1
|
Schmidt N, Kumar A, Korf L, Dinh-Fricke AV, Abendroth F, Koide A, Linne U, Rakwalska-Bange M, Koide S, Essen LO, Vázquez O, Hantschel O. Development of mirror-image monobodies targeting the oncogenic BCR::ABL1 kinase. Nat Commun 2024; 15:10724. [PMID: 39715735 PMCID: PMC11666773 DOI: 10.1038/s41467-024-54901-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/28/2024] [Indexed: 12/25/2024] Open
Abstract
Mirror-image proteins, composed of D-amino acids, are an attractive therapeutic modality, as they exhibit high metabolic stability and lack immunogenicity. Development of mirror-image binding proteins is achieved through chemical synthesis of D-target proteins, phage display library selection of L-binders and chemical synthesis of (mirror-image) D-binders that consequently bind the physiological L-targets. Monobodies are well-established synthetic (L-)binding proteins and their small size (~90 residues) and lack of endogenous cysteine residues make them particularly accessible to chemical synthesis. Here, we develop monobodies with nanomolar binding affinities against the D-SH2 domain of the leukemic tyrosine kinase BCR::ABL1. Two crystal structures of heterochiral monobody-SH2 complexes reveal targeting of the pY binding pocket by an unconventional binding mode. We then prepare potent D-monobodies by either ligating two chemically synthesized D-peptides or by self-assembly without ligation. Their proper folding and stability are determined and high-affinity binding to the L-target is shown. D-monobodies are protease-resistant, show long-term plasma stability, inhibit BCR::ABL1 kinase activity and bind BCR::ABL1 in cell lysates and permeabilized cells. Hence, we demonstrate that functional D-monobodies can be developed readily. Our work represents an important step towards possible future therapeutic use of D-monobodies when combined with emerging methods to enable cytoplasmic delivery of monobodies.
Collapse
Affiliation(s)
- Nina Schmidt
- Institute of Physiological Chemistry, Faculty of Medicine, Philipps University of Marburg, Marburg, Germany
| | - Amit Kumar
- Institute of Physiological Chemistry, Faculty of Medicine, Philipps University of Marburg, Marburg, Germany
| | - Lukas Korf
- Faculty of Chemistry and Unit for Structural Biology, Philipps University of Marburg, Marburg, Germany
| | | | - Frank Abendroth
- Faculty of Chemistry and Unit for Chemical Biology, Philipps University of Marburg, Marburg, Germany
| | - Akiko Koide
- Department of Medicine, New York University School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Uwe Linne
- Faculty of Chemistry and Unit for Mass Spectrometry, Philipps University of Marburg, Marburg, Germany
| | - Magdalena Rakwalska-Bange
- Institute of Physiological Chemistry, Faculty of Medicine, Philipps University of Marburg, Marburg, Germany
| | - Shohei Koide
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Lars-Oliver Essen
- Faculty of Chemistry and Unit for Structural Biology, Philipps University of Marburg, Marburg, Germany
| | - Olalla Vázquez
- Faculty of Chemistry and Unit for Chemical Biology, Philipps University of Marburg, Marburg, Germany.
- Center for Synthetic Microbiology (SYNMIKRO), Philipps University of Marburg, Marburg, Germany.
| | - Oliver Hantschel
- Institute of Physiological Chemistry, Faculty of Medicine, Philipps University of Marburg, Marburg, Germany.
| |
Collapse
|
2
|
Terakawa A, Hu Y, Kokaji T, Yugi K, Morita K, Ohno S, Pan Y, Bai Y, Parkhitko AA, Ni X, Asara JM, Bulyk ML, Perrimon N, Kuroda S. Trans-omics analysis of insulin action reveals a cell growth subnetwork which co-regulates anabolic processes. iScience 2022; 25:104231. [PMID: 35494245 PMCID: PMC9044165 DOI: 10.1016/j.isci.2022.104231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/09/2022] [Accepted: 04/06/2022] [Indexed: 12/16/2022] Open
Abstract
Insulin signaling promotes anabolic metabolism to regulate cell growth through multi-omic interactions. To obtain a comprehensive view of the cellular responses to insulin, we constructed a trans-omic network of insulin action in Drosophila cells that involves the integration of multi-omic data sets. In this network, 14 transcription factors, including Myc, coordinately upregulate the gene expression of anabolic processes such as nucleotide synthesis, transcription, and translation, consistent with decreases in metabolites such as nucleotide triphosphates and proteinogenic amino acids required for transcription and translation. Next, as cell growth is required for cell proliferation and insulin can stimulate proliferation in a context-dependent manner, we integrated the trans-omic network with results from a CRISPR functional screen for cell proliferation. This analysis validates the role of a Myc-mediated subnetwork that coordinates the activation of genes involved in anabolic processes required for cell growth. A trans-omic network of insulin action in Drosophila cells was constructed Insulin co-regulates various anabolic processes in a time-dependent manner The trans-omic network and a CRISPR screen for cell proliferation were integrated A Myc-mediated subnetwork promoting anabolic processes is required for cell growth
Collapse
Affiliation(s)
- Akira Terakawa
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Toshiya Kokaji
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Data Science Center, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara, Japan
| | - Katsuyuki Yugi
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Institute for Advanced Biosciences, Keio University, Fujisawa, 252-8520, Japan
| | - Keigo Morita
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Satoshi Ohno
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Yifei Pan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Yunfan Bai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Andrey A. Parkhitko
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Aging Institute of UPMC and the University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaochun Ni
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - John M. Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02175, USA
| | - Martha L. Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham & Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Corresponding author
| | - Shinya Kuroda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
- Corresponding author
| |
Collapse
|
3
|
Wang S, Zhang C, Li M, Zhao C, Zheng Y. A System-Wide Spatiotemporal Characterization of ErbB Receptor Complexes by Subcellular Fractionation Integrated Quantitative Mass Spectrometry. Anal Chem 2021; 93:7933-7941. [PMID: 34033713 DOI: 10.1021/acs.analchem.1c00651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Precise spatiotemporal regulation of protein complex assembly is essential for cells to achieve a meaningful rely of information flow via intracellular signaling networks in response to extracellular cues, whose disruption would lead to disease. Although various attempts have been made for spatial and/or temporal analysis of protein complexes, it is still a challenge to track cell-wide dynamics of a particular protein complex under physiological conditions. Here we describe a workflow that combines endogenous expression of tagged proteins, organelle marker distribution-directed subcellular fractionation, scaffold protein-mediated receptor complex purification, and targeted proteomics for spatiotemporal quantification of protein complexes in whole cell scale. We applied our method to investigate the assembly kinetics of EGF-dependent ErbB receptor complexes. After fractionation using the density gradient centrifugation and organelle assignment based on organelle markers, endogenous ErbB complex in different subcellular fractionation was efficiently enriched. By using targeted mass spectrometry, ErbB complex components that expressed medium to low level was precisely quantified with in-depth coverage, simultaneously in time and subcellular spaces. Our results revealed a sophisticated scheme of complex behaviors characterized by multiple subcomplexes with distinct molecular composition formed across subcellular fractions enriched with cytosol, plasma membrane, endosome, or mitochondria, implying organelle-specific ErbB functions. Remarkably, our results demonstrated for the first time that activated ErbB receptors might increase their signaling range through promoting a cytosolic, receptor-free subcomplex, consisting of Shc1, Grb2, Arhgef5, Garem1, and Lrrk1. These findings emphasize the potential of our strategy as a powerful tool to study spatiotemporal dynamics of protein complexes.
Collapse
Affiliation(s)
- Shujuan Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Cunjie Zhang
- SickKids Research Institute, cell biology 686 Bay St, Toronto, Ontario CAN M5G 0A4, Canada
| | - Mansheng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Chao Zhao
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yong Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
4
|
Genes That Predict Poor Prognosis in Breast Cancer via Bioinformatical Analysis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6649660. [PMID: 33959662 PMCID: PMC8075678 DOI: 10.1155/2021/6649660] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/01/2021] [Accepted: 04/07/2021] [Indexed: 12/19/2022]
Abstract
Background Breast cancer is one of the most commonly diagnosed cancers all over the world, and it is now the leading cause of cancer death among females. The aim of this study was to find DEGs (differentially expressed genes) which can predict poor prognosis in breast cancer and be effective targets for breast cancer patients via bioinformatical analysis. Methods GSE86374, GSE5364, and GSE70947 were chosen from the GEO database. DEGs between breast cancer tissues and normal breast tissues were picked out by GEO2R and Venn diagram software. Then, DAVID (Database for Annotation, Visualization, and Integrated Discovery) was used to analyze these DEGs in gene ontology (GO) including molecular function (MF), cellular component (CC), and biological process (BP) and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway. Next, STRING (Search Tool for the Retrieval of Interacting Genes) was used to investigate potential protein-protein interaction (PPI) relationships among DEGs and these DEGs were analyzed by Molecular Complex Detection (MCODE) in Cytoscape. After that, UALCAN, GEPIA (gene expression profiling interactive analysis), and KM (Kaplan-Meier plotter) were used for the prognostic information and core genes were qualified. Results There were 96 upregulated genes and 98 downregulated genes in this study. 55 upregulated genes were selected as hub genes in the PPI network. For validation in UALCAN, GEPIA, and KM, 5 core genes (KIF4A, RACGAP1, CKS2, SHCBP1, and HMMR) were found to highly expressed in breast cancer tissues with poor prognosis. They differentially expressed between different subclasses of breast cancer. Conclusion These five genes (KIF4A, RACGAP1, CKS2, SHCBP1, and HMMR) could be potential targets for therapy in breast cancer and prediction of prognosis on the basis of bioinformatical analysis.
Collapse
|
5
|
Bury M, Le Calvé B, Ferbeyre G, Blank V, Lessard F. New Insights into CDK Regulators: Novel Opportunities for Cancer Therapy. Trends Cell Biol 2021; 31:331-344. [PMID: 33676803 DOI: 10.1016/j.tcb.2021.01.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022]
Abstract
Cyclins and their catalytic partners, the cyclin-dependent kinases (CDKs), control the transition between different phases of the cell cycle. CDK/cyclin activity is regulated by CDK inhibitors (CKIs), currently comprising the CDK-interacting protein/kinase inhibitory protein (CIP/KIP) family and the inhibitor of kinase (INK) family. Recent studies have identified a third group of CKIs, called ribosomal protein-inhibiting CDKs (RPICs). RPICs were discovered in the context of cellular senescence, a stable cell cycle arrest with tumor-suppressing abilities. RPICs accumulate in the nonribosomal fraction of senescent cells due to a decrease in rRNA biogenesis. Accordingly, RPICs are often downregulated in human cancers together with other ribosomal proteins, the tumor-suppressor functions of which are still under study. In this review, we discuss unique therapies that have been developed to target CDK activity in the context of cancer treatment or senescence-associated pathologies, providing novel tools for precision medicine.
Collapse
Affiliation(s)
- Marina Bury
- De Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | | | - Gerardo Ferbeyre
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, H3C 3J7, Canada.
| | - Volker Blank
- Lady Davis Institute for Medical Research, Departments of Medicine and Physiology, McGill University, Montreal, QC, H3T 1E2, Canada.
| | - Frédéric Lessard
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, H3C 3J7, Canada.
| |
Collapse
|
6
|
Buljan M, Ciuffa R, van Drogen A, Vichalkovski A, Mehnert M, Rosenberger G, Lee S, Varjosalo M, Pernas LE, Spegg V, Snijder B, Aebersold R, Gstaiger M. Kinase Interaction Network Expands Functional and Disease Roles of Human Kinases. Mol Cell 2020; 79:504-520.e9. [PMID: 32707033 PMCID: PMC7427327 DOI: 10.1016/j.molcel.2020.07.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 02/14/2020] [Accepted: 06/30/2020] [Indexed: 12/30/2022]
Abstract
Protein kinases are essential for signal transduction and control of most cellular processes, including metabolism, membrane transport, motility, and cell cycle. Despite the critical role of kinases in cells and their strong association with diseases, good coverage of their interactions is available for only a fraction of the 535 human kinases. Here, we present a comprehensive mass-spectrometry-based analysis of a human kinase interaction network covering more than 300 kinases. The interaction dataset is a high-quality resource with more than 5,000 previously unreported interactions. We extensively characterized the obtained network and were able to identify previously described, as well as predict new, kinase functional associations, including those of the less well-studied kinases PIM3 and protein O-mannose kinase (POMK). Importantly, the presented interaction map is a valuable resource for assisting biomedical studies. We uncover dozens of kinase-disease associations spanning from genetic disorders to complex diseases, including cancer.
Collapse
Affiliation(s)
- Marija Buljan
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland; Empa, Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland
| | - Rodolfo Ciuffa
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Audrey van Drogen
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Anton Vichalkovski
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Martin Mehnert
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - George Rosenberger
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland; Columbia University Department of Systems Biology, New York, NY 10032, USA
| | - Sohyon Lee
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
| | - Lucia Espona Pernas
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Vincent Spegg
- Department of Molecular Mechanisms of Disease, University of Zurich, 8057 Zurich, Switzerland
| | - Berend Snijder
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Ruedi Aebersold
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland; Faculty of Science, University of Zurich, Zurich, Switzerland.
| | - Matthias Gstaiger
- Institute of Molecular Systems Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
7
|
Chung AH, Leisner TM, Dardis GJ, Bivins MM, Keller AL, Parise LV. CIB1 depletion with docetaxel or TRAIL enhances triple-negative breast cancer cell death. Cancer Cell Int 2019; 19:26. [PMID: 30740034 PMCID: PMC6360800 DOI: 10.1186/s12935-019-0740-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Background Patients diagnosed with triple negative breast cancer (TNBC) have limited treatment options and often suffer from resistance and toxicity due to chemotherapy. We previously found that depleting calcium and integrin-binding protein 1 (CIB1) induces cell death selectively in TNBC cells, while sparing normal cells. Therefore, we asked whether CIB1 depletion further enhances tumor-specific killing when combined with either the commonly used chemotherapeutic, docetaxel, or the cell death-inducing ligand, TRAIL. Methods We targeted CIB1 by RNA interference in MDA-MB-436, MDA-MB-231, MDA-MB-468, docetaxel-resistant MDA-MB-436 TNBC cells and ME16C normal breast epithelial cells alone or combination with docetaxel or TRAIL. Cell death was quantified via trypan blue exclusion using flow cytometry and cell death mechanisms were analyzed by Western blotting. Cell surface levels of TRAIL receptors were measured by flow cytometry analysis. Results CIB1 depletion combined with docetaxel significantly enhanced tumor-specific cell death relative to each treatment alone. The enhanced cell death strongly correlated with caspase-8 activation, a hallmark of death receptor-mediated apoptosis. The death receptor TRAIL-R2 was upregulated in response to CIB1 depletion, which sensitized TNBC cells to the ligand TRAIL, resulting in a synergistic increase in cell death. In addition to death receptor-mediated apoptosis, both combination treatments activated a non-apoptotic mechanism, called paraptosis. Interestingly, these combination treatments also induced nearly complete death of docetaxel-resistant MDA-MB-436 cells, again via apoptosis and paraptosis. In contrast, neither combination treatment induced cell death in normal ME16C cells. Conclusion Novel combinations of CIB1 depletion with docetaxel or TRAIL selectively enhance naive and docetaxel-resistant TNBC cell death while sparing normal cell. Therefore, combination therapies that target CIB1 could prove to be a safe and durable strategy for treatment of TNBC and potentially other cancers. Electronic supplementary material The online version of this article (10.1186/s12935-019-0740-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexander H Chung
- 1Department of Pharmacology, University of North Carolina at Chapel Hill, CB #7365, Chapel Hill, NC 27599 USA
| | - Tina M Leisner
- 2Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, CB #7260, Chapel Hill, NC 27599 USA
| | - Gabrielle J Dardis
- 2Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, CB #7260, Chapel Hill, NC 27599 USA
| | - Marissa M Bivins
- 1Department of Pharmacology, University of North Carolina at Chapel Hill, CB #7365, Chapel Hill, NC 27599 USA
| | - Alana L Keller
- 2Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, CB #7260, Chapel Hill, NC 27599 USA
| | - Leslie V Parise
- 2Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, CB #7260, Chapel Hill, NC 27599 USA.,3Lineberger Comprehensive Cancer Center, Chapel Hill, NC USA
| |
Collapse
|
8
|
Scumaci D, Oliva A, Concolino A, Curcio A, Fiumara CV, Tammè L, Campuzano O, Pascali VL, Coll M, Iglesias A, Berne P, Casu G, Olivo E, Ausania F, Ricci P, Indolfi C, Brugada J, Brugada R, Cuda G. Integration of "Omics" Strategies for Biomarkers Discovery and for the Elucidation of Molecular Mechanisms Underlying Brugada Syndrome. Proteomics Clin Appl 2018; 12:e1800065. [PMID: 29956481 DOI: 10.1002/prca.201800065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/26/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE The Brugada syndrome (BrS) is a severe inherited cardiac disorder. Given the high genetic and phenotypic heterogeneity of this disease, three different "omics" approaches are integrated in a synergic way to elucidate the molecular mechanisms underlying the pathophysiology of BrS as well as for identifying reliable diagnostic/prognostic markers. EXPERIMENTAL DESIGN The profiling of plasma Proteome and MiRNome is perfomed in a cohort of Brugada patients that were preliminary subjected to genomic analysis to assess a peculiar gene mutation profile. RESULTS The integrated analysis of "omics" data unveiled a cooperative activity of mutated genes, deregulated miRNAs and proteins in orchestrating transcriptional and post-translational events that are critical determining factors for the development of the Brugada pattern. CONCLUSIONS AND CLINICAL RELEVANCE This study provides the basis to shed light on the specific molecular fingerprints underlying BrS development and to gain further insights on the pathogenesis of this life-threatening cardiac disease.
Collapse
Affiliation(s)
- Domenica Scumaci
- Laboratory of Proteomics, Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Antonio Oliva
- Fondazione Policlinico A. Gemelli IRCCS, Roma, Università Cattolica del Sacro Cuore, Large Francesco Vito 1, 00168, Rome, Italy
| | - Antonio Concolino
- Laboratory of Proteomics, Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Antonio Curcio
- Division of Cardiology, Department of Medical and Surgical Science, University "Magna Graecia" of Catanzaro, 88100, Catanzaro, Italy
| | - Claudia Vincenza Fiumara
- Laboratory of Proteomics, Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Laura Tammè
- Laboratory of Proteomics, Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Oscar Campuzano
- Cardiovascular Genetics Center, Gencardio Institut d'Investigacions Biomèdiques de Girona,, 17290, Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) 17007, Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004, Girona, Spain
| | - Vincenzo L Pascali
- Fondazione Policlinico A. Gemelli IRCCS, Roma, Università Cattolica del Sacro Cuore, Large Francesco Vito 1, 00168, Rome, Italy
| | - Monica Coll
- Cardiovascular Genetics Center, Gencardio Institut d'Investigacions Biomèdiques de Girona,, 17290, Girona, Spain
| | - Anna Iglesias
- Cardiovascular Genetics Center, Gencardio Institut d'Investigacions Biomèdiques de Girona,, 17290, Girona, Spain
| | - Paola Berne
- Unità Operativa Complessa di Cardiologia Ospedale "San Francesco", 08100, Nuoro, Italy
| | - Gavino Casu
- Unità Operativa Complessa di Cardiologia Ospedale "San Francesco", 08100, Nuoro, Italy
| | - Erika Olivo
- Laboratory of Proteomics, Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Francesco Ausania
- Fondazione Policlinico A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Roma
| | - Pietrantonio Ricci
- Department of Medical Sciences, School of Medicine, University of Girona, 17004, Girona, Spain
- Institute of Legal Medicine, University "Magna Graecia" of Catanzaro, 88100, Catanzaro, Italy
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Science, University "Magna Graecia" of Catanzaro, 88100, Catanzaro, Italy
| | - Josep Brugada
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) 17007, Girona, Spain
- Arrhythmia's Unit, Hospital Clinic, 08036, Barcelona, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Center, Gencardio Institut d'Investigacions Biomèdiques de Girona,, 17290, Girona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV) 17007, Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17004, Girona, Spain
- Cardiology Service, Hospital Josep Trueta, 17007, Girona, Spain
| | - Giovanni Cuda
- Laboratory of Proteomics, Research Center of Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| |
Collapse
|
9
|
Ke M, Liu J, Chen W, Chen L, Gao W, Qin Y, He A, Chu B, Tang J, Xu R, Deng Y, Tian R. Integrated and Quantitative Proteomic Approach for Charting Temporal and Endogenous Protein Complexes. Anal Chem 2018; 90:12574-12583. [PMID: 30280895 DOI: 10.1021/acs.analchem.8b02667] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | - Jun Tang
- Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Shenzhen 518020, China
| | - Ruilian Xu
- Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, Shenzhen 518020, China
| | - Yi Deng
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen 518055, China
| | - Ruijun Tian
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen 518055, China
| |
Collapse
|
10
|
van Hasselt JGC, Iyengar R. Systems Pharmacology: Defining the Interactions of Drug Combinations. Annu Rev Pharmacol Toxicol 2018; 59:21-40. [PMID: 30260737 DOI: 10.1146/annurev-pharmtox-010818-021511] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The majority of diseases are associated with alterations in multiple molecular pathways and complex interactions at the cellular and organ levels. Single-target monotherapies therefore have intrinsic limitations with respect to their maximum therapeutic benefits. The potential of combination drug therapies has received interest for the treatment of many diseases and is well established in some areas, such as oncology. Combination drug treatments may allow us to identify synergistic drug effects, reduce adverse drug reactions, and address variability in disease characteristics between patients. Identification of combination therapies remains challenging. We discuss current state-of-the-art systems pharmacology approaches to enable rational identification of combination therapies. These approaches, which include characterization of mechanisms of disease and drug action at a systems level, can enable understanding of drug interactions at the molecular, cellular, physiological, and organismal levels. Such multiscale understanding can enable precision medicine by promoting the rational development of combination therapy at the level of individual patients for many diseases.
Collapse
Affiliation(s)
- J G Coen van Hasselt
- Department of Pharmacological Sciences, Systems Biology Center, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; .,Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, 2333 Leiden, Netherlands;
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Systems Biology Center, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
11
|
O'Mara TA, Glubb DM, Amant F, Annibali D, Ashton K, Attia J, Auer PL, Beckmann MW, Black A, Bolla MK, Brauch H, Brenner H, Brinton L, Buchanan DD, Burwinkel B, Chang-Claude J, Chanock SJ, Chen C, Chen MM, Cheng THT, Clarke CL, Clendenning M, Cook LS, Couch FJ, Cox A, Crous-Bous M, Czene K, Day F, Dennis J, Depreeuw J, Doherty JA, Dörk T, Dowdy SC, Dürst M, Ekici AB, Fasching PA, Fridley BL, Friedenreich CM, Fritschi L, Fung J, García-Closas M, Gaudet MM, Giles GG, Goode EL, Gorman M, Haiman CA, Hall P, Hankison SE, Healey CS, Hein A, Hillemanns P, Hodgson S, Hoivik EA, Holliday EG, Hopper JL, Hunter DJ, Jones A, Krakstad C, Kristensen VN, Lambrechts D, Marchand LL, Liang X, Lindblom A, Lissowska J, Long J, Lu L, Magliocco AM, Martin L, McEvoy M, Meindl A, Michailidou K, Milne RL, Mints M, Montgomery GW, Nassir R, Olsson H, Orlow I, Otton G, Palles C, Perry JRB, Peto J, Pooler L, Prescott J, Proietto T, Rebbeck TR, Risch HA, Rogers PAW, Rübner M, Runnebaum I, Sacerdote C, Sarto GE, Schumacher F, Scott RJ, Setiawan VW, Shah M, Sheng X, Shu XO, Southey MC, Swerdlow AJ, Tham E, et alO'Mara TA, Glubb DM, Amant F, Annibali D, Ashton K, Attia J, Auer PL, Beckmann MW, Black A, Bolla MK, Brauch H, Brenner H, Brinton L, Buchanan DD, Burwinkel B, Chang-Claude J, Chanock SJ, Chen C, Chen MM, Cheng THT, Clarke CL, Clendenning M, Cook LS, Couch FJ, Cox A, Crous-Bous M, Czene K, Day F, Dennis J, Depreeuw J, Doherty JA, Dörk T, Dowdy SC, Dürst M, Ekici AB, Fasching PA, Fridley BL, Friedenreich CM, Fritschi L, Fung J, García-Closas M, Gaudet MM, Giles GG, Goode EL, Gorman M, Haiman CA, Hall P, Hankison SE, Healey CS, Hein A, Hillemanns P, Hodgson S, Hoivik EA, Holliday EG, Hopper JL, Hunter DJ, Jones A, Krakstad C, Kristensen VN, Lambrechts D, Marchand LL, Liang X, Lindblom A, Lissowska J, Long J, Lu L, Magliocco AM, Martin L, McEvoy M, Meindl A, Michailidou K, Milne RL, Mints M, Montgomery GW, Nassir R, Olsson H, Orlow I, Otton G, Palles C, Perry JRB, Peto J, Pooler L, Prescott J, Proietto T, Rebbeck TR, Risch HA, Rogers PAW, Rübner M, Runnebaum I, Sacerdote C, Sarto GE, Schumacher F, Scott RJ, Setiawan VW, Shah M, Sheng X, Shu XO, Southey MC, Swerdlow AJ, Tham E, Trovik J, Turman C, Tyrer JP, Vachon C, VanDen Berg D, Vanderstichele A, Wang Z, Webb PM, Wentzensen N, Werner HMJ, Winham SJ, Wolk A, Xia L, Xiang YB, Yang HP, Yu H, Zheng W, Pharoah PDP, Dunning AM, Kraft P, De Vivo I, Tomlinson I, Easton DF, Spurdle AB, Thompson DJ. Identification of nine new susceptibility loci for endometrial cancer. Nat Commun 2018; 9:3166. [PMID: 30093612 PMCID: PMC6085317 DOI: 10.1038/s41467-018-05427-7] [Show More Authors] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 07/02/2018] [Indexed: 01/01/2023] Open
Abstract
Endometrial cancer is the most commonly diagnosed cancer of the female reproductive tract in developed countries. Through genome-wide association studies (GWAS), we have previously identified eight risk loci for endometrial cancer. Here, we present an expanded meta-analysis of 12,906 endometrial cancer cases and 108,979 controls (including new genotype data for 5624 cases) and identify nine novel genome-wide significant loci, including a locus on 12q24.12 previously identified by meta-GWAS of endometrial and colorectal cancer. At five loci, expression quantitative trait locus (eQTL) analyses identify candidate causal genes; risk alleles at two of these loci associate with decreased expression of genes, which encode negative regulators of oncogenic signal transduction proteins (SH2B3 (12q24.12) and NF1 (17q11.2)). In summary, this study has doubled the number of known endometrial cancer risk loci and revealed candidate causal genes for future study.
Collapse
Affiliation(s)
- Tracy A O'Mara
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006, QLD, Australia.
| | - Dylan M Glubb
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006, QLD, Australia
| | - Frederic Amant
- Department of Obstetrics and Gynecology, University Hospitals KU Leuven, University of Leuven, Division of Gynecologic Oncology, Leuven, 3000, Belgium
| | - Daniela Annibali
- Department of Obstetrics and Gynecology, University Hospitals KU Leuven, University of Leuven, Division of Gynecologic Oncology, Leuven, 3000, Belgium
| | - Katie Ashton
- John Hunter Hospital, Hunter Medical Research Institute, Newcastle, 2305, NSW, Australia
- University of Newcastle, Centre for Information Based Medicine, Callaghan, 2308, NSW, Australia
- University of Newcastle, Discipline of Medical Genetics, School of Biomedical Sciences and Pharmacy, Faculty of Health, Callaghan, 2308, NSW, Australia
| | - John Attia
- John Hunter Hospital, Hunter Medical Research Institute, Newcastle, 2305, NSW, Australia
- University of Newcastle, Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, Callaghan, 2308, NSW, Australia
| | - Paul L Auer
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
- University of Wisconsin-Milwaukee, Zilber School of Public Health, Milwaukee, 53205, WI, USA
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Comprehensive Cancer Center ER-EMN, Erlangen, 91054, Germany
| | - Amanda Black
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, 20892, MD, USA
| | - Manjeet K Bolla
- Department of Public Health and Primary Care, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
| | - Hiltrud Brauch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, 70376, Germany
- University of Tübingen, Tübingen, 72074, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, 69120, Germany
| | - Hermann Brenner
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, 69120, Germany
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, 69120, Germany
| | - Louise Brinton
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, 20892, MD, USA
| | - Daniel D Buchanan
- Department of Clinical Pathology, The University of Melbourne, Melbourne, 3010, VIC, Australia
- The University of Melbourne, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, Melbourne, 3010, VIC, Australia
- Genetic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, 3010, VIC, Australia
- Victorian Comprehensive Cancer Centre, University of Melbourne Centre for Cancer Research, Parkville, 3010, VIC, Australia
| | - Barbara Burwinkel
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, 69120, Germany
- Molecular Epidemiology Group, C080, German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, 69120, Germany
- University Medical Center Hamburg-Eppendorf, Cancer Epidemiology, University Cancer Center Hamburg (UCCH), Hamburg, 20246, Germany
| | - Stephen J Chanock
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, 20892, MD, USA
| | - Chu Chen
- Epidemiology Program, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Maxine M Chen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Timothy H T Cheng
- University of Oxford, Wellcome Trust Centre for Human Genetics and Oxford NIHR Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Christine L Clarke
- University of Sydney, Westmead Institute for Medical Research, Sydney, 2145, NSW, Australia
| | - Mark Clendenning
- Department of Clinical Pathology, The University of Melbourne, Melbourne, 3010, VIC, Australia
- Victorian Comprehensive Cancer Centre, University of Melbourne Centre for Cancer Research, Parkville, 3010, VIC, Australia
| | - Linda S Cook
- University of New Mexico, University of New Mexico Health Sciences Center, Albuquerque, 87131, NM, USA
- Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, T2N 4N2, AB, Canada
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Angela Cox
- Department of Oncology and Metabolism, University of Sheffield, Sheffield Institute for Nucleic Acids (SInFoNiA), Sheffield, S10 2TN, UK
| | - Marta Crous-Bous
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
- Department of Medicine, Harvard Medical School, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, 02115, MA, USA
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 171 65, Sweden
| | - Felix Day
- University of Cambridge, MRC Epidemiology Unit, School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Joe Dennis
- Department of Public Health and Primary Care, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
| | - Jeroen Depreeuw
- Department of Obstetrics and Gynecology, University Hospitals KU Leuven, University of Leuven, Division of Gynecologic Oncology, Leuven, 3000, Belgium
- VIB, Vesalius Research Center, Leuven, 3000, Belgium
- Department of Human Genetics, University of Leuven, Laboratory for Translational Genetics, Leuven, 3000, Belgium
| | - Jennifer Anne Doherty
- Cancer Research Huntsman Cancer Institute Department of Population Health Sciences, University of Utah, Salt Lake City, 84112, UT, USA
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, 30625, Germany
| | - Sean C Dowdy
- Department of Obstetrics and Gynecology, Mayo Clinic, Division of Gynecologic Oncology, Rochester, 55905, MN, USA
| | - Matthias Dürst
- Department of Gynaecology, Jena University Hospital - Friedrich Schiller University, Jena, 07743, Germany
| | - Arif B Ekici
- Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Institute of Human Genetics, University Hospital Erlangen, Erlangen, 91054, Germany
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Comprehensive Cancer Center ER-EMN, Erlangen, 91054, Germany
- Department of Medicine, University of California at Los Angeles, David Geffen School of Medicine, Division of Hematology and Oncology, Los Angeles, 90095, CA, USA
| | - Brooke L Fridley
- Department of Biostatistics, Kansas University Medical Center, Kansas City, 66160, KS, USA
| | - Christine M Friedenreich
- Department of Cancer Epidemiology and Prevention Research, Alberta Health Services, Calgary, T2N 4N2, AB, Canada
| | - Lin Fritschi
- Curtin University, School of Public Health, Perth, 6102, WA, Australia
| | - Jenny Fung
- University of Queensland, Institute for Molecular Bioscience, Brisbane, 4072, QLD, Australia
| | - Montserrat García-Closas
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, 20892, MD, USA
- Institute of Cancer Research, Division of Genetics and Epidemiology, London, SM2 5NG, UK
| | - Mia M Gaudet
- American Cancer Society, Epidemiology Research Program, Atlanta, 30303, GA, USA
| | - Graham G Giles
- The University of Melbourne, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, Melbourne, 3010, VIC, Australia
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, 3004, VIC, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, 3004, VIC, Australia
| | - Ellen L Goode
- Department of Health Science Research, Mayo Clinic, Division of Epidemiology, Rochester, 55905, MN, USA
| | - Maggie Gorman
- University of Oxford, Wellcome Trust Centre for Human Genetics and Oxford NIHR Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Christopher A Haiman
- Department of Preventive Medicine, University of Southern California, Keck School of Medicine, Los Angeles, 90033, CA, USA
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 171 65, Sweden
- Department of Oncology, South General Hospital, Stockholm, 118 83, Sweden
| | - Susan E Hankison
- Department of Medicine, Harvard Medical School, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, 02115, MA, USA
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, 01003, MA, USA
| | - Catherine S Healey
- Department of Oncology, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
| | - Alexander Hein
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Comprehensive Cancer Center ER-EMN, Erlangen, 91054, Germany
| | - Peter Hillemanns
- Gynaecology Research Unit, Hannover Medical School, Hannover, 30625, Germany
| | - Shirley Hodgson
- Department of Clinical Genetics, St George's, University of London, London, SW17 0RE, UK
| | - Erling A Hoivik
- Department of Clinical Science, University of Bergen, Centre for Cancer Biomarkers, Bergen, 5020, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, 5021, Norway
| | - Elizabeth G Holliday
- John Hunter Hospital, Hunter Medical Research Institute, Newcastle, 2305, NSW, Australia
- University of Newcastle, Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, Callaghan, 2308, NSW, Australia
| | - John L Hopper
- The University of Melbourne, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, Melbourne, 3010, VIC, Australia
| | - David J Hunter
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Angela Jones
- University of Oxford, Wellcome Trust Centre for Human Genetics and Oxford NIHR Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - Camilla Krakstad
- Department of Clinical Science, University of Bergen, Centre for Cancer Biomarkers, Bergen, 5020, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, 5021, Norway
| | - Vessela N Kristensen
- Department of Cancer Genetics, Oslo University Hospital, Radiumhospitalet, Institute for Cancer Research, Oslo, 0379, Norway
- University of Oslo, Institute of Clinical Medicine, Faculty of Medicine, Oslo, 0450, Norway
- Department of Clinical Molecular Biology, University of Oslo, Oslo University Hospital, Oslo, 0450, Norway
| | - Diether Lambrechts
- Department of Human Genetics, University of Leuven, Laboratory for Translational Genetics, Leuven, 3000, Belgium
- VIB, VIB Center for Cancer Biology, Leuven, 3001, Belgium
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, 96813, HI, USA
| | - Xiaolin Liang
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, 10065, NY, USA
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, M. Sklodowska-Curie Cancer Center-Oncology Institute, Warsaw, 02-034, Poland
| | - Jirong Long
- Department of Medicine, Vanderbilt University School of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Nashville, 37232, TN, USA
| | - Lingeng Lu
- Chronic Disease Epidemiology, Yale School of Public Health, New Haven, 06510, CT, USA
| | - Anthony M Magliocco
- Department of Anatomic Pathology, Moffitt Cancer Center and Research Institute, Tampa, 33612, FL, USA
| | - Lynn Martin
- University of Birmingham, Institute of Cancer and Genomic Sciences, Birmingham, B15 2TT, UK
| | - Mark McEvoy
- University of Newcastle, Centre for Clinical Epidemiology and Biostatistics, School of Medicine and Public Health, Callaghan, 2308, NSW, Australia
| | - Alfons Meindl
- Department of Gynecology and Obstetrics, Ludwig-Maximilians University of Munich, Munich, 80336, Germany
| | - Kyriaki Michailidou
- Department of Public Health and Primary Care, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
- Department of Electron Microscopy/Molecular Pathology, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Roger L Milne
- The University of Melbourne, Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, Melbourne, 3010, VIC, Australia
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, Melbourne, 3004, VIC, Australia
| | - Miriam Mints
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, 171 76, Sweden
| | - Grant W Montgomery
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006, QLD, Australia
- University of Queensland, Institute for Molecular Bioscience, Brisbane, 4072, QLD, Australia
| | - Rami Nassir
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, 95817, CA, USA
| | - Håkan Olsson
- Department of Cancer Epidemiology, Clinical Sciences, Lund University, Lund, 222 42, Sweden
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, 10065, NY, USA
| | - Geoffrey Otton
- University of Newcastle, School of Medicine and Public Health, Callaghan, 2308, NSW, Australia
| | - Claire Palles
- University of Oxford, Wellcome Trust Centre for Human Genetics and Oxford NIHR Biomedical Research Centre, Oxford, OX3 7BN, UK
| | - John R B Perry
- University of Cambridge, MRC Epidemiology Unit, School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Julian Peto
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Loreall Pooler
- Department of Preventive Medicine, University of Southern California, Keck School of Medicine, Los Angeles, 90033, CA, USA
| | - Jennifer Prescott
- Department of Medicine, Harvard Medical School, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, 02115, MA, USA
| | - Tony Proietto
- University of Newcastle, School of Medicine and Public Health, Callaghan, 2308, NSW, Australia
| | - Timothy R Rebbeck
- Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
- Dana-Farber Cancer Institute, Boston, 02115, MA, USA
| | - Harvey A Risch
- Chronic Disease Epidemiology, Yale School of Public Health, New Haven, 06510, CT, USA
| | - Peter A W Rogers
- Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women's Hospital, Gynaecology Research Centre, Parkville, 3052, VIC, Australia
| | - Matthias Rübner
- Department of Gynaecology and Obstetrics, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Erlangen, 91054, Germany
| | - Ingo Runnebaum
- Department of Gynaecology, Jena University Hospital - Friedrich Schiller University, Jena, 07743, Germany
| | - Carlotta Sacerdote
- Center for Cancer Prevention (CPO-Peimonte), Turin, 10126, Italy
- Human Genetics Foundation (HuGeF), Turino, 10126, Italy
| | - Gloria E Sarto
- Department of Obstetrics and Gynecology, University of Wisconsin, School of Medicine and Public Health, Madison, 53715, WI, USA
| | - Fredrick Schumacher
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Rodney J Scott
- John Hunter Hospital, Hunter Medical Research Institute, Newcastle, 2305, NSW, Australia
- University of Newcastle, Centre for Information Based Medicine, Callaghan, 2308, NSW, Australia
- University of Newcastle, Discipline of Medical Genetics, School of Biomedical Sciences and Pharmacy, Faculty of Health, Callaghan, 2308, NSW, Australia
- John Hunter Hospital, Division of Molecular Medicine, Pathology North, Newcastle, 2308, NSW, Australia
| | - V Wendy Setiawan
- Department of Preventive Medicine, University of Southern California, Keck School of Medicine, Los Angeles, 90033, CA, USA
| | - Mitul Shah
- Department of Oncology, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
| | - Xin Sheng
- Department of Preventive Medicine, University of Southern California, Keck School of Medicine, Los Angeles, 90033, CA, USA
| | - Xiao-Ou Shu
- Department of Medicine, Vanderbilt University School of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Nashville, 37232, TN, USA
| | - Melissa C Southey
- Department of Clinical Pathology, The University of Melbourne, Melbourne, 3010, VIC, Australia
- Monash University, Precision Medicine, School of Clinical Sciences at Monash Health, Clayton, 3168, VIC, Australia
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
- Division of Breast Cancer Research, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Emma Tham
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, 171 76, Sweden
- Karolinska Institutet, Clinical Genetics, Stockholm, 171 76, Sweden
| | - Jone Trovik
- Department of Clinical Science, University of Bergen, Centre for Cancer Biomarkers, Bergen, 5020, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, 5021, Norway
| | - Constance Turman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Jonathan P Tyrer
- Department of Oncology, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
| | - Celine Vachon
- Department of Health Sciences Research, Mayo Clinic, Rochester, 55905, MN, USA
| | - David VanDen Berg
- Department of Preventive Medicine, University of Southern California, Keck School of Medicine, Los Angeles, 90033, CA, USA
| | - Adriaan Vanderstichele
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, Division of Gynecologic Oncology, Leuven Cancer Institute, Leuven, 3000, Belgium
| | - Zhaoming Wang
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, 20892, MD, USA
| | - Penelope M Webb
- Department of Population Health, QIMR Berghofer Medical Research Institute, Brisbane, 4006, QLD, Australia
| | - Nicolas Wentzensen
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, 20892, MD, USA
| | - Henrica M J Werner
- Department of Clinical Science, University of Bergen, Centre for Cancer Biomarkers, Bergen, 5020, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, 5021, Norway
| | - Stacey J Winham
- Department of Health Science Research, Mayo Clinic, Division of Biomedical Statistics and Informatics, Rochester, 55905, MN, USA
| | - Alicja Wolk
- Department of Environmental Medicine, Karolinska Institutet, Division of Nutritional Epidemiology, Stockholm, 171 77, Sweden
| | - Lucy Xia
- Department of Preventive Medicine, University of Southern California, Keck School of Medicine, Los Angeles, 90033, CA, USA
| | - Yong-Bing Xiang
- Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, State Key Laboratory of Oncogene and Related Genes, Shanghai, China
| | - Hannah P Yang
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Bethesda, 20892, MD, USA
| | - Herbert Yu
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, 96813, HI, USA
| | - Wei Zheng
- Department of Medicine, Vanderbilt University School of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Nashville, 37232, TN, USA
| | - Paul D P Pharoah
- Department of Public Health and Primary Care, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
- Department of Oncology, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
| | - Alison M Dunning
- Department of Oncology, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
- Department of Medicine, Harvard Medical School, Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, 02115, MA, USA
| | - Ian Tomlinson
- University of Oxford, Wellcome Trust Centre for Human Genetics and Oxford NIHR Biomedical Research Centre, Oxford, OX3 7BN, UK
- University of Birmingham, Institute of Cancer and Genomic Sciences, Birmingham, B15 2TT, UK
| | - Douglas F Easton
- Department of Public Health and Primary Care, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
- Department of Oncology, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK
| | - Amanda B Spurdle
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006, QLD, Australia.
| | - Deborah J Thompson
- Department of Public Health and Primary Care, University of Cambridge, Centre for Cancer Genetic Epidemiology, Cambridge, CB1 8RN, UK.
| |
Collapse
|
12
|
Hu Y, Hu L, Gong D, Lu H, Xuan Y, Wang R, Wu D, Chen D, Zhang K, Gao F, Che L. Genome-wide DNA methylation analysis in jejunum of Sus scrofa with intrauterine growth restriction. Mol Genet Genomics 2018; 293:807-818. [PMID: 29392408 PMCID: PMC6061055 DOI: 10.1007/s00438-018-1422-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 01/27/2018] [Indexed: 01/01/2023]
Abstract
Intrauterine growth restriction (IUGR) may elicit a series of postnatal body developmental and metabolic diseases due to their impaired growth and development in the mammalian embryo/fetus during pregnancy. In the present study, we hypothesized that IUGR may lead to abnormally regulated DNA methylation in the intestine, causing intestinal dysfunctions. We applied reduced representation bisulfite sequencing (RRBS) technology to study the jejunum tissues from four newborn IUGR piglets and their normal body weight (NBW) littermates. The results revealed extensively regional DNA methylation changes between IUGR/NBW pairs from different gilts, affecting dozens of genes. Hiseq-based bisulfite sequencing PCR (Hiseq-BSP) was used for validations of 19 genes with epigenetic abnormality, confirming three genes (AIFM1, MTMR1, and TWIST2) in extra samples. Furthermore, integrated analysis of these 19 genes with proteome data indicated that there were three main genes (BCAP31, IRAK1, and AIFM1) interacting with important immunity- or metabolism-related proteins, which could explain the potential intestinal dysfunctions of IUGR piglets. We conclude that IUGR can lead to disparate DNA methylation in the intestine and these changes may affect several important biological processes such as cell apoptosis, cell differentiation, and immunity, which provides more clues linking IUGR and its long-term complications.
Collapse
Affiliation(s)
- Yue Hu
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China
| | - Liang Hu
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - Desheng Gong
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China
| | - Hanlin Lu
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China
| | - Yue Xuan
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - Ru Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - De Wu
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - Keying Zhang
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China
| | - Fei Gao
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China.
| | - Lianqiang Che
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, 625014, Sichuan, China.
| |
Collapse
|
13
|
Rabalski AJ, Gyenis L, Litchfield DW. Molecular Pathways: Emergence of Protein Kinase CK2 (CSNK2) as a Potential Target to Inhibit Survival and DNA Damage Response and Repair Pathways in Cancer Cells. Clin Cancer Res 2018; 22:2840-7. [PMID: 27306791 DOI: 10.1158/1078-0432.ccr-15-1314] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/04/2016] [Indexed: 11/16/2022]
Abstract
Protein kinase CK2 (designated CSNK2) is a constitutively active protein kinase with a vast repertoire of putative substrates that has been implicated in several human cancers, including cancer of the breast, lung, colon, and prostate, as well as hematologic malignancies. On the basis of these observations, CSNK2 has emerged as a candidate for targeted therapy, with two CSNK2 inhibitors in ongoing clinical trials. CX-4945 is a bioavailable small-molecule ATP-competitive inhibitor targeting its active site, and CIGB-300 is a cell-permeable cyclic peptide that prevents phosphorylation of the E7 protein of HPV16 by CSNK2. In preclinical models, either of these inhibitors exhibit antitumor efficacy. Furthermore, in combinations with chemotherapeutics such as cisplatin or gemcitabine, either CX-4945 or CIGB-300 promote synergistic induction of apoptosis. While CSNK2 is a regulatory participant in many processes related to cancer, its potential to modulate caspase action may be particularly pertinent to its emergence as a therapeutic target. Because the substrate recognition motifs for CSNK2 and caspases are remarkably similar, CSNK2 can block the cleavage of many caspase substrates through the phosphorylation of sites adjacent to cleavage sites. Phosphoproteomic strategies have also revealed previously underappreciated roles for CSNK2 in the phosphorylation of several key constituents of DNA damage and DNA repair pathways. Going forward, applications of proteomic strategies to interrogate responses to CSNK2 inhibitors are expected to reveal signatures for CSNK2 inhibition and molecular insights to guide new strategies to interfere with its potential to inhibit caspase action or enhance the susceptibility of cancer cells to DNA damage. Clin Cancer Res; 22(12); 2840-7. ©2016 AACR.
Collapse
Affiliation(s)
- Adam J Rabalski
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Laszlo Gyenis
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - David W Litchfield
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada. Department of Oncology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
14
|
Lau ATY, Xu YM. Regulation of human mitogen-activated protein kinase 15 (extracellular signal-regulated kinase 7/8) and its functions: A recent update. J Cell Physiol 2018; 234:75-88. [PMID: 30070699 DOI: 10.1002/jcp.27053] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/25/2018] [Indexed: 02/05/2023]
Abstract
Mitogen-activated protein kinase 15 (MAPK15), originally also known as extracellular signal-regulated kinase 7/8, is the most recently identified atypical MAPK and the least studied so far. Examinations of the role of MAPK15 in various cell lines and model systems indicate that MAPK15 participates in a variety of cellular activities such as promoting cell proliferation, cell transformation, and apoptosis; stimulating autophagy; regulating cell division, ciliogenesis, and protein secretion; and maintaining genome stability. As multiple roles of MAPK15 were observed among these studies, therefore, it remains unclear whether MAPK15 acts as a proto-oncogene or tumor suppressor. Here, the recent literature on human MAPK15 and the resulting functions will be discussed.
Collapse
Affiliation(s)
- Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| |
Collapse
|
15
|
Meshalkina DA, Shevtsov MA, Dobrodumov AV, Komarova EY, Voronkina IV, Lazarev VF, Margulis BA, Guzhova IV. Knock-down of Hdj2/DNAJA1 co-chaperone results in an unexpected burst of tumorigenicity of C6 glioblastoma cells. Oncotarget 2017; 7:22050-63. [PMID: 26959111 PMCID: PMC5008343 DOI: 10.18632/oncotarget.7872] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/20/2016] [Indexed: 01/04/2023] Open
Abstract
The chaperone system based on Hsp70 and proteins of the DnaJ family is known to protect tumor cells from a variety of cytotoxic factors, including anti-tumor therapy. To analyze whether this also functions in a highly malignant brain tumor, we knocked down the expression of Hsp70 (HSPA1A) and its two most abundant co-chaperones, Hdj1 (DNAJB1) and Hdj2 (DNAJA1) in a C6 rat glioblastoma cell line. As expected, tumor depletion of Hsp70 caused a substantial reduction in its growth rate and increased the survival of tumor-bearing animals, whereas the reduction of Hdj1 expression had no effect. Unexpectedly, a reduction in the expression of Hdj2 led to the enhanced aggressiveness of the C6 tumor, demonstrated by its rapid growth, metastasis formation and a 1.5-fold reduction in the lifespan of tumor-bearing animals. The in vitro reduction of Hdj2 expression reduced spheroid density and simultaneously enhanced the migration and invasion of C6 cells. At the molecular level, a knock-down of Hdj2 led to the relocation of N-cadherin and the enhanced activity of metalloproteinases 1, 2, 8 and 9, which are markers of highly malignant cancer cells. The changes in the actin cytoskeleton in Hdj2-depleted cells indicate that the protein is also important for prevention of the amoeboid-like transition of tumor cells. The results of this study uncover a completely new role for the Hdj2 co-chaperone in tumorigenicity and suggest that the protein is a potential drug target.
Collapse
Affiliation(s)
- Darya A Meshalkina
- Institute of Cytology of Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Maxim A Shevtsov
- Institute of Cytology of Russian Academy of Sciences, St. Petersburg 194064, Russia.,First I.P. Pavlov State Medical University of St. Petersburg, St. Petersburg 197022, Russia
| | - Anatoliy V Dobrodumov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, St. Petersburg 199004, Russia
| | - Elena Y Komarova
- Institute of Cytology of Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Irina V Voronkina
- Institute of Cytology of Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Vladimir F Lazarev
- Institute of Cytology of Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Boris A Margulis
- Institute of Cytology of Russian Academy of Sciences, St. Petersburg 194064, Russia
| | - Irina V Guzhova
- Institute of Cytology of Russian Academy of Sciences, St. Petersburg 194064, Russia
| |
Collapse
|
16
|
Ma S, Sun J, Guo Y, Zhang P, Liu Y, Zheng D, Shi J. Combination of AAV-TRAIL with miR-221-Zip Therapeutic Strategy Overcomes the Resistance to TRAIL Induced Apoptosis in Liver Cancer. Am J Cancer Res 2017; 7:3228-3242. [PMID: 28900506 PMCID: PMC5595128 DOI: 10.7150/thno.19893] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/29/2017] [Indexed: 02/06/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) possesses the capacity to induce apoptosis in a wide variety of tumor cells without affecting most normal cells. However, it has now emerged that many primary cancer cells are resistant to TRAIL monotherapy. Overcoming the intrinsic or acquired TRAIL resistance is desirable for TRAIL-mediated cancer therapy. In this study, we found that the miR-221/222 cluster was up-regulated in TRAIL-resistant liver cancer cells. Specific inhibitors of miR-221 and/or miR-222, called sponge, TuD and miR-Zip were constructed, and their ability to overcome TRAIL resistance was compared. Among them, AAV-mediated gene therapy using co-expression of TRAIL with miR-221-Zip showed the most synergistic activity in the induction of apoptosis in vitro. In vivo treatment of nude mice bearing human TRAIL-resistant liver cancer xenografts with AAV-TRAIL-miR-221-Zip also led to growth inhibition. This sensitizing effect of miR-221-Zip was associated with increased expression of PTEN, the miR-221 target, as well as with decreasing levels of Survivin. Moreover, miR-221 expression was concomitant with promotion of Survivin expression and suppression of PTEN expression. TRAIL sensitivity of cancer cells isolated from liver cancer tissues or from patients was significantly correlated with miR-221 expression. And miR-221 blood expression levels in liver cancer patients were correlated with TRAIL sensitivity, thus it had the potential to be a predictor of TRAIL sensitivity in liver cancer. These data suggested the potential of combining AAV-TRAIL with miR-221-Zip as a therapeutic intervention for liver cancer.
Collapse
|
17
|
Xie R, Gao CC, Yang XZ, Wu SN, Wang HG, Zhang JL, Yan W, Ma TH. Combining TRAIL and liquiritin exerts synergistic effects against human gastric cancer cells and xenograft in nude mice through potentiating apoptosis and ROS generation. Biomed Pharmacother 2017; 93:948-960. [PMID: 28715876 DOI: 10.1016/j.biopha.2017.06.095] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is one of the most factors, leading to cancer-related death worldwide. However, the therapies to prevent gastric cancer are still limited and the emergence of drug resistance leads to development of new anti-cancer drugs and combinational chemotherapy regimens. Our study was aimed to explore the anti-gastric cancer effects of liquiritin (LIQ), a major constituent of Glycyrrhiza Radix, which possesses a variety of pharmacological activities. The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) preferentially inhibited tumor cells over other normal cells, when used in alone or in combination. The results indicated that LIQ, when applied in single, was moderately effective to suppress proliferation, and migration, as well as to induce apoptosis and reactive oxygen species (ROS) generation of human gastric cancer cell lines, AGS and SNU-216, which are TRAIL-resistant. Significantly, when used in combination, the two drugs functioned synergistically to impede the progression and growth of human gastric cancer cells in vitro and gastric cancer cell xenograft nude mice in vivo. Both intrinsic and extrinsic apoptosis were induced by the two in combination via activating Caspases. And c-Jun N-terminal kinase (JNK) activity was dramatically induced by TRAIL/LIQ. Importantly, TRAIL/LIQ-triggered apoptosis and JNK were dependent on ROS production. The data indicated that application of TRAIL/LIQ in combination had a potential value for clinical use to synergistically prevent human gastric cancer development.
Collapse
Affiliation(s)
- Rui Xie
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Cheng-Cheng Gao
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Xiao-Zhong Yang
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Shang-Nong Wu
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Hong-Gang Wang
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Jia-Ling Zhang
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Wei Yan
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, PR China
| | - Tian-Heng Ma
- Department of Gastroenterology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, PR China.
| |
Collapse
|
18
|
Abu-Jamous B, Buffa FM, Harris AL, Nandi AK. In vitro downregulated hypoxia transcriptome is associated with poor prognosis in breast cancer. Mol Cancer 2017; 16:105. [PMID: 28619028 PMCID: PMC5472949 DOI: 10.1186/s12943-017-0673-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 06/02/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Hypoxia is a characteristic of breast tumours indicating poor prognosis. Based on the assumption that those genes which are up-regulated under hypoxia in cell-lines are expected to be predictors of poor prognosis in clinical data, many signatures of poor prognosis were identified. However, it was observed that cell line data do not always concur with clinical data, and therefore conclusions from cell line analysis should be considered with caution. As many transcriptomic cell-line datasets from hypoxia related contexts are available, integrative approaches which investigate these datasets collectively, while not ignoring clinical data, are required. RESULTS We analyse sixteen heterogeneous breast cancer cell-line transcriptomic datasets in hypoxia-related conditions collectively by employing the unique capabilities of the method, UNCLES, which integrates clustering results from multiple datasets and can address questions that cannot be answered by existing methods. This has been demonstrated by comparison with the state-of-the-art iCluster method. From this collection of genome-wide datasets include 15,588 genes, UNCLES identified a relatively high number of genes (>1000 overall) which are consistently co-regulated over all of the datasets, and some of which are still poorly understood and represent new potential HIF targets, such as RSBN1 and KIAA0195. Two main, anti-correlated, clusters were identified; the first is enriched with MYC targets participating in growth and proliferation, while the other is enriched with HIF targets directly participating in the hypoxia response. Surprisingly, in six clinical datasets, some sub-clusters of growth genes are found consistently positively correlated with hypoxia response genes, unlike the observation in cell lines. Moreover, the ability to predict bad prognosis by a combined signature of one sub-cluster of growth genes and one sub-cluster of hypoxia-induced genes appears to be comparable and perhaps greater than that of known hypoxia signatures. CONCLUSIONS We present a clustering approach suitable to integrate data from diverse experimental set-ups. Its application to breast cancer cell line datasets reveals new hypoxia-regulated signatures of genes which behave differently when in vitro (cell-line) data is compared with in vivo (clinical) data, and are of a prognostic value comparable or exceeding the state-of-the-art hypoxia signatures.
Collapse
Affiliation(s)
- Basel Abu-Jamous
- Department of Electronic and Computer Engineering, Brunel University London, Uxbridge, Middlesex, UB8 3PH UK
- Department of Plant Sciences, University of Oxford, Oxford, OX1 3RB UK
| | - Francesca M. Buffa
- Cancer Research UK, Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS UK
| | - Adrian L. Harris
- Cancer Research UK, Department of Oncology, Weatherall Institute of Molecular Medicine, Oxford, OX3 9DS UK
| | - Asoke K. Nandi
- Department of Electronic and Computer Engineering, Brunel University London, Uxbridge, Middlesex, UB8 3PH UK
- The Key Laboratory of Embedded Systems and Service Computing, College of Electronic and Information Engineering, Tongji University, Shanghai, Peoples, Republic of China
| |
Collapse
|
19
|
Luan L, Shi J, Yu Z, Andl T. The major miR-31 target genes STK40 and LATS2 and their implications in the regulation of keratinocyte growth and hair differentiation. Exp Dermatol 2017; 26:497-504. [PMID: 28419554 DOI: 10.1111/exd.13355] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2017] [Indexed: 02/06/2023]
Abstract
Emerging evidence indicates that even subtle changes in the expression of key genes of signalling pathways can have profound effects. MicroRNAs (miRNAs) are masters of subtlety and generally have only mild effects on their target genes. The microRNA miR-31 is one of the major microRNAs in many cutaneous conditions associated with activated keratinocytes, such as the hyperproliferative diseases psoriasis, non-melanoma skin cancer and hair follicle growth. miR-31 is a marker of the hair growth phase, and in our miR-31 transgenic mouse model it impairs the function of keratinocytes. This leads to aberrant proliferation, apoptosis, and differentiation that results in altered hair growth, while the loss of miR-31 leads to increased hair growth. Through in vitro and in vivo studies, we have defined a set of conserved miR-31 target genes, including LATS2 and STK40, which serve as new players in the regulation of keratinocyte growth and hair follicle biology. LATS2 can regulate growth of keratinocytes and we have identified a function of STK40 that can promote the expression of key hair follicle programme regulators such as HR, DLX3 and HOXC13.
Collapse
Affiliation(s)
- Liming Luan
- Division of Dermatology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jianyun Shi
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zhengquan Yu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Thomas Andl
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
20
|
Portes LL, Aguirre LA. Matrix formulation and singular-value decomposition algorithm for structured varimax rotation in multivariate singular spectrum analysis. Phys Rev E 2016; 93:052216. [PMID: 27300889 DOI: 10.1103/physreve.93.052216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Indexed: 06/06/2023]
Abstract
Groth and Ghil [Phys. Rev. E 84, 036206 (2011)PLEEE81539-375510.1103/PhysRevE.84.036206] developed a modified varimax rotation aimed at enhancing the ability of the multivariate singular spectrum analysis (M-SSA) to characterize phase synchronization in systems of coupled chaotic oscillators. Due to the special structure of the M-SSA eigenvectors, the modification proposed by Groth and Ghil imposes a constraint in the rotation of blocks of components associated with the different subsystems. Accordingly, here we call it a structured varimax rotation (SVR). The SVR was presented as successive pairwise rotations of the eigenvectors. The aim of this paper is threefold. First, we develop a closed matrix formulation for the entire family of structured orthomax rotation criteria, for which the SVR is a special case. Second, this matrix approach is used to enable the use of known singular value algorithms for fast computation, allowing a simultaneous rotation of the M-SSA eigenvectors (a Python code is provided in the Appendix). This could be critical in the characterization of phase synchronization phenomena in large real systems of coupled oscillators. Furthermore, the closed algebraic matrix formulation could be used in theoretical studies of the (modified) M-SSA approach. Third, we illustrate the use of the proposed singular value algorithm for the SVR in the context of the two benchmark examples of Groth and Ghil: the Rössler system in the chaotic (i) phase-coherent and (ii) funnel regimes. Comparison with the results obtained with Kaiser's original (unstructured) varimax rotation (UVR) reveals that both SVR and UVR give the same result for the phase-coherent scenario, but for the more complex behavior (ii) only the SVR improves on the M-SSA.
Collapse
Affiliation(s)
- Leonardo L Portes
- Departamento de Engenharia Eletrônica, Universidade Federal de Minas Gerais-Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte MG, Brazil
| | - Luis A Aguirre
- Departamento de Engenharia Eletrônica, Universidade Federal de Minas Gerais-Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte MG, Brazil
| |
Collapse
|
21
|
Federspiel JD, Codreanu SG, Palubinsky AM, Winland AJ, Betanzos CM, McLaughlin B, Liebler DC. Assembly Dynamics and Stoichiometry of the Apoptosis Signal-regulating Kinase (ASK) Signalosome in Response to Electrophile Stress. Mol Cell Proteomics 2016; 15:1947-61. [PMID: 27006476 DOI: 10.1074/mcp.m115.057364] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Indexed: 01/29/2023] Open
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) is a key sensor kinase in the mitogen-activated protein kinase pathway that transduces cellular responses to oxidants and electrophiles. ASK1 is regulated by a large, dynamic multiprotein signalosome complex, potentially including over 90 reported ASK1-interacting proteins. We employed both shotgun and targeted mass spectrometry assays to catalogue the ASK1 protein-protein interactions in HEK-293 cells treated with the prototypical lipid electrophile 4-hydroxy-2-nonenal (HNE). Using both epitope-tagged overexpression and endogenous expression cell systems, we verified most of the previously reported ASK1 protein-protein interactions and identified 14 proteins that exhibited dynamic shifts in association with ASK1 in response to HNE stress. We used precise stable isotope dilution assays to quantify protein stoichiometry in the ASK signalosome complex and identified ASK2 at a 1:1 stoichiometric ratio with ASK1 and 14-3-3 proteins (YWHAQ, YWHAB, YWHAH, and YWHAE) collectively at a 0.5:1 ratio with ASK1 as the main components. Several other proteins, including ASK3, PARK7, PRDX1, and USP9X were detected with stoichiometries of 0.1:1 or less. These data support an ASK signalosome comprising a multimeric core complex of ASK1, ASK2, and 14-3-3 proteins, which dynamically engages other binding partners needed to mediate diverse stress-response signaling events. This study further demonstrates the value of combining global and targeted MS approaches to interrogate multiprotein complex composition and dynamics.
Collapse
Affiliation(s)
- Joel D Federspiel
- From the ‡Department of Biochemistry, Vanderbilt University School of Medicine
| | - Simona G Codreanu
- From the ‡Department of Biochemistry, Vanderbilt University School of Medicine
| | - Amy M Palubinsky
- §Neuroscience Graduate Program, Vanderbilt Brain Institute, Vanderbilt University
| | - Ama J Winland
- ¶Department of Neurology, Vanderbilt University, Nashville, Tennessee, 37232
| | | | - BethAnn McLaughlin
- ¶Department of Neurology, Vanderbilt University, Nashville, Tennessee, 37232
| | - Daniel C Liebler
- From the ‡Department of Biochemistry, Vanderbilt University School of Medicine;
| |
Collapse
|
22
|
Campbell J, Ryan CJ, Brough R, Bajrami I, Pemberton HN, Chong IY, Costa-Cabral S, Frankum J, Gulati A, Holme H, Miller R, Postel-Vinay S, Rafiq R, Wei W, Williamson CT, Quigley DA, Tym J, Al-Lazikani B, Fenton T, Natrajan R, Strauss SJ, Ashworth A, Lord CJ. Large-Scale Profiling of Kinase Dependencies in Cancer Cell Lines. Cell Rep 2016; 14:2490-501. [PMID: 26947069 PMCID: PMC4802229 DOI: 10.1016/j.celrep.2016.02.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/07/2015] [Accepted: 02/01/2016] [Indexed: 12/27/2022] Open
Abstract
One approach to identifying cancer-specific vulnerabilities and therapeutic targets is to profile genetic dependencies in cancer cell lines. Here, we describe data from a series of siRNA screens that identify the kinase genetic dependencies in 117 cancer cell lines from ten cancer types. By integrating the siRNA screen data with molecular profiling data, including exome sequencing data, we show how vulnerabilities/genetic dependencies that are associated with mutations in specific cancer driver genes can be identified. By integrating additional data sets into this analysis, including protein-protein interaction data, we also demonstrate that the genetic dependencies associated with many cancer driver genes form dense connections on functional interaction networks. We demonstrate the utility of this resource by using it to predict the drug sensitivity of genetically or histologically defined subsets of tumor cell lines, including an increased sensitivity of osteosarcoma cell lines to FGFR inhibitors and SMAD4 mutant tumor cells to mitotic inhibitors.
Collapse
MESH Headings
- Cell Line, Tumor
- Gene Expression Profiling
- Humans
- Mutation
- Neoplasms/enzymology
- Neoplasms/genetics
- Neoplasms/pathology
- Protein Kinases/chemistry
- Protein Kinases/genetics
- Protein Kinases/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Smad4 Protein/antagonists & inhibitors
- Smad4 Protein/genetics
- Smad4 Protein/metabolism
Collapse
Affiliation(s)
- James Campbell
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Colm J Ryan
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Rachel Brough
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Ilirjana Bajrami
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Helen N Pemberton
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Irene Y Chong
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; Royal Marsden Hospital, London SW3 6JJ, UK
| | - Sara Costa-Cabral
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Jessica Frankum
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Aditi Gulati
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Harriet Holme
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Rowan Miller
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Sophie Postel-Vinay
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK; Gustave Roussy Cancer Campus, 94805 Villejuif, France
| | - Rumana Rafiq
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Wenbin Wei
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Chris T Williamson
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - David A Quigley
- UCSF Helen Diller Family Comprehensive Cancer Centre, San Francisco, CA 94158, USA
| | - Joe Tym
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Bissan Al-Lazikani
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, Sutton SM2 5NG, UK
| | - Timothy Fenton
- UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Rachael Natrajan
- Functional Genomics Laboratory, The Breast Cancer Now Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Sandra J Strauss
- UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alan Ashworth
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK.
| | - Christopher J Lord
- The Breast Cancer Now Research Centre and CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK.
| |
Collapse
|
23
|
Abstract
Over the past decade, rapid advances in genomics, proteomics and functional genomics technologies that enable in-depth interrogation of cancer genomes and proteomes and high-throughput analysis of gene function have enabled characterization of the kinome 'at large' in human cancers, providing crucial insights into how members of the protein kinase superfamily are dysregulated in malignancy, the context-dependent functional role of specific kinases in cancer and how kinome remodelling modulates sensitivity to anticancer drugs. The power of these complementary approaches, and the insights gained from them, form the basis of this Analysis article.
Collapse
Affiliation(s)
- Emmy D G Fleuren
- Department of Medical Oncology, Radboud University Medical Centre, Geert Grooteplein-Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Luxi Zhang
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
- University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Jianmin Wu
- Cancer Division, Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria Street, Sydney, New South Wales 2010, Australia
| | - Roger J Daly
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
24
|
Mutational Analysis of Glycogen Synthase Kinase 3β Protein Kinase Together with Kinome-Wide Binding and Stability Studies Suggests Context-Dependent Recognition of Kinases by the Chaperone Heat Shock Protein 90. Mol Cell Biol 2016; 36:1007-18. [PMID: 26755559 DOI: 10.1128/mcb.01045-15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/05/2016] [Indexed: 11/20/2022] Open
Abstract
The heat shock protein 90 (HSP90) and cell division cycle 37 (CDC37) chaperones are key regulators of protein kinase folding and maturation. Recent evidence suggests that thermodynamic properties of kinases, rather than primary sequences, are recognized by the chaperones. In concordance, we observed a striking difference in HSP90 binding between wild-type (WT) and kinase-dead (KD) glycogen synthase kinase 3β (GSK3β) forms. Using model cell lines stably expressing these two GSK3β forms, we observed no interaction between WT GSK3β and HSP90, in stark contrast to KD GSK3β forming a stable complex with HSP90 at a 1:1 ratio. In a survey of 91 ectopically expressed kinases in DLD-1 cells, we compared two parameters to measure HSP90 dependency: static binding and kinase stability following HSP90 inhibition. We observed no correlation between HSP90 binding and reduced stability of a kinase after pharmacological inhibition of HSP90. We expanded our stability study to >50 endogenous kinases across four cell lines and demonstrated that HSP90 dependency is context dependent. These observations suggest that HSP90 binds to its kinase client in a particular conformation that we hypothesize to be associated with the nucleotide-processing cycle. Lastly, we performed proteomics profiling of kinases and phosphopeptides in DLD-1 cells to globally define the impact of HSP90 inhibition on the kinome.
Collapse
|
25
|
Affiliation(s)
- Nicholas M. Riley
- Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Joshua J. Coon
- Genome Center of Wisconsin, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
26
|
Sample Preparation for Mass Spectrometry Analysis of Protein-Protein Interactions in Cancer Cell Lines and Tissues. Methods Mol Biol 2016; 1458:339-47. [PMID: 27581032 DOI: 10.1007/978-1-4939-3801-8_23] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A precisely controlled network of protein-protein interactions constitutes the basis for functional signaling pathways. This equilibrium is more often than not disrupted in cancer cells, by the aberrant expression or activation of oncogenic proteins. Therefore, the analysis of protein interaction networks in cancer cells has become crucial to expand our comprehension of the molecular underpinnings of tumor formation and progression. This protocol describes a sample preparation method for the analysis of signaling complexes by mass spectrometry (MS), following the affinity purification of a protein of interest from a cancer cell line or a solid tumor. In particular, we provide a spin tip-based protease digestion procedure that offers a more rapid and controlled alternative to other gel-based and gel-free methods. This sample preparation protocol represents a useful strategy to identify protein interactions and to gain insight into the molecular mechanisms that contribute to a given cancer phenotype.
Collapse
|
27
|
Amarante-Mendes GP, Griffith TS. Therapeutic applications of TRAIL receptor agonists in cancer and beyond. Pharmacol Ther 2015; 155:117-31. [PMID: 26343199 DOI: 10.1016/j.pharmthera.2015.09.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
TRAIL/Apo-2L is a member of the TNF superfamily first described as an apoptosis-inducing cytokine in 1995. Similar to TNF and Fas ligand, TRAIL induces apoptosis in caspase-dependent manner following TRAIL death receptor trimerization. Because tumor cells were shown to be particularly sensitive to this cytokine while normal cells/tissues proved to be resistant along with being able to synthesize and release TRAIL, it was rapidly appreciated that TRAIL likely served as one of our major physiologic weapons against cancer. In line with this, a number of research laboratories and pharmaceutical companies have attempted to exploit the ability of TRAIL to kill cancer cells by developing recombinant forms of TRAIL or TRAIL receptor agonists (e.g., receptor-specific mAb) for therapeutic purposes. In this review article we will describe the biochemical pathways used by TRAIL to induce different cell death programs. We will also summarize the clinical trials related to this pathway and discuss possible novel uses of TRAIL-related therapies. In recent years, the physiological importance of TRAIL has expanded beyond being a tumoricidal molecule to one critical for a number of clinical settings - ranging from infectious disease and autoimmunity to cardiovascular anomalies. We will also highlight some of these conditions where modulation of the TRAIL/TRAIL receptor system may be targeted in the future.
Collapse
Affiliation(s)
- Gustavo P Amarante-Mendes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, SP, Brazil; Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia, Brazil.
| | - Thomas S Griffith
- Department of Urology, Masonic Cancer Center, Center for Immunology, University of Minnesota, Minneapolis, MN, USA; Minneapolis VA Health Care System, Minneapolis, MN 55417, USA.
| |
Collapse
|