These data suggest that miR-15a, miR-15b and miR-16 exert a tumor-suppressive function in NB by targeting MYCN. Therefore, these miRs could be considered as potential targets for NB treatment.
Collapse
Affiliation(s)
- Srinivas Chava
- Department of Biochemistry and Molecular Biology & the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - C Patrick Reynolds
- Childhood Cancer Repository, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Anup S Pathania
- Department of Biochemistry and Molecular Biology & the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Don W Coulter
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Uttar Pradesh, India
| | - Manoj K Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Kishore B Challagundla
- Department of Biochemistry and Molecular Biology & the Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
19
|
Sai E, Miwa Y, Takeyama R, Kojima S, Ueno T, Yashiro M, Seto Y, Mano H. Identification of candidates for driver oncogenes in scirrhous-type gastric cancer cell lines. Cancer Sci 2019; 110:2643-2651. [PMID: 31222839 PMCID: PMC6676123 DOI: 10.1111/cas.14111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/17/2019] [Accepted: 06/19/2019] [Indexed: 02/06/2023] Open
Abstract
Scirrhous‐type gastric cancer (SGC) is one of the most intractable cancer subtypes in humans, and its therapeutic targets have been rarely identified to date. Exploration of somatic mutations in the SGC genome with the next‐generation sequencers has been hampered by markedly increased fibrous tissues. Thus, SGC cell lines may be useful resources for searching for novel oncogenes. Here we have conducted whole exome sequencing and RNA sequencing on 2 SGC cell lines, OCUM‐8 and OCUM‐9. Interestingly, most of the mutations thus identified have not been reported. In OCUM‐8 cells, a novel CD44‐IGF1R fusion gene is discovered, the protein product of which ligates the amino‐terminus of CD44 to the transmembrane and tyrosine‐kinase domains of IGF1R. Furthermore, both CD44 and IGF1R are markedly amplified in the OCUM‐8 genome and abundantly expressed. CD44‐IGF1R has a transforming ability, and the suppression of its kinase activity leads to rapid cell death of OCUM‐8. To the best of our knowledge, this is the first report describing the transforming activity of IGF1R fusion genes. However, OCUM‐9 seems to possess multiple oncogenic events in its genome. In particular, a novel BORCS5‐ETV6 fusion gene is identified in the OCUM‐9 genome. BORCS5‐ETV6 possesses oncogenic activity, and suppression of its message partially inhibits cell growth. Prevalence of these novel fusion genes among SGC awaits further investigation, but we validate the significance of cell lines as appropriate reagents for detailed genomic analyses of SGC.
Collapse
Affiliation(s)
- Eirin Sai
- Department of Medical Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshiyuki Miwa
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Reina Takeyama
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Shinya Kojima
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Toshihide Ueno
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Mano
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Cellular Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| |
Collapse
|
20
|
Paglia S, Sollazzo M, Di Giacomo S, Strocchi S, Grifoni D. Exploring MYC relevance to cancer biology from the perspective of cell competition. Semin Cancer Biol 2019; 63:49-59. [PMID: 31102666 DOI: 10.1016/j.semcancer.2019.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/08/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022]
Abstract
Cancer has long been regarded and treated as a foreign body appearing by mistake inside a living organism. However, now we know that cancer cells communicate with neighbours, thereby creating modified environments able to support their unusual need for nutrients and space. Understanding the molecular basis of these bi-directional interactions is thus mandatory to approach the complex nature of cancer. Since their discovery, MYC proteins have been showing to regulate a steadily increasing number of processes impacting cell fitness, and are consistently found upregulated in almost all human tumours. Of interest, MYC takes part in cell competition, an evolutionarily conserved fitness comparison strategy aimed at detecting weakened cells, which are then committed to death, removed from the tissue and replaced by fitter neighbours. During physiological development, MYC-mediated cell competition is engaged to eliminate cells with suboptimal MYC levels, so as to guarantee selective growth of the fittest and proper homeostasis, while transformed cells expressing high levels of MYC coopt cell competition to subvert tissue constraints, ultimately disrupting homeostasis. Therefore, the interplay between cells with different MYC levels may result in opposite functional outcomes, depending on the nature of the players. In the present review, we describe the most recent findings on the role of MYC-mediated cell competition in different contexts, with a special emphasis on its impact on cancer initiation and progression. We also discuss the relevance of competition-associated cell death to cancer disease.
Collapse
Affiliation(s)
- Simona Paglia
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| | - Manuela Sollazzo
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| | - Simone Di Giacomo
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| | - Silvia Strocchi
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| | - Daniela Grifoni
- CanceЯEvolutionLab, University of Bologna, Department of Pharmacy and Biotechnology, Via Selmi 3, 40126, Bologna, Italy.
| |
Collapse
|
21
|
Yang TW, Sahu D, Chang YW, Hsu CL, Hsieh CH, Huang HC, Juan HF. RNA-Binding Proteomics Reveals MATR3 Interacting with lncRNA SNHG1 To Enhance Neuroblastoma Progression. J Proteome Res 2018; 18:406-416. [PMID: 30516047 DOI: 10.1021/acs.jproteome.8b00693] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The interaction of long noncoding RNAs (lncRNAs) with one or more RNA-binding proteins (RBPs) is important to a plethora of cellular and physiological processes. The lncRNA SNHG1 was reported to be aberrantly expressed and associated with poor patient prognosis in several cancers including neuroblastoma. However, the interacting RBPs and biological functions associated with SNHG1 in neuroblastoma remain unknown. In this study, we identified 283, 31, and 164 SNHG1-interacting proteins in SK-N-BE(2)C, SK-N-DZ, and SK-N-AS neuroblastoma cells, respectively, using a RNA-protein pull-down assay coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS). Twenty-four SNHG1-interacting RBPs were identified in common from these three neuroblastoma cell lines. RBPs MATR3, YBX1, and HNRNPL have the binding sites for SNHG1 predicted by DeepBind motif analysis. Furthermore, the direct binding of MATR3 with SNHG1 was validated by Western blot and confirmed by RNA immunoprecipitation assay (RIP). Coexpression analysis revealed that the expression of SNHG1 is positively correlated with MATR3 ( P = 3.402 × 10-13). The high expression of MATR3 is associated with poor event-free survival ( P = 0.00711) and overall survival ( P = 0.00064). Biological functions such as ribonucleoprotein complex biogenesis, RNA processing, and RNA splicing are significantly enriched and in common between SNHG1 and MATR3. In conclusion, we identified MATR3 as binding to SNHG1 and the interaction might be involved in splicing events that enhance neuroblastoma progression.
Collapse
Affiliation(s)
| | - Divya Sahu
- Institute of Biomedical Informatics , National Yang-Ming University , Taipei 112 , Taiwan
| | | | - Chia-Lang Hsu
- Department of Medical Research , National Taiwan University Hospital , Taipei 100 , Taiwan
| | | | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics , National Yang-Ming University , Taipei 112 , Taiwan
| | | |
Collapse
|
22
|
Felgenhauer J, Tomino L, Selich-Anderson J, Bopp E, Shah N. Dual BRD4 and AURKA Inhibition Is Synergistic against MYCN-Amplified and Nonamplified Neuroblastoma. Neoplasia 2018; 20:965-974. [PMID: 30153557 PMCID: PMC6111011 DOI: 10.1016/j.neo.2018.08.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/02/2018] [Accepted: 08/02/2018] [Indexed: 11/29/2022] Open
Abstract
A majority of cases of high-risk neuroblastoma, an embryonal childhood cancer, are driven by MYC or MYCN-driven oncogenic signaling. While considered to be directly “undruggable” therapeutically, MYC and MYCN can be repressed transcriptionally by inhibition of Bromodomain-containing protein 4 (BRD4) or destabilized posttranslationally by inhibition of Aurora Kinase A (AURKA). Preclinical and early-phase clinical studies of BRD4 and AURKA inhibitors, however, show limited efficacy against neuroblastoma when used alone. We report our studies on the concomitant use of the BRD4 inhibitor I-BET151 and AURKA inhibitor alisertib. We show that, in vitro, the drugs act synergistically to inhibit viability in four models of high-risk neuroblastoma. We demonstrate that this synergy is driven, in part, by the ability of I-BET151 to mitigate reflexive upregulation of AURKA, MYC, and MYCN in response to AURKA inhibition. We then demonstrate that I-BET151 and alisertib are effective in prolonging survival in four xenograft neuroblastoma models in vivo, and this efficacy is augmented by the addition of the antitubule chemotherapeutic vincristine. These data suggest that epigenetic and posttranslational inhibition of MYC/MYCN-driven pathways may have significant clinical efficacy against neuroblastoma.
Collapse
Affiliation(s)
- Joshua Felgenhauer
- Nationwide Children's Hospital, Center for Childhood Cancer and Blood Disorders, 700 Children's Drive, Columbus, OH, 43205
| | - Laura Tomino
- Nationwide Children's Hospital, Center for Childhood Cancer and Blood Disorders, 700 Children's Drive, Columbus, OH, 43205
| | - Julia Selich-Anderson
- Nationwide Children's Hospital, Center for Childhood Cancer and Blood Disorders, 700 Children's Drive, Columbus, OH, 43205
| | - Emily Bopp
- The Ohio State University, College of Arts and Sciences, 186 University Hall, Columbus, OH, 43210
| | - Nilay Shah
- Nationwide Children's Hospital, Center for Childhood Cancer and Blood Disorders, 700 Children's Drive, Columbus, OH, 43205; The Ohio State University, College of Medicine, Department of Pediatrics, 370 W 9th Ave, Columbus, OH 43210.
| |
Collapse
|
23
|
Enhanced expression of MycN/CIP2A drives neural crest toward a neural stem cell-like fate: Implications for priming of neuroblastoma. Proc Natl Acad Sci U S A 2018; 115:E7351-E7360. [PMID: 30021854 DOI: 10.1073/pnas.1800039115] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma is a neural crest-derived childhood tumor of the peripheral nervous system in which MycN amplification is a hallmark of poor prognosis. Here we show that MycN is expressed together with phosphorylation-stabilizing factor CIP2A in regions of the neural plate destined to form the CNS, but MycN is excluded from the neighboring neural crest stem cell domain. Interestingly, ectopic expression of MycN or CIP2A in the neural crest domain biases cells toward CNS-like neural stem cells that express Sox2. Consistent with this, some forms of neuroblastoma have been shown to share transcriptional resemblance with CNS neural stem cells. As high MycN/CIP2A levels correlate with poor prognosis, we posit that a MycN/CIP2A-mediated cell-fate bias may reflect a possible mechanism underlying early priming of some aggressive forms of neuroblastoma. In contrast to MycN, its paralogue cMyc is normally expressed in the neural crest stem cell domain and typically is associated with better overall survival in clinical neuroblastoma, perhaps reflecting a more "normal" neural crest-like state. These data suggest that priming for some forms of aggressive neuroblastoma may occur before neural crest emigration from the CNS and well before sympathoadrenal specification.
Collapse
|
24
|
Abstract
Neuroblastoma (NB) is the most common solid childhood tumor outside the brain and causes 15% of childhood cancer-related mortality. The main drivers of NB formation are neural crest cell-derived sympathoadrenal cells that undergo abnormal genetic arrangements. Moreover, NB is a complex disease that has high heterogeneity and is therefore difficult to target for successful therapy. Thus, a better understanding of NB development helps to improve treatment and increase the survival rate. One of the major causes of sporadic NB is known to be MYCN amplification and mutations in ALK (anaplastic lymphoma kinase) are responsible for familial NB. Many other genetic abnormalities can be found; however, they are not considered as driver mutations, rather they support tumor aggressiveness. Tumor cell elimination via cell death is widely accepted as a successful technique. Therefore, in this review, we provide a thorough overview of how different modes of cell death and treatment strategies, such as immunotherapy or spontaneous regression, are or can be applied for NB elimination. In addition, several currently used and innovative approaches and their suitability for clinical testing and usage will be discussed. Moreover, significant attention will be given to combined therapies that show more effective results with fewer side effects than drugs targeting only one specific protein or pathway.
Collapse
|
25
|
Rickman DS, Schulte JH, Eilers M. The Expanding World of N-MYC–Driven Tumors. Cancer Discov 2018; 8:150-163. [DOI: 10.1158/2159-8290.cd-17-0273] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/04/2017] [Accepted: 10/18/2017] [Indexed: 11/16/2022]
|
26
|
Bo L, Wei B, Wang Z, Kong D, Gao Z, Miao Z. Bioinformatics analysis of the CDK2 functions in neuroblastoma. Mol Med Rep 2018; 17:3951-3959. [PMID: 29328425 DOI: 10.3892/mmr.2017.8368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 11/14/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to elucidate the potential mechanism of cyclin-dependent kinase 2 (CDK2) in neuroblastoma progression and to identify the candidate genes associated with neuroblastoma with CDK2 silencing. The microarray data of GSE16480 were obtained from the gene expression omnibus database. This dataset contained 15 samples: Neuroblastoma cell line IMR32 transfected with CDK2 shRNA at 0, 8, 24, 48 and 72 h (n=3 per group; total=15). Significant clusters associated with differentially expressed genes (DEGs) were identified using fuzzy C‑Means algorithm in the Mfuzz package. Gene ontology and pathway enrichment analysis of DEGs in each cluster were performed, and a protein‑protein interaction (PPI) network was constructed. Additionally, functional annotation of DEGs in clusters was performed for the detection of transcription factors and tumor‑associated genes. A total of 4 clusters with significant change tendency and 1,683 DEGs were identified. The hub nodes of the PPI network constructed by DEGs in Cluster 1, Cluster 2, Cluster 3 and Cluster 4 were MDM2 oncogene, E3 ubiquitin protein ligase (MDM2), cyclin‑dependent kinase 1 (CDK1), proteasome (prosome, macropain) 26S subunit, non‑ATPase, 14 (PSMD14) and translocator protein (18 kDa) (TSPO) respectively. These genes were significantly enriched in the p53 signaling pathway, cell cycle, proteasome and systemic lupus erythematosus pathways. MDM2, CDK1, PSMD14 and TSPO may be key target genes of CDK2. CDK2 may have key functions in neuroblastoma progression by regulating the expression of these genes.
Collapse
Affiliation(s)
- Lijuan Bo
- Department of Infections, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Bo Wei
- Department of Neurosurgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhanfeng Wang
- Department of Neurosurgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Daliang Kong
- Department of Orthopaedics, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zheng Gao
- Department of Neurosurgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhuang Miao
- Department of Neurosurgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
27
|
Heimburg T, Kolbinger FR, Zeyen P, Ghazy E, Herp D, Schmidtkunz K, Melesina J, Shaik TB, Erdmann F, Schmidt M, Romier C, Robaa D, Witt O, Oehme I, Jung M, Sippl W. Structure-Based Design and Biological Characterization of Selective Histone Deacetylase 8 (HDAC8) Inhibitors with Anti-Neuroblastoma Activity. J Med Chem 2017; 60:10188-10204. [DOI: 10.1021/acs.jmedchem.7b01447] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Tino Heimburg
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Fiona R. Kolbinger
- Clinical
Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
- Preclinical Program, Hopp Children’s Cancer Center at NCT Heidelberg (KiTZ), 69120 Heidelberg, Germany
| | - Patrik Zeyen
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Ehab Ghazy
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Daniel Herp
- Institute
of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany
| | - Karin Schmidtkunz
- Institute
of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany
| | - Jelena Melesina
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Tajith Baba Shaik
- Département
de Biologie Structurale Intégrative, Institut de Génétique
et Biologie Moléculaire et Cellulaire (IGBMC), Université
de Strasbourg (UDS), CNRS, INSERM, 67404 Illkirch Cedex, France
| | - Frank Erdmann
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Matthias Schmidt
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Christophe Romier
- Département
de Biologie Structurale Intégrative, Institut de Génétique
et Biologie Moléculaire et Cellulaire (IGBMC), Université
de Strasbourg (UDS), CNRS, INSERM, 67404 Illkirch Cedex, France
| | - Dina Robaa
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| | - Olaf Witt
- Clinical
Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
- Preclinical Program, Hopp Children’s Cancer Center at NCT Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Department
of Pediatric Oncology, Hematology and Immunology, University of Heidelberg Medical Center, 69120 Heidelberg, Germany
| | - Ina Oehme
- Clinical
Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), INF 280, 69120 Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
- Preclinical Program, Hopp Children’s Cancer Center at NCT Heidelberg (KiTZ), 69120 Heidelberg, Germany
| | - Manfred Jung
- Institute
of Pharmaceutical Sciences, University of Freiburg, 79104 Freiburg, Germany
| | - Wolfgang Sippl
- Institute
of Pharmacy, Martin-Luther University of Halle-Wittenberg, 06120 Halle/Saale, Germany
| |
Collapse
|
28
|
Riccardo F, Réal A, Voena C, Chiarle R, Cavallo F, Barutello G. Maternal Immunization: New Perspectives on Its Application Against Non-Infectious Related Diseases in Newborns. Vaccines (Basel) 2017; 5:E20. [PMID: 28763018 PMCID: PMC5620551 DOI: 10.3390/vaccines5030020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022] Open
Abstract
The continuous evolution in preventive medicine has anointed vaccination a versatile, human-health improving tool, which has led to a steady decline in deaths in the developing world. Maternal immunization represents an incisive step forward for the field of vaccination as it provides protection against various life-threatening diseases in pregnant women and their children. A number of studies to improve prevention rates and expand protection against the largest possible number of infections are still in progress. The complex unicity of the mother-infant interaction, both during and after pregnancy and which involves immune system cells and molecules, is an able partner in the success of maternal immunization, as intended thus far. Interestingly, new studies have shed light on the versatility of maternal immunization in protecting infants from non-infectious related diseases, such as allergy, asthma and congenital metabolic disorders. However, barely any attempt at applying maternal immunization to the prevention of childhood cancer has been made. The most promising study reported in this new field is a recent proof of concept on the efficacy of maternal immunization in protecting cancer-prone offspring against mammary tumor progression. New investigations into the possibility of exploiting maternal immunization to prevent the onset and/or progression of neuroblastoma, one of the most common childhood malignancies, are therefore justified. Maternal immunization is presented in a new guise in this review. Attention will be focused on its versatility and potential applications in preventing tumor progression in neuroblastoma-prone offspring.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Aline Réal
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, Center for Experimental Research and Medical Studies, University of Torino, Torino 10126, Italy.
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, Center for Experimental Research and Medical Studies, University of Torino, Torino 10126, Italy.
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA.
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| |
Collapse
|
29
|
Megiorni F, Colaiacovo M, Cialfi S, McDowell HP, Guffanti A, Camero S, Felsani A, Losty PD, Pizer B, Shukla R, Cappelli C, Ferrara E, Pizzuti A, Moles A, Dominici C. A sketch of known and novel MYCN-associated miRNA networks in neuroblastoma. Oncol Rep 2017; 38:3-20. [PMID: 28586032 PMCID: PMC5492854 DOI: 10.3892/or.2017.5701] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 04/27/2017] [Indexed: 12/12/2022] Open
Abstract
Neuroblastoma (NB) originates from neural crest-derived precursors and represents the most common childhood extracranial solid tumour. MicroRNAs (miRNAs), a class of small non-coding RNAs that participate in a wide variety of biological processes by regulating gene expression, appear to play an essential role within the NB context. High-throughput next generation sequencing (NGS) was applied to study the miRNA transcriptome in a cohort of NB tumours with and without MYCN-amplification (MNA and MNnA, respectively) and in dorsal root ganglia (DRG), as a control. Out of the 128 miRNAs differentially expressed in the NB vs. DRG comparison, 47 were expressed at higher levels, while 81 were expressed at lower levels in the NB tumours. We also found that 23 miRNAs were differentially expressed in NB with or without MYCN-amplification, with 17 miRNAs being upregulated and 6 being downregulated in the MNA subtypes. Functional annotation analysis of the target genes of these differentially expressed miRNAs demonstrated that many mRNAs were involved in cancer-related pathways, such as DNA-repair and apoptosis as well as FGFR and EGFR signalling. In particular, we found that miR-628-3p negatively affects MYCN gene expression. Furthermore, we identified a novel miRNA candidate with variable expression in MNA vs. MNnA tumours, whose putative target genes are implicated in the mTOR pathway. The present study provides further insight into the molecular mechanisms that correlate miRNA dysregulation to NB development and progression.
Collapse
Affiliation(s)
- Francesca Megiorni
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, I-00161 Rome, Italy
| | | | - Samantha Cialfi
- Department of Molecular Medicine, Sapienza University of Rome, I-00161 Rome, Italy
| | - Heather P McDowell
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, I-00161 Rome, Italy
| | | | - Simona Camero
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, I-00161 Rome, Italy
| | | | - Paul D Losty
- Department of Paediatric Surgery, Alder Hey Children's NHS Foundation Trust, L12 2AP Liverpool, UK
| | - Barry Pizer
- Department of Oncology, Alder Hey Children's NHS Foundation Trust, L12 2AP Liverpool, UK
| | - Rajeev Shukla
- Department of Perinatal and Paediatric Pathology, Alder Hey Children's NHS Foundation Trust, L12 2AP Liverpool, UK
| | - Carlo Cappelli
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, I-00161 Rome, Italy
| | - Eva Ferrara
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, I-00161 Rome, Italy
| | - Antonio Pizzuti
- Department of Experimental Medicine, Sapienza University of Rome, I-00161 Rome, Italy
| | - Anna Moles
- Genomnia s.r.l., I-20091 Bresso, MI, Italy
| | - Carlo Dominici
- Department of Paediatrics and Infantile Neuropsychiatry, Sapienza University of Rome, I-00161 Rome, Italy
| |
Collapse
|
30
|
MYCN amplified neuroblastoma requires the mRNA translation regulator eEF2 kinase to adapt to nutrient deprivation. Cell Death Differ 2017; 24:1564-1576. [PMID: 28574509 DOI: 10.1038/cdd.2017.79] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 04/08/2017] [Accepted: 04/13/2017] [Indexed: 01/01/2023] Open
Abstract
MYC family proteins are implicated in many human cancers, but their therapeutic targeting has proven challenging. MYCN amplification in childhood neuroblastoma (NB) is associated with aggressive disease and high mortality. Novel and effective therapeutic strategies are therefore urgently needed for these tumors. MYC-driven oncogenic transformation impairs cell survival under nutrient deprivation (ND), a characteristic stress condition within the tumor microenvironment. We recently identified eukaryotic Elongation Factor 2 Kinase (eEF2K) as a pivotal mediator of the adaptive response of tumor cells to ND. We therefore hypothesized that eEF2K facilitates the adaptation of MYCN amplified NB to ND, and that inhibiting this pathway can impair MYCN-driven NB progression. To test our hypothesis, we first analyzed publicly available genomic databases and tissue microarrays for eEF2K expression in NB, and for links between eEF2K, MYCN, and clinical outcome in NB. Effects of eEF2K inhibition were evaluated on survival of MYCN amplified versus non-amplified NB cell lines under ND. Finally, NB xenograft mouse models were used to confirm in vitro observations. Our results indicate that high eEF2K expression and activity are strongly predictive of poor outcome in NB, and correlates significantly with MYCN amplification. Inhibition of eEF2K markedly decreases survival of MYCN amplified NB cell lines in vitro under ND. Growth of MYCN amplified NB xenografts is markedly impaired by eEF2K knockdown, particularly under caloric restriction. In summary, eEF2K protects MYCN overexpressing NB cells from ND in vitro and in vivo, highlighting this kinase as a critical mediator of the adaptive response of MYCN amplified NB cells to metabolic stress.
Collapse
|
31
|
The MYCN Protein in Health and Disease. Genes (Basel) 2017; 8:genes8040113. [PMID: 28358317 PMCID: PMC5406860 DOI: 10.3390/genes8040113] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022] Open
Abstract
MYCN is a member of the MYC family of proto-oncogenes. It encodes a transcription factor, MYCN, involved in the control of fundamental processes during embryonal development. The MYCN protein is situated downstream of several signaling pathways promoting cell growth, proliferation and metabolism of progenitor cells in different developing organs and tissues. Conversely, deregulated MYCN signaling supports the development of several different tumors, mainly with a childhood onset, including neuroblastoma, medulloblastoma, rhabdomyosarcoma and Wilms’ tumor, but it is also associated with some cancers occurring during adulthood such as prostate and lung cancer. In neuroblastoma, MYCN-amplification is the most consistent genetic aberration associated with poor prognosis and treatment failure. Targeting MYCN has been proposed as a therapeutic strategy for the treatment of these tumors and great efforts have allowed the development of direct and indirect MYCN inhibitors with potential clinical use.
Collapse
|
32
|
Vaughan L, Clarke PA, Barker K, Chanthery Y, Gustafson CW, Tucker E, Renshaw J, Raynaud F, Li X, Burke R, Jamin Y, Robinson SP, Pearson A, Maira M, Weiss WA, Workman P, Chesler L. Inhibition of mTOR-kinase destabilizes MYCN and is a potential therapy for MYCN-dependent tumors. Oncotarget 2016; 7:57525-57544. [PMID: 27438153 PMCID: PMC5295370 DOI: 10.18632/oncotarget.10544] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023] Open
Abstract
MYC oncoproteins deliver a potent oncogenic stimulus in several human cancers, making them major targets for drug development, but efforts to deliver clinically practical therapeutics have not yet been realized. In childhood cancer, aberrant expression of MYC and MYCN genes delineates a group of aggressive tumours responsible for a major proportion of pediatric cancer deaths. We designed a chemical-genetic screen that identifies compounds capable of enhancing proteasomal elimination of MYCN oncoprotein. We isolated several classes of compound that selectively kill MYCN expressing cells and we focus on inhibitors of PI3K/mTOR pathway in this study. We show that PI3K/mTOR inhibitors selectively killed MYCN-expressing neuroblastoma tumor cells, and induced significant apoptosis of transgenic MYCN-driven neuroblastoma tumors concomitant with elimination of MYCN protein in vivo. Mechanistically, the ability of these compounds to degrade MYCN requires complete blockade of mTOR but not PI3 kinase activity and we highlight NVP-BEZ235 as a PI3K/mTOR inhibitor with an ideal activity profile. These data establish that MYCN expression is a marker indicative of likely clinical sensitivity to mTOR inhibition, and provide a rationale for the selection of clinical candidate MYCN-destabilizers likely to be useful for the treatment of MYCN-driven cancers.
Collapse
Affiliation(s)
- Lynsey Vaughan
- Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey, UK
- Present address: Cell Signalling Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Paul A. Clarke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Signal Transduction and Molecular Pharmacology Team, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Karen Barker
- Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Yvan Chanthery
- Department of Neurology, Pediatrics, Neurosurgery, Brain Tumor Research Center and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Clay W. Gustafson
- Department of Neurology, Pediatrics, Neurosurgery, Brain Tumor Research Center and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Elizabeth Tucker
- Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Jane Renshaw
- Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Florence Raynaud
- Cancer Research UK Cancer Therapeutics Unit, Clinical Pharmacology and Trials Team, Sutton, Surrey, UK
| | - Xiaodun Li
- Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey, UK
- Present address: MRC Cancer Unit, University of Cambridge, Cambridge, UK
| | - Rosemary Burke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Target Selection and Hit Discovery Team, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Yann Jamin
- Cancer Research UK & Engineering and Physical Sciences Research Council Cancer Imaging Centre, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Simon P. Robinson
- Cancer Research UK & Engineering and Physical Sciences Research Council Cancer Imaging Centre, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Andrew Pearson
- Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Michel Maira
- Novartis Pharma AG, Basel, Switzerland
- Present address: Basilea Pharmaceutica International AG, Basel, Switzerland
| | - William A. Weiss
- Department of Neurology, Pediatrics, Neurosurgery, Brain Tumor Research Center and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Signal Transduction and Molecular Pharmacology Team, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Louis Chesler
- Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey, UK
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Signal Transduction and Molecular Pharmacology Team, The Institute of Cancer Research, Sutton, Surrey, UK
- The Royal Marsden NHS Trust, Children and Young People's Unit, Sutton, Surrey, UK
| |
Collapse
|
33
|
Marrano P, Irwin MS, Thorner PS. Heterogeneity of MYCN amplification in neuroblastoma at diagnosis, treatment, relapse, and metastasis. Genes Chromosomes Cancer 2016; 56:28-41. [PMID: 27465929 DOI: 10.1002/gcc.22398] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 12/14/2022] Open
Abstract
Amplification of the MYCN gene in neuroblastoma is associated with a poor prognosis and is considered to remain unchanged in post-treatment specimens and metastases. While heterogeneity of MYCN copy number in tumor cells has been reported, serial samples have only been studied in a limited way, and the biologic relevance of this finding is not well understood. We used in situ hybridization on paraffin sections of 102 specimens from 30 patients with MYCN-amplified neuroblastoma to determine MYCN copy number in the primary tumor, pre- and post-treatment, and in metastatic samples. Nineteen cases (63%) showed diffuse MYCN amplification in all samples tested. Nine cases (30%) showed a reduction in MYCN copy number: five cases with diffuse amplification subsequently showed focal amplification, one case with diffuse MYCN amplification showed MYCN gain after treatment, and three focally amplified cases were non-amplified in later specimens. In two cases (7%), focal amplification became diffuse in subsequent samples. Histology was not predictive of the temporal or spatial pattern of MYCN amplification for a particular tumor. If extent of amplification (focal vs. diffuse) is not considered, 26/30 (87%) of cases were consistently MYCN-amplified. However, our data suggest that MYCN status can be heterogeneous between tumor sites, during tumor progression or following treatment, challenging the notion that MYCN copy number does not change for a particular neuroblastoma. Assessing the biologic significance of MYCN heterogeneity will require larger studies of clinically annotated tumor samples, and will depend on interpreting heterogeneity in MYCN status in combination with other genetic changes. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Paula Marrano
- Department of Pediatric Laboratory Medicine, the Hospital for Sick Children, Toronto, Canada
| | - Meredith S Irwin
- Department of Pediatrics, the Hospital for Sick Children, Toronto, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Paul S Thorner
- Department of Pediatric Laboratory Medicine, the Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
34
|
Schwab M, Corvi R, Amler LC. N-MYC Oncogene Amplification: A Consequence of Genomic Instability in Human Neuroblastoma. Neuroscientist 2016. [DOI: 10.1177/107385849500100505] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Increase of the dosage of cellular oncogenes by DNA amplification is a frequent genetic alteration of cancer cells and arises as the consequence of genomic instability. The presence of amplified cellular oncogenes is usually signaled by conspicuous chromosomal abnormalities "double minutes," or "homogeneously staining chromosomal regions." Some human cancers carry a specific amplified oncogene at high incidence. In neuroblastomas, which are tumors of the peripheral nervous system that arise from primitive neuroectodermal cells derived from neural crest, the amplification of the gene N-MYC has been associated with aggressively growing cancers and is an indicator for poor prognosis. N-MYC amplification is of predictive value for iden tifying neuroblastoma patients who either require specific therapeutic regimens or who do not benefit from chemotherapy. The Neuroscientist 1:277-285, 1995
Collapse
Affiliation(s)
- Manfred Schwab
- German Cancer Research Center Division of Cytogenetics
Heidelberg, Germany
| | - Raffaella Corvi
- German Cancer Research Center Division of Cytogenetics
Heidelberg, Germany
| | - Lukas C. Amler
- German Cancer Research Center Division of Cytogenetics
Heidelberg, Germany
| |
Collapse
|
35
|
Nicolai S, Pieraccioli M, Peschiaroli A, Melino G, Raschellà G. Neuroblastoma: oncogenic mechanisms and therapeutic exploitation of necroptosis. Cell Death Dis 2015; 6:e2010. [PMID: 26633716 PMCID: PMC4720889 DOI: 10.1038/cddis.2015.354] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/17/2015] [Accepted: 10/19/2015] [Indexed: 12/20/2022]
Abstract
Neuroblastoma (NB) is the most common extracranial childhood tumor classified in five stages (1, 2, 3, 4 and 4S), two of which (3 and 4) identify chemotherapy-resistant, highly aggressive disease. High-risk NB frequently displays MYCN amplification, mutations in ALK and ATRX, and genomic rearrangements in TERT genes. These NB subtypes are also characterized by reduced susceptibility to programmed cell death induced by chemotherapeutic drugs. The latter feature is a major cause of failure in the treatment of advanced NB patients. Thus, proper reactivation of apoptosis or of other types of programmed cell death pathways in response to treatment is relevant for the clinical management of aggressive forms of NB. In this short review, we will discuss the most relevant genomic rearrangements that define high-risk NB and the role that destabilization of p53 and p73 can have in NB aggressiveness. In addition, we will propose a strategy to stabilize p53 and p73 by using specific inhibitors of their ubiquitin-dependent degradation. Finally, we will introduce necroptosis as an alternative strategy to kill NB cells and increase tumor immunogenicity.
Collapse
Affiliation(s)
- S Nicolai
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - M Pieraccioli
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - A Peschiaroli
- Institute of Cell Biology and Neurobiology (IBCN), CNR, Via E. Ramarini 32, Rome 00015, Italy
| | - G Melino
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy.,Medical Research Council, Toxicology Unit, Hodgkin Building, Leicester University, Lancaster Road, PO Box 138, Leicester LE1 9HN, UK
| | - G Raschellà
- ENEA Research Center Casaccia, Laboratory of Biosafety and Risk Assessment, Via Anguillarese, 301, Rome 00123, Italy
| |
Collapse
|
36
|
Zhang Z, Faouzi M, Huang J, Geerts D, Yu H, Fleig A, Penner R. N-Myc-induced up-regulation of TRPM6/TRPM7 channels promotes neuroblastoma cell proliferation. Oncotarget 2015; 5:7625-34. [PMID: 25277194 PMCID: PMC4202149 DOI: 10.18632/oncotarget.2283] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Intracellular levels of the divalent cations Ca2+ and Mg2+ are important regulators of cell cycle and proliferation. However, the precise mechanisms by which they are regulated in cancer remain incompletely understood. The channel kinases TRPM6 and TRPM7 are gatekeepers of human Ca2+/Mg2+ metabolism. Here, we investigated the human neuroblastoma cell line SHEP-21N in which the MYCN oncogene (encoding N-Myc) can be reversibly expressed under control of an inducible repressor. We report that N-Myc expression increases cell growth and up-regulates both TRPM6 and TRPM7 expression. Membrane current analyses reveal that endogenous TRPM6/TRPM7 currents exhibit reduced Mg·ATP suppression, increased Mg2+ sensitivity, and diminished sensitivity to 2-APB inhibition. These properties are consistent with N-Myc-induced increase of heteromeric TRPM7/TRPM6 channels promoting Ca2+ and Mg2+ uptake. Genetic suppression of TRPM6/TRPM7 through siRNA inhibits cell proliferation, suggesting that N-Myc can promote neuroblastoma cell proliferation through up-regulation of divalent cation-transporting channels.
Collapse
Affiliation(s)
- Zheng Zhang
- Center for Biomedical Research, The Queen's Medical Center, University of Hawaii Cancer Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, U.S.A. Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China. Contributed equally to this work
| | - Malika Faouzi
- Center for Biomedical Research, The Queen's Medical Center, University of Hawaii Cancer Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, U.S.A.. Contributed equally to this work
| | - Junhao Huang
- Center for Biomedical Research, The Queen's Medical Center, University of Hawaii Cancer Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, U.S.A
| | - Dirk Geerts
- Department of Pediatric Oncology/Hematology, Erasmus University Medical Center, Dr. Molewaterplein 50, GE Rotterdam, the Netherlands
| | - Haijie Yu
- Center for Biomedical Research, The Queen's Medical Center, University of Hawaii Cancer Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, U.S.A
| | - Andrea Fleig
- Center for Biomedical Research, The Queen's Medical Center,University of Hawaii Cancer Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, U.S.A
| | - Reinhold Penner
- Center for Biomedical Research, The Queen's Medical Center, University of Hawaii Cancer Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, U.S.A
| |
Collapse
|
37
|
Expression quantitative trait loci and receptor pharmacology implicate Arg1 and the GABA-A receptor as therapeutic targets in neuroblastoma. Cell Rep 2014; 9:1034-46. [PMID: 25437558 DOI: 10.1016/j.celrep.2014.09.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 08/14/2014] [Accepted: 09/25/2014] [Indexed: 02/05/2023] Open
Abstract
The development of targeted therapeutics for neuroblastoma, the third most common tumor in children, has been limited by a poor understanding of growth signaling mechanisms unique to the peripheral nerve precursors from which tumors arise. In this study, we combined genetics with gene-expression analysis in the peripheral sympathetic nervous system to implicate arginase 1 and GABA signaling in tumor formation in vivo. In human neuroblastoma cells, either blockade of ARG1 or benzodiazepine-mediated activation of GABA-A receptors induced apoptosis and inhibited mitogenic signaling through AKT and MAPK. These results suggest that ARG1 and GABA influence both neural development and neuroblastoma and that benzodiazepines in clinical use may have potential applications for neuroblastoma therapy.
Collapse
|
38
|
Brodeur GM, Iyer R, Croucher JL, Zhuang T, Higashi M, Kolla V. Therapeutic targets for neuroblastomas. Expert Opin Ther Targets 2014; 18:277-92. [PMID: 24387342 DOI: 10.1517/14728222.2014.867946] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Neuroblastoma (NB) is the most common and deadly solid tumor in children. Despite recent improvements, the long-term outlook for high-risk NB is still < 50%. Further, there is considerable short- and long-term toxicity. More effective, less toxic therapy is needed, and the development of targeted therapies offers great promise. AREAS COVERED Relevant literature was reviewed to identify current and future therapeutic targets that are critical to malignant transformation and progression of NB. The potential or actual NB therapeutic targets are classified into four categories: i) genes activated by amplification, mutation, translocation or autocrine overexpression; ii) genes inactivated by deletion, mutation or epigenetic silencing; iii) membrane-associated genes expressed on most NBs but few other tissues; or iv) common target genes relevant to NB as well as other tumors. EXPERT OPINION Therapeutic approaches have been developed to some of these targets, but many remain untargeted at the present time. It is unlikely that single targeted agents will be sufficient for long-term cure, at least for high-risk NBs. The challenge will be how to integrate targeted agents with each other and with conventional therapy to enhance their efficacy, while simultaneously reducing systemic toxicity.
Collapse
Affiliation(s)
- Garrett M Brodeur
- Children's Hospital of Philadelphia, Division of Oncology , CTRB Rm. 3018, 3501 Civic Center Blvd., Philadelphia, PA 19104-4302 , USA +1 215 590 2817 ; +1 215 590 3770 ;
| | | | | | | | | | | |
Collapse
|
39
|
Suenaga Y, Islam SMR, Alagu J, Kaneko Y, Kato M, Tanaka Y, Kawana H, Hossain S, Matsumoto D, Yamamoto M, Shoji W, Itami M, Shibata T, Nakamura Y, Ohira M, Haraguchi S, Takatori A, Nakagawara A. NCYM, a Cis-antisense gene of MYCN, encodes a de novo evolved protein that inhibits GSK3β resulting in the stabilization of MYCN in human neuroblastomas. PLoS Genet 2014; 10:e1003996. [PMID: 24391509 PMCID: PMC3879166 DOI: 10.1371/journal.pgen.1003996] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 10/18/2013] [Indexed: 11/19/2022] Open
Abstract
The rearrangement of pre-existing genes has long been thought of as the major mode of new gene generation. Recently, de novo gene birth from non-genic DNA was found to be an alternative mechanism to generate novel protein-coding genes. However, its functional role in human disease remains largely unknown. Here we show that NCYM, a cis-antisense gene of the MYCN oncogene, initially thought to be a large non-coding RNA, encodes a de novo evolved protein regulating the pathogenesis of human cancers, particularly neuroblastoma. The NCYM gene is evolutionally conserved only in the taxonomic group containing humans and chimpanzees. In primary human neuroblastomas, NCYM is 100% co-amplified and co-expressed with MYCN, and NCYM mRNA expression is associated with poor clinical outcome. MYCN directly transactivates both NCYM and MYCN mRNA, whereas NCYM stabilizes MYCN protein by inhibiting the activity of GSK3β, a kinase that promotes MYCN degradation. In contrast to MYCN transgenic mice, neuroblastomas in MYCN/NCYM double transgenic mice were frequently accompanied by distant metastases, behavior reminiscent of human neuroblastomas with MYCN amplification. The NCYM protein also interacts with GSK3β, thereby stabilizing the MYCN protein in the tumors of the MYCN/NCYM double transgenic mice. Thus, these results suggest that GSK3β inhibition by NCYM stabilizes the MYCN protein both in vitro and in vivo. Furthermore, the survival of MYCN transgenic mice bearing neuroblastoma was improved by treatment with NVP-BEZ235, a dual PI3K/mTOR inhibitor shown to destabilize MYCN via GSK3β activation. In contrast, tumors caused in MYCN/NCYM double transgenic mice showed chemo-resistance to the drug. Collectively, our results show that NCYM is the first de novo evolved protein known to act as an oncopromoting factor in human cancer, and suggest that de novo evolved proteins may functionally characterize human disease. The MYCN oncogene has a central role in the genesis and progression of neuroblastomas, and its amplification is associated with an unfavorable prognosis. We have found that NCYM, a MYCN cis-antisense RNA, is translated in humans to a de novo evolved protein. NCYM inhibits GSK3β to stabilize MYCN, whereas MYCN induces NCYM transcription. The positive feedback regulation formed in the MYCN/NCYM-amplified tumors promotes the aggressive nature of human neuroblastoma. MYCN transgenic mice, which express human MYCN in sympathoadrenal tissues, spontaneously develop neuroblastomas. However, unlike human neuroblastoma, distant metastasis is infrequent. We established MYCN/NCYM double transgenic mice as a new animal model for studying neuroblastoma pathogenesis. We found that NCYM expression promoted both the metastasis and chemo-resistance of the neuroblastomas formed in the double transgenic mice. These results demonstrate that NCYM may be a potential target for developing novel therapeutic tools against high-risk neuroblastomas in humans, and that the MYCN/NCYM double transgenic mouse may be a suitable model for the screening of these new drugs.
Collapse
Affiliation(s)
- Yusuke Suenaga
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - S. M. Rafiqul Islam
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Jennifer Alagu
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Yoshiki Kaneko
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Mamoru Kato
- Division of Cancer Genomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Yukichi Tanaka
- Department of Diagnostic Pathology, Research Institute, Kanagawa Children's Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Japan
| | - Hidetada Kawana
- Division of Surgical Pathology, Chiba Cancer Center, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Shamim Hossain
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Daisuke Matsumoto
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Mami Yamamoto
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Wataru Shoji
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
- Department of Pediatric Surgery, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Makiko Itami
- Division of Surgical Pathology, Chiba Cancer Center, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, Japan
| | - Yohko Nakamura
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Miki Ohira
- Laboratory of Cancer Genomics, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Seiki Haraguchi
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Atsushi Takatori
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
| | - Akira Nakagawara
- Division of Biochemistry and Innovative Cancer Therapeutics and Children's Cancer Research Center, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuo-ku, Chiba, Japan
- * E-mail:
| |
Collapse
|
40
|
Jay V, MacNeill S, Zielenska M. MYCN Amplification in Pediatric Brain Tumors: Detection by Differential Polymerase Chain Reaction. J Histotechnol 2013. [DOI: 10.1179/his.1997.20.2.115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
41
|
Marotta M, Chen X, Inoshita A, Stephens R, Budd GT, Crowe JP, Lyons J, Kondratova A, Tubbs R, Tanaka H. A common copy-number breakpoint of ERBB2 amplification in breast cancer colocalizes with a complex block of segmental duplications. Breast Cancer Res 2012. [PMID: 23181561 PMCID: PMC4053137 DOI: 10.1186/bcr3362] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introduction Segmental duplications (low-copy repeats) are the recently duplicated genomic segments in the human genome that display nearly identical (> 90%) sequences and account for about 5% of euchromatic regions. In germline, duplicated segments mediate nonallelic homologous recombination and thus cause both non-disease-causing copy-number variants and genomic disorders. To what extent duplicated segments play a role in somatic DNA rearrangements in cancer remains elusive. Duplicated segments often cluster and form genomic blocks enriched with both direct and inverted repeats (complex genomic regions). Such complex regions could be fragile and play a mechanistic role in the amplification of the ERBB2 gene in breast tumors, because repeated sequences are known to initiate gene amplification in model systems. Methods We conducted polymerase chain reaction (PCR)-based assays for primary breast tumors and analyzed publically available array-comparative genomic hybridization data to map a common copy-number breakpoint in ERBB2-amplified primary breast tumors. We further used molecular, bioinformatics, and population-genetics approaches to define duplication contents, structural variants, and haplotypes within the common breakpoint. Results We found a large (> 300-kb) block of duplicated segments that was colocalized with a common-copy number breakpoint for ERBB2 amplification. The breakpoint that potentially initiated ERBB2 amplification localized in a region 1.5 megabases (Mb) on the telomeric side of ERBB2. The region is very complex, with extensive duplications of KRTAP genes, structural variants, and, as a result, a paucity of single-nucleotide polymorphism (SNP) markers. Duplicated segments are varied in size and degree of sequence homology, indicating that duplications have occurred recurrently during genome evolution. Conclusions Amplification of the ERBB2 gene in breast tumors is potentially initiated by a complex region that has unusual genomic features and thus requires rigorous, labor-intensive investigation. The haplotypes we provide could be useful to identify the potential association between the complex region and ERBB2 amplification.
Collapse
|
42
|
Sottile F, Gnemmi I, Cantilena S, D'Acunto WC, Sala A. A chemical screen identifies the chemotherapeutic drug topotecan as a specific inhibitor of the B-MYB/MYCN axis in neuroblastoma. Oncotarget 2012; 3:535-45. [PMID: 22619121 PMCID: PMC3388183 DOI: 10.18632/oncotarget.498] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The transcription factor MycN is the prototypical neuroblastoma oncogene and a potential therapeutic target. However, its strong expression caused by gene amplification in about 30% of neuroblastoma patients is a considerable obstacle to the development of therapeutic approaches aiming at eliminating its tumourigenic activity. We have previously reported that B-Myb is essentially required for transcription of the MYCN amplicon and have also shown that B-MYB and MYCN are engaged in a feed forward loop promoting the survival/proliferation of neuroblastoma cells. We postulated that pharmacological strategies breaking the B-MYB/MYCN axis should result in clinically desirable effects. Thus, we implemented a high throughput chemical screen, using a curated library of ~1500 compounds from the National Cancer Institute, whose endpoint was the identification of small molecules that inhibited B-Myb. At the end of the screening, we found that the compounds pinafide, ellipticine and camptothecin inhibited B-Myb transcriptional activity in luciferase assays. One of the compounds, the topoisomerase-1 inhibitor camptothecin, is of considerable clinical interest since its derivatives topotecan and irinotecan are currently used as first and second line treatment agents for various types of cancer, including neuroblastoma. We found that neuroblastoma cells with amplification of MYCN are more sensitive than MYCN negative cells to camptothecin and topotecan killing. Campothecin and topotecan caused selective down-regulation of B-Myb and MycN expression in neuroblastoma cells. Notably, forced overexpression of B-Myb could antagonize the killing effect of topotecan and camptothecin, demonstrating that the transcription factor is a key target of the drugs. These results suggest that camptothecin and its analogues should be more effective in patients whose tumours feature amplification of MYCN and/or overexpression of B-MYB.
Collapse
|
43
|
Lorenzi S, Forloni M, Cifaldi L, Antonucci C, Citti A, Boldrini R, Pezzullo M, Castellano A, Russo V, van der Bruggen P, Giacomini P, Locatelli F, Fruci D. IRF1 and NF-kB restore MHC class I-restricted tumor antigen processing and presentation to cytotoxic T cells in aggressive neuroblastoma. PLoS One 2012; 7:e46928. [PMID: 23071666 PMCID: PMC3465322 DOI: 10.1371/journal.pone.0046928] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Accepted: 09/06/2012] [Indexed: 02/06/2023] Open
Abstract
Neuroblastoma (NB), the most common solid extracranial cancer of childhood, displays a remarkable low expression of Major Histocompatibility Complex class I (MHC-I) and Antigen Processing Machinery (APM) molecules, including Endoplasmic Reticulum (ER) Aminopeptidases, and poorly presents tumor antigens to Cytotoxic T Lymphocytes (CTL). We have previously shown that this is due to low expression of the transcription factor NF-kB p65. Herein, we show that not only NF-kB p65, but also the Interferon Regulatory Factor 1 (IRF1) and certain APM components are low in a subset of NB cell lines with aggressive features. Whereas single transfection with either IRF1, or NF-kB p65 is ineffective, co-transfection results in strong synergy and substantial reversion of the MHC-I/APM-low phenotype in all NB cell lines tested. Accordingly, linked immunohistochemistry expression patterns between nuclear IRF1 and p65 on the one hand, and MHC-I on the other hand, were observed in vivo. Absence and presence of the three molecules neatly segregated between high-grade and low-grade NB, respectively. Finally, APM reconstitution by double IRF1/p65 transfection rendered a NB cell line susceptible to killing by anti MAGE-A3 CTLs, lytic efficiency comparable to those seen upon IFN-γ treatment. This is the first demonstration that a complex immune escape phenotype can be rescued by reconstitution of a limited number of master regulatory genes. These findings provide molecular insight into defective MHC-I expression in NB cells and provide the rational for T cell-based immunotherapy in NB variants refractory to conventional therapy.
Collapse
Affiliation(s)
- Silvia Lorenzi
- Paediatric Haematology/Oncology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Matteo Forloni
- Paediatric Haematology/Oncology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Loredana Cifaldi
- Paediatric Haematology/Oncology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Chiara Antonucci
- Paediatric Haematology/Oncology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Arianna Citti
- Pathology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Renata Boldrini
- Pathology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Marco Pezzullo
- Paediatric Haematology/Oncology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Aurora Castellano
- Paediatric Haematology/Oncology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Vincenzo Russo
- Cancer Gene Therapy Unit, Scientific Institute San Raffaele, Milan, Italy
| | | | - Patrizio Giacomini
- Ludwig Institute for Cancer Research and Université Catholique de Louvain, de Duve Institute, Brussels, Belgium
| | - Franco Locatelli
- Paediatric Haematology/Oncology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- University of Pavia, Pavia, Italy
| | - Doriana Fruci
- Paediatric Haematology/Oncology Department, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- * E-mail:
| |
Collapse
|
44
|
Berry T, Luther W, Bhatnagar N, Jamin Y, Poon E, Sanda T, Pei D, Sharma B, Vetharoy WR, Hallsworth A, Ahmad Z, Barker K, Moreau L, Webber H, Wang W, Liu Q, Perez-Atayde A, Rodig S, Cheung NK, Raynaud F, Hallberg B, Robinson SP, Gray NS, Pearson AD, Eccles SA, Chesler L, George RE. The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma. Cancer Cell 2012; 22:117-30. [PMID: 22789543 PMCID: PMC3417812 DOI: 10.1016/j.ccr.2012.06.001] [Citation(s) in RCA: 229] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Revised: 03/18/2012] [Accepted: 06/05/2012] [Indexed: 01/03/2023]
Abstract
The ALK(F1174L) mutation is associated with intrinsic and acquired resistance to crizotinib and cosegregates with MYCN in neuroblastoma. In this study, we generated a mouse model overexpressing ALK(F1174L) in the neural crest. Compared to ALK(F1174L) and MYCN alone, co-expression of these two oncogenes led to the development of neuroblastomas with earlier onset, higher penetrance, and enhanced lethality. ALK(F1174L)/MYCN tumors exhibited increased MYCN dosage due to ALK(F1174L)-induced activation of the PI3K/AKT/mTOR and MAPK pathways, coupled with suppression of MYCN pro-apoptotic effects. Combined treatment with the ATP-competitive mTOR inhibitor Torin2 overcame the resistance of ALK(F1174L)/MYCN tumors to crizotinib. Our findings demonstrate a pathogenic role for ALK(F1174L) in neuroblastomas overexpressing MYCN and suggest a strategy for improving targeted therapy for ALK-positive neuroblastoma.
Collapse
Affiliation(s)
- Teeara Berry
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - William Luther
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Namrata Bhatnagar
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Yann Jamin
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Evon Poon
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Takaomi Sanda
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Desheng Pei
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Bandana Sharma
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Winston R. Vetharoy
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Albert Hallsworth
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Zai Ahmad
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Karen Barker
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Lisa Moreau
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Hannah Webber
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Wenchao Wang
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Qingsong Liu
- Departments of Cancer Biology, Dana Farber Cancer Institute and Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | | | - Scott Rodig
- Department of Pathology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Nai-Kong Cheung
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Florence Raynaud
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Bengt Hallberg
- Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Simon P. Robinson
- Division of Radiotherapy & Imaging, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Nathanael S. Gray
- Departments of Cancer Biology, Dana Farber Cancer Institute and Biological Chemistry & Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Andrew D.J. Pearson
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
- The Children and Young People’s Unit, The Royal Marsden NHS Trust, Sutton, Surrey, UK
| | - Suzanne A. Eccles
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
| | - Louis Chesler
- Divisions of Clinical Studies & Cancer Therapeutics, The Institute of Cancer Research, Sutton, Surrey, UK
- The Children and Young People’s Unit, The Royal Marsden NHS Trust, Sutton, Surrey, UK
| | - Rani E. George
- Department of Pediatric Hematology & Oncology, Dana-Farber Cancer Institute and Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Ye S, Tan L, Yang R, Fang B, Qu S, Schulze EN, Song H, Ying Q, Li P. Pleiotropy of glycogen synthase kinase-3 inhibition by CHIR99021 promotes self-renewal of embryonic stem cells from refractory mouse strains. PLoS One 2012; 7:e35892. [PMID: 22540008 PMCID: PMC3335080 DOI: 10.1371/journal.pone.0035892] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 03/23/2012] [Indexed: 12/20/2022] Open
Abstract
Background Inhibition of glycogen synthase kinase-3 (GSK-3) improves the efficiency of embryonic stem (ES) cell derivation from various strains of mice and rats, as well as dramatically promotes ES cell self-renewal potential. β-catenin has been reported to be involved in the maintenance of self-renewal of ES cells through TCF dependent and independent pathway. But the intrinsic difference between ES cell lines from different species and strains has not been characterized. Here, we dissect the mechanism of GSK-3 inhibition by CHIR99021 in mouse ES cells from refractory mouse strains. Methodology/Principal Findings We found that CHIR99021, a GSK-3 specific inhibitor, promotes self-renewal of ES cells from recalcitrant C57BL/6 (B6) and BALB/c mouse strains through stabilization of β-catenin and c-Myc protein levels. Stabilized β-catenin promoted ES self-renewal through two mechanisms. First, β-catenin translocated into the nucleus to maintain stem cell pluripotency in a lymphoid-enhancing factor/T-cell factor–independent manner. Second, β-catenin binds plasma membrane-localized E-cadherin, which ensures a compact, spherical morphology, a hallmark of ES cells. Further, elevated c-Myc protein levels did not contribute significantly to CH-mediated ES cell self-renewal. Instead, the role of c-Myc is dependent on its transformation activity and can be replaced by N-Myc but not L-Myc. β-catenin and c-Myc have similar effects on ES cells derived from both B6 and BALB/c mice. Conclusions/Significance Our data demonstrated that GSK-3 inhibition by CH promotes self-renewal of mouse ES cells with non-permissive genetic backgrounds by regulation of multiple signaling pathways. These findings would be useful to improve the availability of normally non-permissive mouse strains as research tools.
Collapse
Affiliation(s)
- Shoudong Ye
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Shanghai, People's Republic of China
| | - Li Tan
- Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Rongqing Yang
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Shanghai, People's Republic of China
| | - Bo Fang
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Shanghai, People's Republic of China
| | - Su Qu
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Shanghai, People's Republic of China
| | - Eric N. Schulze
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Houyan Song
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Shanghai, People's Republic of China
| | - Qilong Ying
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Ping Li
- The Key Laboratory of Molecular Medicine, Ministry of Education, Shanghai Medical College, Shanghai, People's Republic of China
- * E-mail:
| |
Collapse
|
46
|
Bagci O, Tumer S, Olgun N, Altungoz O. Copy number status and mutation analyses of anaplastic lymphoma kinase (ALK) gene in 90 sporadic neuroblastoma tumors. Cancer Lett 2011; 317:72-7. [PMID: 22085494 DOI: 10.1016/j.canlet.2011.11.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 11/05/2011] [Accepted: 11/08/2011] [Indexed: 11/19/2022]
Abstract
Somatic and germline mutations of the anaplastic lymphoma kinase (ALK) gene were recently described in neuroblastoma (NB). In this study, we investigated the association of ALK copy number alterations with copy number status 2p24.1 amplicon harboring DEAD box polypeptide 1 (DDX1), MYCN and neuroblastoma-amplified (NAG) genes in 90 primary tumors of sporadic NB cases by multiplex ligation-dependent probe amplification (MLPA). We also performed mutation analysis of ALK gene by directly sequencing the exons 20-28 which cover the region that encodes juxtamembrane and kinase domains. A total of 39 (43.3%) NB cases revealed copy numbers alterations of ALK gene. There was highly significant association of ALK copy number gains with gains of one or more of the genes at 2p24.1 (DDX1, MYCN or NAG) in MYCN unamplified tumors (P<0.000). In addition, 15 of 17 MYCN amplified cases (88.2%) had aberrant ALK status. Solitary gain of ALK with normal copy number status of all other genes was observed only in one case. DNA sequencing of exons 20-28 of ALK revealed two different nucleotide changes in three cases leading to amino acid substitutions of F1245V and R1275Q in tyrosine kinase domain. In conclusion, the frequency of ALK mutations in NB is low and solitary copy number change of it is rarely observed.
Collapse
Affiliation(s)
- Ozkan Bagci
- Department of Medical Biology and Genetics, Dokuz Eylul University, School of Medicine, Izmir, Turkey
| | | | | | | |
Collapse
|
47
|
Jiang M, Stanke J, Lahti JM. The connections between neural crest development and neuroblastoma. Curr Top Dev Biol 2011; 94:77-127. [PMID: 21295685 DOI: 10.1016/b978-0-12-380916-2.00004-8] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neuroblastoma (NB), the most common extracranial solid tumor in childhood, is an extremely heterogeneous disease both biologically and clinically. Although significant progress has been made in identifying molecular and genetic markers for NB, this disease remains an enigmatic challenge. Since NB is thought to be an embryonal tumor that is derived from precursor cells of the peripheral (sympathetic) nervous system, understanding the development of normal sympathetic nervous system may highlight abnormal events that contribute to NB initiation. Therefore, this review focuses on the development of the peripheral trunk neural crest, the current understanding of how developmental factors may contribute to NB and on recent advances in the identification of important genetic lesions and signaling pathways involved in NB tumorigenesis and metastasis. Finally, we discuss how future advances in identification of molecular alterations in NB may lead to more effective, less toxic therapies, and improve the prognosis for NB patients.
Collapse
Affiliation(s)
- Manrong Jiang
- Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | |
Collapse
|
48
|
Wong JV, Yao G, Nevins JR, You L. Viral-mediated noisy gene expression reveals biphasic E2f1 response to MYC. Mol Cell 2011; 41:275-85. [PMID: 21292160 DOI: 10.1016/j.molcel.2011.01.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Revised: 07/08/2010] [Accepted: 11/24/2010] [Indexed: 12/28/2022]
Abstract
Gene expression mediated by viral vectors is subject to cell-to-cell variability, which limits the accuracy of gene delivery. When coupled with single-cell measurements, however, such variability provides an efficient means to quantify signaling dynamics in mammalian cells. Here, we illustrate the utility of this approach by mapping the E2f1 response to MYC, serum stimulation, or both. Our results revealed an underappreciated mode of gene regulation: E2f1 expression first increased, then decreased as MYC input increased. This biphasic pattern was also reflected in other nodes of the network, including the miR-17-92 microRNA cluster and p19Arf. A mathematical model of the network successfully predicted modulation of the biphasic E2F response by serum and a CDK inhibitor. In addition to demonstrating how noise can be exploited to probe signaling dynamics, our results reveal how coordination of the MYC/RB/E2F pathway enables dynamic discrimination of aberrant and normal levels of growth stimulation.
Collapse
Affiliation(s)
- Jeffrey V Wong
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | | | | |
Collapse
|
49
|
Forloni M, Albini S, Limongi MZ, Cifaldi L, Boldrini R, Nicotra MR, Giannini G, Natali PG, Giacomini P, Fruci D. NF-kappaB, and not MYCN, regulates MHC class I and endoplasmic reticulum aminopeptidases in human neuroblastoma cells. Cancer Res 2010; 70:916-24. [PMID: 20103633 DOI: 10.1158/0008-5472.can-09-2582] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neuroblastoma (NB) is the most common solid extracranial cancer of childhood. Amplification and overexpression of the MYCN oncogene characterize the most aggressive forms and are believed to severely downregulate MHC class I molecules by transcriptional inhibition of the p50 NF-kappaB subunit. In this study, we found that in human NB cell lines, high MYCN expression is not responsible for low MHC class I expression because neither transfection-mediated overexpression nor small interfering RNA suppression of MYCN affects MHC class I and p50 levels. Furthermore, we identified NF-kappaB as the immediate upstream regulator of MHC class I because the p65 NF-kappaB subunit binds MHC class I promoter in chromatin immunoprecipitation experiments, and MHC class I expression is enhanced by p65 transfection and reduced by (a) the chemical NF-kappaB inhibitor sulfasalazine, (b) a dominant-negative IKBalpha gene, and (c) p65 silencing. Moreover, we showed that the endoplasmic reticulum aminopeptidases ERAP1 and ERAP2, which generate MHC class I binding peptides, are regulated by NF-kappaB, contain functional NF-kappaB-binding elements in their promoters, and mimic MHC class I molecules in the expression pattern. Consistent with these findings, nuclear p65 was detected in NB cells that express MHC class I molecules in human NB specimens. Thus, the coordinated downregulation of MHC class I, ERAP1, and ERAP2 in aggressive NB cells is attributable to a low transcriptional availability of NF-kappaB, possibly due to an unknown suppressor other than MYCN.
Collapse
|
50
|
Besançon R, Valsesia-Wittmann S, Locher C, Delloye-Bourgeois C, Furhman L, Tutrone G, Bertrand C, Jallas AC, Garin E, Puisieux A. Upstream ORF affects MYCN translation depending on exon 1b alternative splicing. BMC Cancer 2009; 9:445. [PMID: 20017904 PMCID: PMC2810302 DOI: 10.1186/1471-2407-9-445] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 12/17/2009] [Indexed: 12/24/2022] Open
Abstract
Background The MYCN gene is transcribed into two major mRNAs: one full-length (MYCN) and one exon 1b-spliced (MYCNΔ1b) mRNA. But nothing is known about their respective ability to translate the MYCN protein. Methods Plasmids were prepared to enable translation from the upstream (uORF) and major ORF of the two MYCN transcripts. Translation was studied after transfection in neuroblastoma SH-EP cell line. Impact of the upstream AUG on translation was evaluated after directed mutagenesis. Functional study with the two MYCN mRNAs was conducted by a cell viability assay. Existence of a new protein encoded by the MYCNΔ1b uORF was explored by designing a rabbit polyclonal antibody against a specific epitope of this protein. Results Both are translated, but higher levels of protein were seen with MYCNΔ1b mRNA. An upstream ORF was shown to have positive cis-regulatory activity on translation from MYCN but not from MYCNΔ1b mRNA. In transfected SH-EP neuroblastoma cells, high MYCN dosage obtained with MYCNΔ1b mRNA translation induces an antiapoptotic effect after serum deprivation that was not observed with low MYCN expression obtained with MYCN mRNA. Here, we showed that MYCNOT: MYCN Overlap Transcript, a new protein of unknown function is translated from the upstream AUG of MYCNΔ1b mRNA. Conclusions Existence of upstream ORF in MYCN transcripts leads to a new level of MYCN regulation. The resulting MYCN dosage has a weak but significant anti-apoptotic activity after intrinsic apoptosis induction.
Collapse
|