101
|
Wang W, Zhang Y, Chen R, Tian Z, Zhai Y, Janz S, Gu C, Yang Y. Chromosomal instability and acquired drug resistance in multiple myeloma. Oncotarget 2017; 8:78234-78244. [PMID: 29100463 PMCID: PMC5652852 DOI: 10.18632/oncotarget.20829] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/26/2017] [Indexed: 12/29/2022] Open
Abstract
Chromosomal instability (CIN) is an important hallmark of human cancer. CIN not only contributes to all stages of tumor development (initiation, promotion and progression) but also drives, in large measure, the acquisition of drug resistance by cancer cells. Although CIN is a cornerstone of the complex mutational architecture that underlies neoplastic cell development and tumor heterogeneity and has been tightly associated with treatment responses and survival of cancer patients, it may be one of the least understood features of the malignant phenotype in terms of genetic pathways and molecular mechanisms. Here we review new insights into the type of CIN seen in multiple myeloma (MM), a blood cancer of terminally differentiated, immunoglobulin-producing B-lymphocytes called plasma cells that remains incurable in the great majority of cases. We will consider bona fide myeloma CIN genes, methods for measuring CIN in myeloma cells, and novel approaches to CIN-targeted treatments of patients with myeloma. The new findings generate optimism that enhanced understanding of CIN will lead to the design and testing of new therapeutic strategies to overcome drug resistance in MM in the not-so-distant future.
Collapse
Affiliation(s)
- Wang Wang
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Ruini Chen
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhidan Tian
- Department of Pathology, Nanjing First Hospital, Nanjing, 210006, China
| | - Yongpin Zhai
- Department of Hematology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, China
| | - Siegfried Janz
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, 52242, USA
| | - Chunyan Gu
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye Yang
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China.,School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
102
|
Chan SH, Ngeow J. Germline mutation contribution to chromosomal instability. Endocr Relat Cancer 2017; 24:T33-T46. [PMID: 28808044 DOI: 10.1530/erc-17-0062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/18/2017] [Indexed: 12/29/2022]
Abstract
Genomic instability is a feature of cancer that fuels oncogenesis through increased frequency of genetic disruption, leading to loss of genomic integrity and promoting clonal evolution as well as tumor transformation. A form of genomic instability prevalent across cancer types is chromosomal instability, which involves karyotypic changes including chromosome copy number alterations as well as gross structural abnormalities such as transversions and translocations. Defects in cellular mechanisms that are in place to govern fidelity of chromosomal segregation, DNA repair and ultimately genomic integrity are known to contribute to chromosomal instability. In this review, we discuss the association of germline mutations in these pathways with chromosomal instability in the background of related cancer predisposition syndromes. We will also reflect on the impact of genetic predisposition to clinical management of patients and how we can exploit this vulnerability to promote catastrophic genomic instability as a therapeutic strategy.
Collapse
Affiliation(s)
- Sock Hoai Chan
- Division of Medical OncologyCancer Genetics Service, National Cancer Centre Singapore, Singapore
| | - Joanne Ngeow
- Division of Medical OncologyCancer Genetics Service, National Cancer Centre Singapore, Singapore
- Oncology Academic Clinical ProgramDuke-NUS Medical School Singapore, Singapore
| |
Collapse
|
103
|
Agarwal S, Varma D. Targeting mitotic pathways for endocrine-related cancer therapeutics. Endocr Relat Cancer 2017; 24:T65-T82. [PMID: 28615236 PMCID: PMC5557717 DOI: 10.1530/erc-17-0080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/14/2017] [Indexed: 12/21/2022]
Abstract
A colossal amount of basic research over the past few decades has provided unprecedented insights into the highly complex process of cell division. There is an ever-expanding catalog of proteins that orchestrate, participate and coordinate in the exquisite processes of spindle formation, chromosome dynamics and the formation and regulation of kinetochore microtubule attachments. Use of classical microtubule poisons has still been widely and often successfully used to combat a variety of cancers, but their non-selective interference in other crucial physiologic processes necessitate the identification of novel druggable components specific to the cell cycle/division pathway. Considering cell cycle deregulation, unscheduled proliferation, genomic instability and chromosomal instability as a hallmark of tumor cells, there lies an enormous untapped terrain that needs to be unearthed before a drug can pave its way from bench to bedside. This review attempts to systematically summarize the advances made in this context so far with an emphasis on endocrine-related cancers and the avenues for future progress to target mitotic mechanisms in an effort to combat these dreadful cancers.
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Cell and Molecular BiologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Dileep Varma
- Department of Cell and Molecular BiologyFeinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
104
|
Mackenzie KJ, Carroll P, Martin CA, Murina O, Fluteau A, Simpson DJ, Olova N, Sutcliffe H, Rainger JK, Leitch A, Osborn RT, Wheeler AP, Nowotny M, Gilbert N, Chandra T, Reijns MAM, Jackson AP. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 2017; 548:461-465. [PMID: 28738408 PMCID: PMC5870830 DOI: 10.1038/nature23449] [Citation(s) in RCA: 1129] [Impact Index Per Article: 161.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 07/04/2017] [Indexed: 12/12/2022]
Abstract
DNA is strictly compartmentalized within the nucleus to prevent autoimmunity; despite this, cyclic GMP-AMP synthase (cGAS), a cytosolic sensor of double-stranded DNA, is activated in autoinflammatory disorders and by DNA damage. Precisely how cellular DNA gains access to the cytoplasm remains to be determined. Here, we report that cGAS localizes to micronuclei arising from genome instability in a mouse model of monogenic autoinflammation, after exogenous DNA damage and spontaneously in human cancer cells. Such micronuclei occur after mis-segregation of DNA during cell division and consist of chromatin surrounded by its own nuclear membrane. Breakdown of the micronuclear envelope, a process associated with chromothripsis, leads to rapid accumulation of cGAS, providing a mechanism by which self-DNA becomes exposed to the cytosol. cGAS is activated by chromatin, and consistent with a mitotic origin, micronuclei formation and the proinflammatory response following DNA damage are cell-cycle dependent. By combining live-cell laser microdissection with single cell transcriptomics, we establish that interferon-stimulated gene expression is induced in micronucleated cells. We therefore conclude that micronuclei represent an important source of immunostimulatory DNA. As micronuclei formed from lagging chromosomes also activate this pathway, recognition of micronuclei by cGAS may act as a cell-intrinsic immune surveillance mechanism that detects a range of neoplasia-inducing processes.
Collapse
Affiliation(s)
- Karen J Mackenzie
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Paula Carroll
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Carol-Anne Martin
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Olga Murina
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Adeline Fluteau
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Daniel J Simpson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Nelly Olova
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Hannah Sutcliffe
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Jacqueline K Rainger
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Andrea Leitch
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Ruby T Osborn
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Ann P Wheeler
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Marcin Nowotny
- Laboratory of Protein Structure, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Nick Gilbert
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Tamir Chandra
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Martin A M Reijns
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Andrew P Jackson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
105
|
Zhang P, Li Z, Wang N, Duan G, Wang W, Feng Y, Zhao Y, Wang L, Zhu H, Zhang Q, Liu X, Wu W, Wu Y, Yao W, Wang J, Wu Y, Feng F. Coal tar pitch extract could induce chromosomal instability of human bronchial epithelial cells mediated by spindle checkpoint-related proteins. Oncotarget 2017; 8:56506-56517. [PMID: 28915607 PMCID: PMC5593578 DOI: 10.18632/oncotarget.17025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 03/21/2017] [Indexed: 11/25/2022] Open
Abstract
Coal tar pitch (CTP) is a byproduct of coal tar distillation. The workers working with coal tar or in aluminum smelters, potrooms and carbon plants have the opportunities of exposing to coal tar pitch volatiles. Coal tar pitches from which polycyclic aromatic hydrocarbons (PAHs) originate have been shown to exhibit lung carcinogenicity in humans. Chromosomal instability (CIN) is a mechanism in carcinogenesis, however, whether CIN is involved in coal tar pitch-induced lung cancer remains elusive. In this present study, human bronchial epithelial cells (BEAS-2B) were first exposed to coal tar pitch extracts (CTPE) to induce a malignant transformation model. Then, the occurrence of severe chromosomal changes detected using G band, R band and multiplex fluorescence in situ hybridization (M-FISH) staining were examined. It was shown that more clones of transformed BEAS-2B cells at passage 30 following stimulation with CTPE were formed in the soft agar compared with the vehicle control. Moreover, the expression of the spindle checkpoint-related proteins, mitotic arrest defective 2 (Mad2), budding uninhibited in benzimidazole 1 (Bub1), and anaphase-promoting complex (APC), indicators of abnormal chromosomes and carcinogenesis, reduced in CTPE-treated BEAS-2B cells at Passage 30 compared with the vehicle control using real-time PCR and immunohistochemistry. In summary, exposure of BEAS-2B cells to CTPE may induce chromosomal instability through spindle checkpoint-related proteins.
Collapse
Affiliation(s)
- Peng Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China.,The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, Henan, China
| | - Zhitao Li
- Medical College, Henan University of Science and Technology, Luoyang, Henan, China
| | - Na Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Guangcai Duan
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yanming Feng
- Medical College, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yong Zhao
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Lixia Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Hansong Zhu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Qiao Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaozhuan Liu
- Medical College, Henan University of Science and Technology, Luoyang, Henan, China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Wu Yao
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Wang
- Department of Pulmonary, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yiming Wu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Feifei Feng
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
106
|
Huang Q, Feng J, Wu R, Yang Y, Dai C, Li J, Liao Y, Xiang M, Wang D, Du XB. Total Oxidant/Antioxidant Status in Sera of Patients with Esophageal Cancer. Med Sci Monit 2017; 23:3789-3794. [PMID: 28777781 PMCID: PMC5553443 DOI: 10.12659/msm.902210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Oxidative stress parameters such as total oxidant status (TOS), total antioxidant status (TAS), and oxidative stress index (OSI) have been studied in breast, thyroid, and simple esophageal cancers (EC). We evaluated these parameters in patients with EC and analyzed their correlations with treatment outcomes. Material/Method Serum TOS, TAS, and OSI in 92 patients with EC at different clinical stages and in 64 healthy people (controls) were measured. Results Serum TOS, TAS, and OSI were significantly different between patients with EC and healthy controls (all p<0.001); however, there were no significant differences across different clinical stages (all p>0.05). These factors are not correlated with smoking or drinking history (all p>0.05). Patients with EC with higher TOS and OSI and lower TAS had better responses to chemotherapy and/or radiotherapy, but there was no significant correlation with different responses (all p>0.05). In a receiver operating characteristic curve analysis comparing patients with EC with healthy controls, the Youden indices were 0.391, 0.886, and 1, respectively. Conclusions Serum TOS, TAS, and OSI were significantly different between patients with EC and healthy controls. In patients with EC, these factors were not correlated with smoking or drinking history or with clinical stage. Patients with EC with higher TOS and OSI and lower TAS had a trend towards better outcomes but it did not reach significance. Serum TOS and OSI are potential diagnostic biomarkers that can be used to identify cases of EC.
Collapse
Affiliation(s)
- Qingmei Huang
- Department of Oncology, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland).,Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Jiafu Feng
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland)
| | - Rong Wu
- Department of Oncology, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland)
| | - Yuwei Yang
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland)
| | - Chunmei Dai
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland)
| | - Jie Li
- Department of Oncology, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland)
| | - Yao Liao
- Department of Oncology, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland)
| | - Miao Xiang
- Department of Oncology, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland)
| | - Dong Wang
- Department of Surgery, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland)
| | - Xiao-Bo Du
- Department of Oncology, Mianyang Central Hospital, Mianyang, Sichuan, China (mainland).,Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| |
Collapse
|
107
|
Yao YF, Wang ZC, Wu SY, Li QF, Yu C, Liang XY, Lv PC, Duan YT, Zhu HL. Identification of novel 1-indolyl acetate-5-nitroimidazole derivatives of combretastatin A-4 as potential tubulin polymerization inhibitors. Biochem Pharmacol 2017; 137:10-28. [DOI: 10.1016/j.bcp.2017.04.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 04/25/2017] [Indexed: 01/26/2023]
|
108
|
Wolter P, Hanselmann S, Pattschull G, Schruf E, Gaubatz S. Central spindle proteins and mitotic kinesins are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cell lines and are potential targets for therapy. Oncotarget 2017; 8:11160-11172. [PMID: 28061449 PMCID: PMC5355254 DOI: 10.18632/oncotarget.14466] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/26/2016] [Indexed: 12/17/2022] Open
Abstract
The MuvB multiprotein complex, together with B-MYB and FOXM1 (MMB-FOXM1), plays an essential role in cell cycle progression by regulating the transcription of genes required for mitosis and cytokinesis. In many tumors, B-MYB and FOXM1 are overexpressed as part of the proliferation signature. However, the transcriptional targets that are important for oncogenesis have not been identified. Given that mitotic kinesins are highly expressed in cancer cells and that selected kinesins have been reported as target genes of MMB-FOXM1, we sought to determine which mitotic kinesins are directly regulated by MMB-FOXM1. We demonstrate that six mitotic kinesins and two microtubule-associated non-motor proteins (MAPs) CEP55 and PRC1 are direct transcriptional targets of MuvB, B-MYB and FOXM1 in breast cancer cells. Suppression of KIF23 and PRC1 strongly suppressed proliferation of MDA-MB-231 cells. The set of MMB-FOXM1 regulated kinesins genes and 4 additional kinesins which we referred to as the mitotic kinesin signature (MKS) is linked to poor outcome in breast cancer patients. Thus, mitotic kinesins could be used as prognostic biomarker and could be potential therapeutic targets for the treatment of breast cancer.
Collapse
Affiliation(s)
- Patrick Wolter
- Theodor Boveri Institute, Biocenter, University of Wuerzburg and Comprehensive Cancer Center Mainfranken, University of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Steffen Hanselmann
- Theodor Boveri Institute, Biocenter, University of Wuerzburg and Comprehensive Cancer Center Mainfranken, University of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Grit Pattschull
- Theodor Boveri Institute, Biocenter, University of Wuerzburg and Comprehensive Cancer Center Mainfranken, University of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Eva Schruf
- Theodor Boveri Institute, Biocenter, University of Wuerzburg and Comprehensive Cancer Center Mainfranken, University of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Stefan Gaubatz
- Theodor Boveri Institute, Biocenter, University of Wuerzburg and Comprehensive Cancer Center Mainfranken, University of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
109
|
|
110
|
Kim YH, Han ME, Oh SO. The molecular mechanism for nuclear transport and its application. Anat Cell Biol 2017; 50:77-85. [PMID: 28713609 PMCID: PMC5509903 DOI: 10.5115/acb.2017.50.2.77] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 03/31/2017] [Indexed: 12/30/2022] Open
Abstract
Transportation between the cytoplasm and the nucleoplasm is critical for many physiological and pathophysiological processes including gene expression, signal transduction, and oncogenesis. So, the molecular mechanism for the transportation needs to be studied not only to understand cell physiological processes but also to develop new diagnostic and therapeutic targets. Recent progress in the research of the nuclear transportation (import and export) via nuclear pore complex and four important factors affecting nuclear transport (nucleoporins, Ran, karyopherins, and nuclear localization signals/nuclear export signals) will be discussed. Moreover, the clinical significance of nuclear transport and its application will be reviewed. This review will provide some critical insight for the molecular design of therapeutics which need to be targeted inside the nucleus.
Collapse
Affiliation(s)
- Yun Hak Kim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Korea.,BEER, Busan Society of Evidence-Based mEdicine and Research, Busan, Korea.,Gene and Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Korea
| | - Myoung-Eun Han
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Korea.,Gene and Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Korea
| | - Sae-Ock Oh
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Korea.,Gene and Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Korea
| |
Collapse
|
111
|
Lin HT, Okumura T, Yatsuda Y, Ito S, Nakauchi H, Otsu M. Application of Droplet Digital PCR for Estimating Vector Copy Number States in Stem Cell Gene Therapy. Hum Gene Ther Methods 2017; 27:197-208. [PMID: 27763786 PMCID: PMC5111482 DOI: 10.1089/hgtb.2016.059] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Stable gene transfer into target cell populations via integrating viral vectors is widely used in stem cell gene therapy (SCGT). Accurate vector copy number (VCN) estimation has become increasingly important. However, existing methods of estimation such as real-time quantitative PCR are more restricted in practicality, especially during clinical trials, given the limited availability of sample materials from patients. This study demonstrates the application of an emerging technology called droplet digital PCR (ddPCR) in estimating VCN states in the context of SCGT. Induced pluripotent stem cells (iPSCs) derived from a patient with X-linked chronic granulomatous disease were used as clonable target cells for transduction with alpharetroviral vectors harboring codon-optimized CYBB cDNA. Precise primer–probe design followed by multiplex analysis conferred assay specificity. Accurate estimation of per-cell VCN values was possible without reliance on a reference standard curve. Sensitivity was high and the dynamic range of detection was wide. Assay reliability was validated by observation of consistent, reproducible, and distinct VCN clustering patterns for clones of transduced iPSCs with varying numbers of transgene copies. Taken together, use of ddPCR appears to offer a practical and robust approach to VCN estimation with a wide range of clinical and research applications.
Collapse
Affiliation(s)
- Huan-Ting Lin
- 1 Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo , Tokyo, Japan .,2 Division of Stem Cell Processing/Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo , Tokyo, Japan
| | - Takashi Okumura
- 2 Division of Stem Cell Processing/Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo , Tokyo, Japan
| | | | - Satoru Ito
- 3 Life Science Division, Bio-Rad Laboratories , Tokyo, Japan
| | - Hiromitsu Nakauchi
- 1 Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo , Tokyo, Japan .,4 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Makoto Otsu
- 1 Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo , Tokyo, Japan .,2 Division of Stem Cell Processing/Stem Cell Bank, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo , Tokyo, Japan
| |
Collapse
|
112
|
Mitotic Dysfunction Associated with Aging Hallmarks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1002:153-188. [DOI: 10.1007/978-3-319-57127-0_7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
113
|
Yu Z, Sun Y, She X, Wang Z, Chen S, Deng Z, Zhang Y, Liu Q, Liu Q, Zhao C, Li P, Liu C, Feng J, Fu H, Li G, Wu M. SIX3, a tumor suppressor, inhibits astrocytoma tumorigenesis by transcriptional repression of AURKA/B. J Hematol Oncol 2017; 10:115. [PMID: 28595628 PMCID: PMC5465582 DOI: 10.1186/s13045-017-0483-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/31/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND SIX homeobox 3 (SIX3) is a member of the sine oculis homeobox transcription factor family. It plays a vital role in the nervous system development. Our previous study showed that the SIX3 gene is hypermethylated, and its expression is decreased in astrocytoma, but the role of SIX3 remains unknown. METHODS Chromatin-immunoprecipitation (ChIP) and luciferase reporter assay were used to confirm the binding of SIX3 to the promoter regions of aurora kinase A (AURKA) and aurora kinase B (AURKB). Confocal imaging and co-immunoprecipitation (Co-IP) were used to detect the interaction between AURKA and AURKB. Flow cytometry was performed to assess the effect of SIX3 on cell cycle distribution. Colony formation, EdU incorporation, transwell, and intracranial xenograft assays were performed to demonstrate the effect of SIX3 on the malignant phenotype of astrocytoma cells. RESULTS SIX3 is identified as a novel negative transcriptional regulator of AURKA and AURKB, and it decreases the expression of AURKA and AURKB in a dose-dependent manner in astrocytoma cells. Importantly, interactions between AURKA and AURKB stabilize and protect AURKA/B from degradation, and overexpression of SIX3 does not affect these interactions; SIX3 also acts as a tumor suppressor, and it increases p53 activity and expression at the post-translational level by the negative regulation of AURKA or AURKB, reduces the events of numerical centrosomal aberrations and misaligned chromosomes, and significantly inhibits the proliferation, invasion, and tumorigenesis of astrocytoma in vitro and in vivo. Moreover, experiments using primary cultured astrocytoma cells indicate that astrocytoma patients with a low expression of SIX3 and mutant p53 are more sensitive to treatment with aurora kinase inhibitors. CONCLUSION SIX3 is a novel negative transcriptional regulator and acts as a tumor suppressor that directly represses the transcription of AURKA and AURKB in astrocytoma. For the first time, the functional interaction of AURKA and AURKB has been found, which aids in the protection of their stability, and partially explains their constant high expression and activity in cancers. SIX3 is a potential biomarker that could be used to predict the response of astrocytoma patients to aurora kinase inhibitors.
Collapse
Affiliation(s)
- Zhibin Yu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Yingnan Sun
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Xiaoling She
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Zeyou Wang
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shuai Chen
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Zhiyong Deng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Yan Zhang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Qiang Liu
- The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Qing Liu
- The Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chunhua Zhao
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
| | - Peiyao Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Changhong Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Jianbo Feng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Haijuan Fu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Guiyuan Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China.
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
114
|
New Insights in the Cytogenetic Practice: Karyotypic Chaos, Non-Clonal Chromosomal Alterations and Chromosomal Instability in Human Cancer and Therapy Response. Genes (Basel) 2017; 8:genes8060155. [PMID: 28587191 PMCID: PMC5485519 DOI: 10.3390/genes8060155] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 05/26/2017] [Accepted: 05/31/2017] [Indexed: 12/17/2022] Open
Abstract
Recently, non-clonal chromosomal alterations previously unappreciated are being proposed to be included in cytogenetic practice. The aim of this inclusion is to obtain a greater understanding of chromosomal instability (CIN) and tumor heterogeneity and their role in cancer evolution and therapy response. Although several genetic assays have allowed the evaluation of the variation in a population of cancer cells, these assays do not provide information at the level of individual cells, therefore limiting the information of the genomic diversity within tumors (heterogeneity). The karyotype is one of the few available cytogenetic techniques that allow us not only to identify the chromosomal alterations present within a single cell, but also allows us to profile both clonal (CCA) and non-clonal chromosomal alterations (NCCAs). A greater understanding of CIN and tumor heterogeneity in cancer could not only improve existing therapeutic regimens but could also be used as targets for the design of new therapeutic approaches. In this review we indicate the importance and significance of karyotypic chaos, NCCAs and CIN in the prognosis of human cancers.
Collapse
|
115
|
Weiler SME, Pinna F, Wolf T, Lutz T, Geldiyev A, Sticht C, Knaub M, Thomann S, Bissinger M, Wan S, Rössler S, Becker D, Gretz N, Lang H, Bergmann F, Ustiyan V, Kalin TV, Singer S, Lee JS, Marquardt JU, Schirmacher P, Kalinichenko VV, Breuhahn K. Induction of Chromosome Instability by Activation of Yes-Associated Protein and Forkhead Box M1 in Liver Cancer. Gastroenterology 2017; 152:2037-2051.e22. [PMID: 28249813 DOI: 10.1053/j.gastro.2017.02.018] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/07/2017] [Accepted: 02/19/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Many different types of cancer cells have chromosome instability. The hippo pathway leads to phosphorylation of the transcriptional activator yes-associated protein 1 (YAP1, YAP), which regulates proliferation and has been associated with the development of liver cancer. We investigated the effects of hippo signaling via YAP on chromosome stability and hepatocarcinogenesis in humans and mice. METHODS We analyzed transcriptome data from 242 patients with hepatocellular carcinoma (HCC) to search for gene signatures associated with chromosomal instability (CIN); we investigated associations with overall survival time and cancer recurrence using Kaplan-Meier curves. We analyzed changes in expression of these signature genes, at mRNA and protein levels, after small interfering RNA-mediated silencing of YAP in Sk-Hep1, SNU182, HepG2, or pancreatic cancer cells, as well as incubation with thiostrepton (an inhibitor of forkhead box M1 [FOXM1]) or verteporfin (inhibitor of the interaction between YAP and TEA domain transcription factor 4 [TEAD4]). We performed co-immunoprecipitation and chromatin immunoprecipitation experiments. We collected liver tissues from mice that express a constitutively active form of YAP (YAPS127A) and analyzed gene expression signatures and histomorphologic parameters associated with chromosomal instability. Mice were given injections of thiostrepton and livers were collected and analyzed by immunoblotting, immunohistochemistry, histology, and real-time polymerase chain reaction. We performed immunohistochemical analyses on tissue microarrays of 105 HCCs and 7 nontumor liver tissues. RESULTS Gene expression patterns associated with chromosome instability, called CIN25 and CIN70, were detected in HCCs from patients with shorter survival time or early cancer recurrence. TEAD4 and YAP were required for CIN25 and CIN70 signature expression via induction and binding of FOXM1. Disrupting the interaction between YAP and TEAD4 with verteporfin, or inhibiting FOXM1 with thiostrepton, reduced the chromosome instability gene expression patterns. Hyperplastic livers and tumors from YAPS127A mice had increased CIN25 and CIN70 gene expression patterns, aneuploidy, and defects in mitosis. Injection of YAPS127A mice with thiostrepton reduced liver overgrowth and signs of chromosomal instability. In human HCC tissues, high levels of nuclear YAP correlated with increased chromosome instability gene expression patterns and aneuploidy. CONCLUSIONS By analyzing cell lines, genetically modified mice, and HCC tissues, we found that YAP cooperates with FOXM1 to contribute to chromosome instability. Agents that disrupt this pathway might be developed as treatments for liver cancer. Transcriptome data are available in the Gene Expression Omnibus public database (accession numbers: GSE32597 and GSE73396).
Collapse
Affiliation(s)
- Sofia M E Weiler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Federico Pinna
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Wolf
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Teresa Lutz
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Aman Geldiyev
- International Educational-Scientific Center, Ashgabat City, Turkmenistan
| | - Carsten Sticht
- Medical Faculty Mannheim, Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Maria Knaub
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Thomann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michaela Bissinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Shan Wan
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephanie Rössler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Diana Becker
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Center, University of Heidelberg, Mannheim, Germany
| | - Hauke Lang
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany
| | - Frank Bergmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Vladimir Ustiyan
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Tatiana V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stephan Singer
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jens U Marquardt
- Department of Medicine I, Johannes Gutenberg University, Mainz, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Vladimir V Kalinichenko
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
116
|
Intracellular HMGB1 as a novel tumor suppressor of pancreatic cancer. Cell Res 2017; 27:916-932. [PMID: 28374746 PMCID: PMC5518983 DOI: 10.1038/cr.2017.51] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/11/2017] [Accepted: 02/28/2017] [Indexed: 02/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) driven by oncogenic K-Ras remains among the most lethal human cancers despite recent advances in modern medicine. The pathogenesis of PDAC is partly attributable to intrinsic chromosome instability and extrinsic inflammation activation. However, the molecular link between these two events in pancreatic tumorigenesis has not yet been fully established. Here, we show that intracellular high mobility group box 1 (HMGB1) remarkably suppresses oncogenic K-Ras-driven pancreatic tumorigenesis by inhibiting chromosome instability-mediated pro-inflammatory nucleosome release. Conditional genetic ablation of either single or both alleles of HMGB1 in the pancreas renders mice extremely sensitive to oncogenic K-Ras-driven initiation of precursor lesions at birth, including pancreatic intraepithelial neoplasms, intraductal papillary mucinous neoplasms, and mucinous cystic neoplasms. Loss of HMGB1 in the pancreas is associated with oxidative DNA damage and chromosomal instability characterized by chromosome rearrangements and telomere abnormalities. These lead to inflammatory nucleosome release and propagate K-Ras-driven pancreatic tumorigenesis. Extracellular nucleosomes promote interleukin 6 (IL-6) secretion by infiltrating macrophages/neutrophils and enhance oncogenic K-Ras signaling activation in pancreatic lesions. Neutralizing antibodies to IL-6 or histone H3 or knockout of the receptor for advanced glycation end products all limit K-Ras signaling activation, prevent cancer development and metastasis/invasion, and prolong animal survival in Pdx1-Cre;K-RasG12D/+;Hmgb1−/− mice. Pharmacological inhibition of HMGB1 loss by glycyrrhizin limits oncogenic K-Ras-driven tumorigenesis in mice under inflammatory conditions. Diminished nuclear and total cellular expression of HMGB1 in PDAC patients correlates with poor overall survival, supporting intracellular HMGB1 as a novel tumor suppressor with prognostic and therapeutic relevance in PDAC.
Collapse
|
117
|
Prognostic value of CA20, a score based on centrosome amplification-associated genes, in breast tumors. Sci Rep 2017; 7:262. [PMID: 28325915 PMCID: PMC5428291 DOI: 10.1038/s41598-017-00363-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/20/2017] [Indexed: 11/08/2022] Open
Abstract
Centrosome amplification (CA) is a hallmark of cancer, observable in ≥75% of breast tumors. CA drives aggressive cellular phenotypes such as chromosomal instability (CIN) and invasiveness. Thus, assessment of CA may offer insights into the prognosis of breast cancer and identify patients who might benefit from centrosome declustering agents. However, it remains unclear whether CA is correlated with clinical outcomes after adjusting for confounding factors. To gain insights, we developed a signature, “CA20”, comprising centrosome structural genes and genes whose dysregulation is implicated in inducing CA. We found that CA20 was a significant independent predictor of worse survival in two large independent datasets after adjusting for potentially confounding factors. In multivariable analyses including both CA20 and CIN25 (a gene expression-based score that correlates with aneuploidy and has prognostic value in many types of cancer), only CA20 was significant, suggesting CA20 captures the risk-predictive information of CIN25 and offers information beyond it. CA20 correlated strongly with CIN25, so a high CA20 score may reflect tumors with high CIN and potentially other aggressive features that may require more aggressive treatment. Finally, we identified processes and pathways differing between CA20-low and high groups that may be valuable therapeutic targets.
Collapse
|
118
|
Lampson MA, Grishchuk EL. Mechanisms to Avoid and Correct Erroneous Kinetochore-Microtubule Attachments. BIOLOGY 2017; 6:E1. [PMID: 28067761 PMCID: PMC5371994 DOI: 10.3390/biology6010001] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/24/2016] [Accepted: 12/28/2016] [Indexed: 12/19/2022]
Abstract
In dividing vertebrate cells multiple microtubules must connect to mitotic kinetochores in a highly stereotypical manner, with each sister kinetochore forming microtubule attachments to only one spindle pole. The exact sequence of events by which this goal is achieved varies considerably from cell to cell because of the variable locations of kinetochores and spindle poles, and randomness of initial microtubule attachments. These chance encounters with the kinetochores nonetheless ultimately lead to the desired outcome with high fidelity and in a limited time frame, providing one of the most startling examples of biological self-organization. This chapter discusses mechanisms that contribute to accurate chromosome segregation by helping dividing cells to avoid and resolve improper microtubule attachments.
Collapse
Affiliation(s)
- Michael A Lampson
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Ekaterina L Grishchuk
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
119
|
Nähse V, Christ L, Stenmark H, Campsteijn C. The Abscission Checkpoint: Making It to the Final Cut. Trends Cell Biol 2016; 27:1-11. [PMID: 27810282 DOI: 10.1016/j.tcb.2016.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 10/08/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022]
Abstract
Cytokinesis is the final stage of cell division and is concluded by abscission of the intercellular bridge to physically separate the daughter cells. Timing of cytokinetic abscission is monitored by a molecular machinery termed the abscission checkpoint. This machinery delays abscission in cells with persistent chromatin in the intercellular bridge. Recent work has also uncovered its response to high membrane tension, nuclear pore defects, and DNA replication stress. Although it is known that the abscission checkpoint depends on persistent activity of the Aurora B protein kinase, we have only recently begun to understand its molecular basis. We propose here a molecular framework for abscission checkpoint signaling and we discuss outstanding questions relating to its function and physiological relevance.
Collapse
Affiliation(s)
- Viola Nähse
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | - Liliane Christ
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | - Harald Stenmark
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Faculty of Medicine, 7491 Trondheim, Norway.
| | - Coen Campsteijn
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Montebello, 0379 Oslo, Norway; Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway.
| |
Collapse
|
120
|
Cosenza MR, Krämer A. Centrosome amplification, chromosomal instability and cancer: mechanistic, clinical and therapeutic issues. Chromosome Res 2016; 24:105-26. [PMID: 26645976 DOI: 10.1007/s10577-015-9505-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Centrosomes, the main microtubule-organizing centers in most animal cells, are of crucial importance for the assembly of a bipolar mitotic spindle and subsequent faithful segregation of chromosomes into two daughter cells. Centrosome abnormalities can be found in virtually all cancer types and have been linked to chromosomal instability (CIN) and tumorigenesis. Although our knowledge on centrosome structure, replication, and amplification has greatly increased within recent years, still only very little is known on nature, causes, and consequences of centrosome aberrations in primary tumor tissues. In this review, we summarize our current insights into the mechanistic link between centrosome aberrations, aneuploidy, CIN and tumorigenesis. Mechanisms of induction and cellular consequences of aneuploidy, tetraploidization and CIN, as well as origin and effects of supernumerary centrosomes will be discussed. In addition, animal models for both CIN and centrosome amplification will be outlined. Finally, we describe approaches to exploit centrosome amplification, aneuploidy and CIN for novel and specific anticancer treatment strategies based on the modulation of chromosome missegregation rates.
Collapse
Affiliation(s)
- Marco Raffaele Cosenza
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
121
|
Dócs O, Hegyi K, Mokánszky A, Mónusné A, Beke L, András C, Bedekovics J, Méhes G. Mutant KRAS Status Is Associated with Increased KRAS Copy Number Imbalance: a Potential Mechanism of Molecular Heterogeneity. Pathol Oncol Res 2016; 23:417-423. [PMID: 27743339 DOI: 10.1007/s12253-016-0126-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 10/05/2016] [Indexed: 12/30/2022]
Abstract
Mutation rates determined by allele-specific PCR can be highly different in KRAS exon 2 mutant colorectal carcinoma (CRC) samples suggesting intratumoural heterogeneity. To address the effect of KRAS gene copy number on the relative mutant allele frequency the KRAS locus was individually quantified following FISH analysis in 36 cases. We observed, that mutant KRAS status was associated with an elevated KRAS locus number (2.36 ± 0.42 vs 2.63 ± 0.75; p = 0.037) reflecting an increased aneuploidy status but no true amplification of the locus. In parallel, KRAS locus copy numbers showed significant intercellular variability (1-16 copies/cell nucleus) within individual tumours also indicating to a dynamic intratumoural oscillation of the mutant allele copy number. In conclusion, aneusomy is a common feature of KRAS mutant CRC and KRAS copy number variations may have an impact on the relative mutant allele frequency detected by allele specific PCR/sequencing), potentially leading to subclonal diversity and influencing tumour behaviour.
Collapse
Affiliation(s)
- Ottó Dócs
- Department of Pathology, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Katalin Hegyi
- Department of Pathology, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Attila Mokánszky
- Department of Pathology, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Anikó Mónusné
- Department of Pathology, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Lívia Beke
- Department of Pathology, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Csilla András
- Department of Oncology, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Judit Bedekovics
- Department of Pathology, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary
| | - Gábor Méhes
- Department of Pathology, University of Debrecen, Nagyerdei krt. 98, Debrecen, H-4032, Hungary.
| |
Collapse
|
122
|
Deficiency of RITA results in multiple mitotic defects by affecting microtubule dynamics. Oncogene 2016; 36:2146-2159. [DOI: 10.1038/onc.2016.372] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/08/2016] [Accepted: 08/28/2016] [Indexed: 01/31/2023]
|
123
|
ASPP1/2-PP1 complexes are required for chromosome segregation and kinetochore-microtubule attachments. Oncotarget 2016; 6:41550-65. [PMID: 26595804 PMCID: PMC4747173 DOI: 10.18632/oncotarget.6355] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/02/2015] [Indexed: 01/01/2023] Open
Abstract
Regulated interactions between kinetochores and spindle microtubules are critical for maintaining genomic stability during chromosome segregation. Defects in chromosome segregation are widespread phenomenon in human cancers that are thought to serve as the fuel for tumorigenic progression. Tumor suppressor proteins ASPP1 and ASPP2, two members of the apoptosis stimulating proteins of p53 (ASPP) family, are frequently down-regulated in human cancers. Here we report that ASPP1/2 are required for proper mitotic progression. In ASPP1/2 co-depleted cells, the persistence of unaligned chromosomes and the reduction of tension across sister kinetochores on aligned chromosomes resulted in persistent spindle assembly checkpoint (SAC) activation. Using protein affinity purification methods, we searched for functional partners of ASPP1/2, and found that ASPP1/2 were associated with a subset of kinetochore proteins (Hec1, KNL-1, and CENP-F). It was found that ASPP1/2 act as PP1-targeting subunits to facilitate the interaction between PP1 and Hec1, and catalyze Hec1 (Ser165) dephosphorylation during late mitosis. These observations revealed a previously unrecognized function of ASPP1/2 in chromosome segregation and kinetochore-microtubule attachments that likely contributes to their roles in chromosome stability and tumor suppression.
Collapse
|
124
|
Duffy S, Fam HK, Wang YK, Styles EB, Kim JH, Ang JS, Singh T, Larionov V, Shah SP, Andrews B, Boerkoel CF, Hieter P. Overexpression screens identify conserved dosage chromosome instability genes in yeast and human cancer. Proc Natl Acad Sci U S A 2016; 113:9967-76. [PMID: 27551064 PMCID: PMC5018746 DOI: 10.1073/pnas.1611839113] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Somatic copy number amplification and gene overexpression are common features of many cancers. To determine the role of gene overexpression on chromosome instability (CIN), we performed genome-wide screens in the budding yeast for yeast genes that cause CIN when overexpressed, a phenotype we refer to as dosage CIN (dCIN), and identified 245 dCIN genes. This catalog of genes reveals human orthologs known to be recurrently overexpressed and/or amplified in tumors. We show that two genes, TDP1, a tyrosyl-DNA-phosphdiesterase, and TAF12, an RNA polymerase II TATA-box binding factor, cause CIN when overexpressed in human cells. Rhabdomyosarcoma lines with elevated human Tdp1 levels also exhibit CIN that can be partially rescued by siRNA-mediated knockdown of TDP1 Overexpression of dCIN genes represents a genetic vulnerability that could be leveraged for selective killing of cancer cells through targeting of an unlinked synthetic dosage lethal (SDL) partner. Using SDL screens in yeast, we identified a set of genes that when deleted specifically kill cells with high levels of Tdp1. One gene was the histone deacetylase RPD3, for which there are known inhibitors. Both HT1080 cells overexpressing hTDP1 and rhabdomyosarcoma cells with elevated levels of hTdp1 were more sensitive to histone deacetylase inhibitors valproic acid (VPA) and trichostatin A (TSA), recapitulating the SDL interaction in human cells and suggesting VPA and TSA as potential therapeutic agents for tumors with elevated levels of hTdp1. The catalog of dCIN genes presented here provides a candidate list to identify genes that cause CIN when overexpressed in cancer, which can then be leveraged through SDL to selectively target tumors.
Collapse
Affiliation(s)
- Supipi Duffy
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada V6T 1Z4
| | - Hok Khim Fam
- Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada V5Z 4H4; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | - Yi Kan Wang
- BC Cancer Agency, Vancouver, BC, Canada V5Z 4E6
| | - Erin B Styles
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada M5S 1A8; The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada M5S 3E1
| | - Jung-Hyun Kim
- Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892
| | - J Sidney Ang
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada V6T 1Z4
| | - Tejomayee Singh
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada V6T 1Z4
| | - Vladimir Larionov
- Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892
| | | | - Brenda Andrews
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada M5S 1A8; The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada M5S 3E1
| | - Cornelius F Boerkoel
- Child and Family Research Institute, University of British Columbia, Vancouver, BC, Canada V5Z 4H4; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | - Philip Hieter
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada V6T 1Z4; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada V6T 1Z3;
| |
Collapse
|
125
|
Spears M, Yousif F, Lyttle N, Boutros PC, Munro AF, Twelves C, Pritchard KI, Levine MN, Shepherd L, Bartlett JMS. A four gene signature predicts benefit from anthracyclines: evidence from the BR9601 and MA.5 clinical trials. Oncotarget 2016; 6:31693-701. [PMID: 26372731 PMCID: PMC4741633 DOI: 10.18632/oncotarget.5562] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/10/2015] [Indexed: 12/21/2022] Open
Abstract
Chromosome instability (CIN) in solid tumours results in multiple numerical and structural chromosomal aberrations and is associated with poor prognosis in multiple tumour types. Recent evidence demonstrated CEP17 duplication, a CIN marker, is a predictive marker of anthracycline benefit. An analysis of the BR9601 and MA.5 clinical trials was performed to test the role of existing CIN gene expression signatures as predictive markers of anthracycline sensitivity in breast cancer. Univariate analysis demonstrated, high CIN25 expression score was associated with improved distant relapse free survival (DRFS) (HR: 0.74, 95% CI 0.54-0.99, p = 0.046). High tumour CIN70 and CIN25 scores were associated with aggressive clinicopathological phenotype and increased sensitivity to anthracycline therapy compared to low CIN scores. However, in a prospectively planned multivariate analysis only pathological grade, nodal status and tumour size were significant predictors of outcome for CIN25/CIN70. A limited gene signature was generated, patients with low tumour CIN4 scores benefited from anthracycline treatment significantly more than those with high CIN4 scores (HR 0.37, 95% CI 0.20-0.56, p = 0.001). In multivariate analyses the treatment by marker interaction for CIN4/anthracyclines demonstrated hazard ratio of 0.35 (95% CI 0.15-0.80, p = 0.012) for DRFS. This data shows CIN4 is independent predictor of anthracycline benefit for DRFS in breast cancer.
Collapse
Affiliation(s)
- Melanie Spears
- Transformative Pathology, Ontario Institute for Cancer Research, MaRS Centre, Toronto, ON, Canada
| | - Fouad Yousif
- Informatics and Bio-Computing, Ontario Institute for Cancer Research, MaRS Centre, Toronto, ON, Canada
| | - Nicola Lyttle
- Transformative Pathology, Ontario Institute for Cancer Research, MaRS Centre, Toronto, ON, Canada
| | - Paul C Boutros
- Informatics and Bio-Computing, Ontario Institute for Cancer Research, MaRS Centre, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Alison F Munro
- Edinburgh Cancer Research UK Centre, MRC IGMM, University of Edinburgh, Crewe Road South, Edinburgh, UK
| | - Chris Twelves
- Leeds Institute of Cancer and Pathology and Cancer Research UK Centre, St James' University Hospital, Leeds, UK
| | - Kathleen I Pritchard
- Sunnybrook Odette Cancer Centre, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Mark N Levine
- McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada
| | - Lois Shepherd
- NCIC Clinical Trials Group (NCIC CTG] and Queen's University, Kingston, ON, Canada
| | - John M S Bartlett
- Transformative Pathology, Ontario Institute for Cancer Research, MaRS Centre, Toronto, ON, Canada.,Edinburgh Cancer Research UK Centre, MRC IGMM, University of Edinburgh, Crewe Road South, Edinburgh, UK
| |
Collapse
|
126
|
Gao R, Davis A, McDonald TO, Sei E, Shi X, Wang Y, Tsai PC, Casasent A, Waters J, Zhang H, Meric-Bernstam F, Michor F, Navin NE. Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat Genet 2016; 48:1119-30. [PMID: 27526321 PMCID: PMC5042845 DOI: 10.1038/ng.3641] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 07/13/2016] [Indexed: 12/15/2022]
Abstract
Aneuploidy is a hallmark of breast cancer; however, our knowledge of how these complex genomic rearrangements evolve during tumorigenesis is limited. In this study we developed a highly multiplexed single-nucleus-sequencing method to investigate copy number evolution in triple-negative breast cancer patients. We sequenced 1000 single cells from 12 patients and identified 1–3 major clonal subpopulations in each tumor that shared a common evolutionary lineage. We also identified a minor subpopulation of non-clonal cells that were classified as: 1) metastable, 2) pseudo-diploid, or 3) chromazemic. Phylogenetic analysis and mathematical modeling suggest that these data are unlikely to be explained by the gradual accumulation of copy number events over time. In contrast, our data challenge the paradigm of gradual evolution, showing that the majority of copy number aberrations are acquired at the earliest stages of tumor evolution, in short punctuated bursts, followed by stable clonal expansions that form the tumor mass.
Collapse
Affiliation(s)
- Ruli Gao
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alexander Davis
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Thomas O McDonald
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Emi Sei
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xiuqing Shi
- Peking Union Medical College, Department of Medical Oncology, Cancer Hospital and Institute, Chinese Academy of Medical Sciences, Beijing, China
| | - Yong Wang
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pei-Ching Tsai
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anna Casasent
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jill Waters
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hong Zhang
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Funda Meric-Bernstam
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Franziska Michor
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Nicholas E Navin
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
127
|
An important role for Myb-MuvB and its target gene KIF23 in a mouse model of lung adenocarcinoma. Oncogene 2016; 36:110-121. [PMID: 27212033 DOI: 10.1038/onc.2016.181] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/30/2016] [Accepted: 04/15/2016] [Indexed: 12/14/2022]
Abstract
The conserved Myb-MuvB (MMB) multiprotein complex has an important role in transcriptional activation of mitotic genes. MMB target genes are overexpressed in several different cancer types and their elevated expression is associated with an advanced tumor state and a poor prognosis. This suggests that MMB could contribute to tumorigenesis by mediating overexpression of mitotic genes. However, although MMB has been extensively characterized biochemically, the requirement for MMB in tumorigenesis in vivo has not been investigated. Here we demonstrate that MMB is required for tumor formation in a mouse model of lung cancer driven by oncogenic K-RAS. We also identify a requirement for the mitotic kinesin KIF23, a key target gene of MMB, in tumorigenesis. RNA interference-mediated depletion of KIF23 inhibited lung tumor formation in vivo and induced apoptosis in lung cancer cell lines. Our results suggest that inhibition of KIF23 could be a strategy for treatment of lung cancer.
Collapse
|
128
|
Chan A, Singh AJ, Northcote PT, Miller JH. Peloruside A, a microtubule-stabilizing agent, induces aneuploidy in ovarian cancer cells. Invest New Drugs 2016; 34:424-38. [PMID: 27155614 DOI: 10.1007/s10637-016-0355-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 04/27/2016] [Indexed: 11/29/2022]
Abstract
To ensure proper chromosome segregation, mitosis is tightly regulated by the spindle assembly checkpoint (SAC). Low concentrations of microtubule-stabilizing agents can induce aneuploid populations of cells in the absence of G2/M block, suggesting pertubation of the spindle checkpoint. We investigated the effects of peloruside A, a microtubule-stabilizing agent, on expression levels of several key cell cycle proteins, MAD2, BUBR1, p55CDC and cyclin B1. Synchronized 1A9 ovarian carcinoma cells were allowed to progress through the cell cycle in the presence or absence of peloruside A. Co-immunoprecipitation and Western blotting were used to probe the cell cycle kinetics of MAD2 and BUBR1 dissociation from p55CDC. Using confocal microscopy, we investigated whether premature dissociation of MAD2 and BUBR1 at low (40 nM) but not high (100 nM) concentrations of peloruside A was caused by defects in the attachment of chromosomes to the mitotic spindle. An increased frequency of polar chromosomes was observed at low concentrations of peloruside A, suggesting that an increased frequency of pseudo-metaphase cells, which are not detected by the spindle assembly checkpoint, may be underlying the induction of aneuploidy.
Collapse
Affiliation(s)
- Ariane Chan
- School of Biological Sciences, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand.,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand.,Volpara Solutions Limited, Level 12, 86 Victoria Street, Wellington, 6011, New Zealand
| | - A Jonathan Singh
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand.,School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand.,Molecular Targets Laboratory, Center for Cancer Research, National Cancer Institute - Frederick, Frederick, MD, 21702, USA
| | - Peter T Northcote
- Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand.,School of Chemical and Physical Sciences, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand
| | - John H Miller
- School of Biological Sciences, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand. .,Centre for Biodiscovery, Victoria University of Wellington, PO Box 600, Wellington, 6140, New Zealand.
| |
Collapse
|
129
|
Ritter A, Kreis NN, Louwen F, Wordeman L, Yuan J. Molecular insight into the regulation and function of MCAK. Crit Rev Biochem Mol Biol 2016; 51:228-45. [DOI: 10.1080/10409238.2016.1178705] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
130
|
Abstract
Wilms' tumor-1 protein (WT1) is a transcription factor that can either activate or repress genes to regulate cell growth, apoptosis and differentiation. WT1 can act as either a tumor suppressor or an oncogene. The cellular functions of WT1 are predominantly regulated by its various interacting partners. Recently we have found that WT1 can regulate the fidelity of chromosome segregation through its interaction with the spindle assembly checkpoint protein, Mitotic arrest deficient-2 (MAD2). WT1 delays anaphase entry by inhibiting the ubiquitination activity of the Anaphase promoting complex/cyclosome (APC/C). Our findings have revealed an important role of WT1 in the regulation of mitotic checkpoint and genomic stability.
Collapse
Affiliation(s)
- Jayasha Shandilya
- a Department of Biological Sciences ; University at Buffalo ; Buffalo , NY USA
| | | |
Collapse
|
131
|
The role of oxidative stress on breast cancer development and therapy. Tumour Biol 2016; 37:4281-91. [PMID: 26815507 DOI: 10.1007/s13277-016-4873-9] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/15/2016] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are produced by both enzymatic and non-enzymatic systems within eukaryotic cells and play important roles in cellular physiology and pathophysiology. Although physiological concentrations are crucial for ensuring cell survival, ROS overproduction is detrimental to cells, and considered key-factors for the development of several diseases, such as neurodegenerative diseases, cardiovascular disorders, and cancer. Cancer cells are usually submitted to higher ROS levels that further stimulate malignant phenotype through stimulus to sustained proliferation, death evasion, angiogenesis, invasiveness, and metastasis. The role of ROS on breast cancer etiology and progression is being progressively elucidated. However, less attention has been given to the development of redox system-targeted strategies for breast cancer therapy. In this review, we address the basic mechanisms of ROS production and scavenging in breast tumor cells, and the emerging possibilities of breast cancer therapies targeting ROS homeostasis.
Collapse
|
132
|
Teng YC, Shen ZQ, Kao CH, Tsai TF. Hepatocellular carcinoma mouse models: Hepatitis B virus-associated hepatocarcinogenesis and haploinsufficient tumor suppressor genes. World J Gastroenterol 2016; 22:300-325. [PMID: 26755878 PMCID: PMC4698494 DOI: 10.3748/wjg.v22.i1.300] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 10/14/2015] [Accepted: 11/24/2015] [Indexed: 02/06/2023] Open
Abstract
The multifactorial and multistage pathogenesis of hepatocellular carcinoma (HCC) has fascinated a wide spectrum of scientists for decades. While a number of major risk factors have been identified, their mechanistic roles in hepatocarcinogenesis still need to be elucidated. Many tumor suppressor genes (TSGs) have been identified as being involved in HCC. These TSGs can be classified into two groups depending on the situation with respect to allelic mutation/loss in the tumors: the recessive TSGs with two required mutated alleles and the haploinsufficient TSGs with one required mutated allele. Hepatitis B virus (HBV) is one of the most important risk factors associated with HCC. Although mice cannot be infected with HBV due to the narrow host range of HBV and the lack of a proper receptor, one advantage of mouse models for HBV/HCC research is the numerous and powerful genetic tools that help investigate the phenotypic effects of viral proteins and allow the dissection of the dose-dependent action of TSGs. Here, we mainly focus on the application of mouse models in relation to HBV-associated HCC and on TSGs that act either in a recessive or in a haploinsufficient manner. Discoveries obtained using mouse models will have a great impact on HCC translational medicine.
Collapse
|
133
|
Abdul-Sater Z, Cerabona D, Potchanant ES, Sun Z, Enzor R, He Y, Robertson K, Goebel WS, Nalepa G. FANCA safeguards interphase and mitosis during hematopoiesis in vivo. Exp Hematol 2015; 43:1031-1046.e12. [PMID: 26366677 PMCID: PMC4666759 DOI: 10.1016/j.exphem.2015.08.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 12/20/2022]
Abstract
The Fanconi anemia (FA/BRCA) signaling network controls multiple genome-housekeeping checkpoints, from interphase DNA repair to mitosis. The in vivo role of abnormal cell division in FA remains unknown. Here, we quantified the origins of genomic instability in FA patients and mice in vivo and ex vivo. We found that both mitotic errors and interphase DNA damage significantly contribute to genomic instability during FA-deficient hematopoiesis and in nonhematopoietic human and murine FA primary cells. Super-resolution microscopy coupled with functional assays revealed that FANCA shuttles to the pericentriolar material to regulate spindle assembly at mitotic entry. Loss of FA signaling rendered cells hypersensitive to spindle chemotherapeutics and allowed escape from the chemotherapy-induced spindle assembly checkpoint. In support of these findings, direct comparison of DNA crosslinking and anti-mitotic chemotherapeutics in primary FANCA-/- cells revealed genomic instability originating through divergent cell cycle checkpoint aberrations. Our data indicate that FA/BRCA signaling functions as an in vivo gatekeeper of genomic integrity throughout interphase and mitosis, which may have implications for future targeted therapies in FA and FA-deficient cancers.
Collapse
Affiliation(s)
- Zahi Abdul-Sater
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Donna Cerabona
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Elizabeth Sierra Potchanant
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Zejin Sun
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Rikki Enzor
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ying He
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kent Robertson
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - W Scott Goebel
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Grzegorz Nalepa
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana; Bone Marrow Failure Program, Division of Pediatric Hematology-Oncology, Riley Hospital for Children, Indianapolis, Indiana.
| |
Collapse
|
134
|
Bertalan Z, Budrikis Z, La Porta CAM, Zapperi S. Role of the Number of Microtubules in Chromosome Segregation during Cell Division. PLoS One 2015; 10:e0141305. [PMID: 26506005 PMCID: PMC4624697 DOI: 10.1371/journal.pone.0141305] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/07/2015] [Indexed: 12/02/2022] Open
Abstract
Faithful segregation of genetic material during cell division requires alignment of chromosomes between two spindle poles and attachment of their kinetochores to each of the poles. Failure of these complex dynamical processes leads to chromosomal instability (CIN), a characteristic feature of several diseases including cancer. While a multitude of biological factors regulating chromosome congression and bi-orientation have been identified, it is still unclear how they are integrated so that coherent chromosome motion emerges from a large collection of random and deterministic processes. Here we address this issue by a three dimensional computational model of motor-driven chromosome congression and bi-orientation during mitosis. Our model reveals that successful cell division requires control of the total number of microtubules: if this number is too small bi-orientation fails, while if it is too large not all the chromosomes are able to congress. The optimal number of microtubules predicted by our model compares well with early observations in mammalian cell spindles. Our results shed new light on the origin of several pathological conditions related to chromosomal instability.
Collapse
Affiliation(s)
- Zsolt Bertalan
- Institute for Scientific Interchange Foundation, Via Alassio 11/C, 10126 Torino, Italy
| | - Zoe Budrikis
- Institute for Scientific Interchange Foundation, Via Alassio 11/C, 10126 Torino, Italy
| | - Caterina A. M. La Porta
- Center for Complexity and Biosystems, Department of Bioscience, University of Milan, via Celoria 26, 20133 Milano, Italy
- * E-mail: (CAMLP); (SZ)
| | - Stefano Zapperi
- Institute for Scientific Interchange Foundation, Via Alassio 11/C, 10126 Torino, Italy
- Center for Complexity and Biosystems, Department of Physics, University of Milan, via Celoria 16, 20133 Milano, Italy
- CNR - Consiglio Nazionale delle Ricerche, Istituto per l’Energetica e le Interfasi, Via R. Cozzi 53, 20125 Milano, Italy
- Department of Applied Physics, Aalto University, P.O. Box 14100, FIN-00076 Aalto, Espoo, Finland
- * E-mail: (CAMLP); (SZ)
| |
Collapse
|
135
|
Koturbash I, Tolleson WH, Guo L, Yu D, Chen S, Hong H, Mattes W, Ning B. microRNAs as pharmacogenomic biomarkers for drug efficacy and drug safety assessment. Biomark Med 2015; 9:1153-76. [PMID: 26501795 PMCID: PMC5712454 DOI: 10.2217/bmm.15.89] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Much evidence has documented that microRNAs (miRNAs) play an important role in the modulation of interindividual variability in the production of drug metabolizing enzymes and transporters (DMETs) and nuclear receptors (NRs) through multidirectional interactions involving environmental stimuli/stressors, the expression of miRNA molecules and genetic polymorphisms. MiRNA expression has been reported to be affected by drugs and miRNAs themselves may affect drug metabolism and toxicity. In cancer research, miRNA biomarkers have been identified to mediate intrinsic and acquired resistance to cancer therapies. In drug safety assessment, miRNAs have been found associated with cardiotoxicity, hepatotoxicity and nephrotoxicity. This review article summarizes published studies to show that miRNAs can serve as early biomarkers for the evaluation of drug efficacy and drug safety.
Collapse
Affiliation(s)
- Igor Koturbash
- Department of Environmental & Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - William H Tolleson
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Lei Guo
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Dianke Yu
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Si Chen
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Huixiao Hong
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - William Mattes
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| | - Baitang Ning
- National Center for Toxicological Research, US Food & Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
136
|
Hudler P. Challenges of deciphering gastric cancer heterogeneity. World J Gastroenterol 2015; 21:10510-10527. [PMID: 26457012 PMCID: PMC4588074 DOI: 10.3748/wjg.v21.i37.10510] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 06/19/2015] [Accepted: 08/31/2015] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is in decline in most developed countries; however, it still accounts for a notable fraction of global mortality and morbidity related to cancer. High-throughput methods are rapidly changing our view and understanding of the molecular basis of gastric carcinogenesis. Today, it is widely accepted that the molecular complexity and heterogeneity, both inter- and intra-tumour, of gastric adenocarcinomas present significant obstacles in elucidating specific biomarkers for early detection of the disease. Although genome-wide sequencing and gene expression studies have revealed the intricate nature of the molecular changes that occur in tumour landscapes, the collected data and results are complex and sometimes contradictory. Several aberrant molecules have already been tested in clinical trials, although their diagnostic and prognostic utilities have not been confirmed thus far. The gold standard for the detection of sporadic gastric cancer is still the gastric endoscopy, which is considered invasive. In addition, genome-wide association studies have confirmed that genetic variations are important contributors to increased cancer risk and could participate in the initiation of malignant transformation. This hypothesis could in part explain the late onset of sporadic gastric cancers. The elaborate interplay of polymorphic low penetrance genes and lifestyle and environmental risk factors requires additional research to decipher their relative impacts on tumorigenesis. The purpose of this article is to present details of the molecular heterogeneity of sporadic gastric cancers at the DNA, RNA, and proteome levels and to discuss issues relevant to the translation of basic research data to clinically valuable tools. The focus of this work is the identification of relevant molecular changes that could be detected non-invasively.
Collapse
|
137
|
Martinez R, Blasina A, Hallin JF, Hu W, Rymer I, Fan J, Hoffman RL, Murphy S, Marx M, Yanochko G, Trajkovic D, Dinh D, Timofeevski S, Zhu Z, Sun P, Lappin PB, Murray BW. Mitotic Checkpoint Kinase Mps1 Has a Role in Normal Physiology which Impacts Clinical Utility. PLoS One 2015; 10:e0138616. [PMID: 26398286 PMCID: PMC4580473 DOI: 10.1371/journal.pone.0138616] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/25/2015] [Indexed: 12/20/2022] Open
Abstract
Cell cycle checkpoint intervention is an effective therapeutic strategy for cancer when applied to patients predisposed to respond and the treatment is well-tolerated. A critical cell cycle process that could be targeted is the mitotic checkpoint (spindle assembly checkpoint) which governs the metaphase-to-anaphase transition and insures proper chromosomal segregation. The mitotic checkpoint kinase Mps1 was selected to explore whether enhancement in genomic instability is a viable therapeutic strategy. The basal-a subset of triple-negative breast cancer was chosen as a model system because it has a higher incidence of chromosomal instability and Mps1 expression is up-regulated. Depletion of Mps1 reduces tumor cell viability relative to normal cells. Highly selective, extremely potent Mps1 kinase inhibitors were created to investigate the roles of Mps1 catalytic activity in tumor cells and normal physiology (PF-7006, PF-3837; Ki<0.5 nM; cellular IC50 2–6 nM). Treatment of tumor cells in vitro with PF-7006 modulates expected Mps1-dependent biology as demonstrated by molecular and phenotypic measures (reduced pHH3-Ser10 levels, shorter duration of mitosis, micro-nucleation, and apoptosis). Tumor-bearing mice treated with PF-7006 exhibit tumor growth inhibition concomitant with pharmacodynamic modulation of a downstream biomarker (pHH3-Ser10). Unfortunately, efficacy only occurs at drug exposures that cause dose-limiting body weight loss, gastrointestinal toxicities, and neutropenia. Mps1 inhibitor toxicities may be mitigated by inducing G1 cell cycle arrest in Rb1-competent cells with the cyclin-dependent kinase-4/6 inhibitor palbociclib. Using an isogenic cellular model system, PF-7006 is shown to be selectively cytotoxic to Rb1-deficient cells relative to Rb1-competent cells (also a measure of kinase selectivity). Human bone marrow cells pretreated with palbociclib have decreased PF-7006-dependent apoptosis relative to cells without palbociclib pretreatment. Collectively, this study raises a concern that single agent therapies inhibiting Mps1 will not be well-tolerated clinically but may be when combined with a selective CDK4/6 drug.
Collapse
Affiliation(s)
- Ricardo Martinez
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Alessandra Blasina
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Jill F. Hallin
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Wenyue Hu
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Isha Rymer
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Jeffery Fan
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Robert L. Hoffman
- Worldwide Medicinal Chemistry, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Sean Murphy
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Matthew Marx
- Worldwide Medicinal Chemistry, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Gina Yanochko
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Dusko Trajkovic
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Dac Dinh
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Sergei Timofeevski
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Zhou Zhu
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Peiquing Sun
- Department of Cell and Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, United States of America
| | - Patrick B. Lappin
- Drug Safety Research and Development, Pfizer Worldwide Research and Development, 10646 Science Center Drive, San Diego, CA, 92121, United States of America
| | - Brion W. Murray
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10724 Science Center Drive, San Diego, CA, 92121, United States of America
- * E-mail:
| |
Collapse
|
138
|
Somyajit K, Saxena S, Babu S, Mishra A, Nagaraju G. Mammalian RAD51 paralogs protect nascent DNA at stalled forks and mediate replication restart. Nucleic Acids Res 2015; 43:9835-55. [PMID: 26354865 PMCID: PMC4787763 DOI: 10.1093/nar/gkv880] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/23/2015] [Indexed: 12/22/2022] Open
Abstract
Mammalian RAD51 paralogs are implicated in the repair of collapsed replication forks by homologous recombination. However, their physiological roles in replication fork maintenance prior to fork collapse remain obscure. Here, we report on the role of RAD51 paralogs in short-term replicative stress devoid of DSBs. We show that RAD51 paralogs localize to nascent DNA and common fragile sites upon replication fork stalling. Strikingly, RAD51 paralogs deficient cells exhibit elevated levels of 53BP1 nuclear bodies and increased DSB formation, the latter being attributed to extensive degradation of nascent DNA at stalled forks. RAD51C and XRCC3 promote the restart of stalled replication in an ATP hydrolysis dependent manner by disengaging RAD51 and other RAD51 paralogs from the halted forks. Notably, we find that Fanconi anemia (FA)-like disorder and breast and ovarian cancer patient derived mutations of RAD51C fails to protect replication fork, exhibit under-replicated genomic regions and elevated micro-nucleation. Taken together, RAD51 paralogs prevent degradation of stalled forks and promote the restart of halted replication to avoid replication fork collapse, thereby maintaining genomic integrity and suppressing tumorigenesis.
Collapse
Affiliation(s)
- Kumar Somyajit
- Department of Biochemistry, Indian Institute of Science, Bangalore-560012, India
| | - Sneha Saxena
- Department of Biochemistry, Indian Institute of Science, Bangalore-560012, India
| | - Sharath Babu
- Department of Biochemistry, Indian Institute of Science, Bangalore-560012, India
| | - Anup Mishra
- Department of Biochemistry, Indian Institute of Science, Bangalore-560012, India
| | - Ganesh Nagaraju
- Department of Biochemistry, Indian Institute of Science, Bangalore-560012, India
| |
Collapse
|
139
|
Dependence of Human Colorectal Cells Lacking the FBW7 Tumor Suppressor on the Spindle Assembly Checkpoint. Genetics 2015; 201:885-95. [PMID: 26354767 PMCID: PMC4649658 DOI: 10.1534/genetics.115.180653] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/24/2015] [Indexed: 01/24/2023] Open
Abstract
FBW7 (F-box and WD repeat domain containing 7), also known as FBXW7 or hCDC4, is a tumor suppressor gene mutated in a broad spectrum of cancer cell types. As a component of the SCF E3 ubiquitin ligase, FBW7 is responsible for specifically recognizing phosphorylated substrates, many important for tumor progression, and targeting them for ubiquitin-mediated degradation. Although the role of FBW7 as a tumor suppressor is well established, less well studied is how FBW7-mutated cancer cells might be targeted for selective killing. To explore this further, we undertook a genome-wide RNAi screen using WT and FBW7 knockout colorectal cell lines and identified the spindle assembly checkpoint (SAC) protein BUBR1, as a candidate synthetic lethal target. We show here that asynchronous FBW7 knockout cells have increased levels of mitotic APC/C substrates and are sensitive to knockdown of not just BUBR1 but BUB1 and MPS1, other known SAC components, suggesting a dependence of these cells on the mitotic checkpoint. Consistent with this dependence, knockdown of BUBR1 in cells lacking FBW7 results in significant cell aneuploidy and increases in p53 levels. The FBW7 substrate cyclin E was necessary for the genetic interaction with BUBR1. In contrast, the establishment of this dependence on the SAC requires the deregulation of multiple substrates of FBW7. Our work suggests that FBW7 knockout cells are vulnerable in their dependence on the mitotic checkpoint and that this may be a good potential target to exploit in FBW7-mutated cancer cells.
Collapse
|
140
|
Hu S, Lu Y, Orr B, Godek K, Mustachio LM, Kawakami M, Sekula D, Compton DA, Freemantle S, Dmitrovsky E. Specific CP110 Phosphorylation Sites Mediate Anaphase Catastrophe after CDK2 Inhibition: Evidence for Cooperation with USP33 Knockdown. Mol Cancer Ther 2015; 14:2576-85. [PMID: 26304236 DOI: 10.1158/1535-7163.mct-15-0443] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/19/2015] [Indexed: 11/16/2022]
Abstract
Chromosomal instability (CIN) is a hallmark of solid tumor biology and is implicated in carcinogenesis. Preferentially eliminating malignant cells by targeting CIN and aneuploidy is an attractive antineoplastic strategy. We previously reported that CDK2 antagonism causes lung cancer cells to undergo anaphase catastrophe and apoptosis through inhibition of phosphorylation of the centrosomal protein CP110. Cells with activating KRAS mutations were particularly sensitive to CDK2 inhibition due to downregulation of CP110 protein levels. This study investigated mechanisms of CDK2 antagonism that mediate anaphase catastrophe via changes in CP110 protein expression and how activated KRAS affects CP110 levels in lung cancers. Site-directed mutagenesis revealed candidate CDK phosphorylation sites of CP110 (residues Ser 170 and Thr 194) critical for conferring anaphase catastrophe by altering centrosome clustering in mitosis. Intriguingly, KRAS mutation can promote CP110 protein degradation by upregulating the ubiquitin ligase SCF(cyclinF), which targets CP110 protein for destabilization. Finally, CDK2 inhibitor response was enhanced when combined with knockdown of the deubiquitinase USP33 that in turn accelerates CP110 protein degradation. Thus, this study provides molecular pharmacologic insights into how CP110 expression regulates response to CDK2 inhibition. An improved understanding of in vitro antineoplastic mechanisms of combining CDK2 antagonism with induced CP110 repression provides a rationale for exploring clinical consequences of this strategy. Taken together, preclinical findings obtained from combining CDK2 inhibition with USP33 repression have implications for treating patients with non-small cell lung cancers.
Collapse
Affiliation(s)
- Shanhu Hu
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Yun Lu
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Bernardo Orr
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Kristina Godek
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Lisa Maria Mustachio
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Masanori Kawakami
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - David Sekula
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Duane A Compton
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Sarah Freemantle
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Ethan Dmitrovsky
- Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, and Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
| |
Collapse
|
141
|
Peng J, Ma J, Li W, Mo R, Zhang P, Gao K, Jin X, Xiao J, Wang C, Fan J. Stabilization of MCRS1 by BAP1 prevents chromosome instability in renal cell carcinoma. Cancer Lett 2015; 369:167-74. [PMID: 26300492 DOI: 10.1016/j.canlet.2015.08.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/17/2015] [Accepted: 08/17/2015] [Indexed: 01/21/2023]
Abstract
Characterization of the exome and genome of carcinoma (ccRCC) by next-generation sequencing identified numerous genetic alternations. BRCA1-associated protein-1 (BAP1) was identified as one of the most frequently mutated genes in ccRCC, suggesting that BAP1 is a potential key driver for ccRCC cancer initiation and progression. However, how BAP1 mutations contribute to ccRCC remains to be elucidated. BAP1 is a nuclear de-ubiquitinating enzyme and cleaves the ubiquitin chain from the substrates. Here, we identified MCRS1 as a bona fide substrate for BAP1. MCRS1 is a component of the centrosome proteins, and plays an essential role in spindle assembly. BAP1 binds to MCRS1 and stabilizes MCRS1 by de-ubiquitination. BAP1 contributes to chromosome stability partially via MCRS1. A positive correlation was identified between BAP1 and MCRS1 expression in ccRCC tissues. Both BAP1 loss and MCRS1 down-regulation in ccRCC were associated with adverse clinicopathological features. This study revealed a novel mechanism for BAP1 involved in MCRS1 stability regulation, and provided insight in understanding the relationship between BAP1 mutations and chromosome instability in ccRCC.
Collapse
Affiliation(s)
- Jingtao Peng
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China
| | - Jian Ma
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China
| | - Weiguo Li
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China
| | - Ren Mo
- Department of Urology, Inner Mongolia Autonomous Region Peoples Hospital, 20 Zhaowuda Road, Hohhot 010017, China
| | - Pingzhao Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Kun Gao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Xiaofeng Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Jiantao Xiao
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China
| | - Chenji Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200433, China.
| | - Jie Fan
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China.
| |
Collapse
|
142
|
Abstract
Numerical chromosomal instability is a ubiquitous feature of human neoplasms. Due to experimental limitations, fundamental characteristics of karyotypic changes in cancer are poorly understood. Using an experimentally inspired stochastic model, based on the potency and chromosomal distribution of oncogenes and tumor suppressor genes, we show that cancer cells have evolved to exist within a narrow range of chromosome missegregation rates that optimizes phenotypic heterogeneity and clonal survival. Departure from this range reduces clonal fitness and limits subclonal diversity. Mapping of the aneuploid fitness landscape reveals a highly favorable, commonly observed, near-triploid state onto which evolving diploid- and tetraploid-derived populations spontaneously converge, albeit at a much lower fitness cost for the latter. Finally, by analyzing 1,368 chromosomal translocation events in five human cancers, we find that karyotypic evolution also shapes chromosomal translocation patterns by selecting for more oncogenic derivative chromosomes. Thus, chromosomal instability can generate the heterogeneity required for Darwinian tumor evolution.
Collapse
|
143
|
Ohashi A, Ohori M, Iwai K, Nakayama Y, Nambu T, Morishita D, Kawamoto T, Miyamoto M, Hirayama T, Okaniwa M, Banno H, Ishikawa T, Kandori H, Iwata K. Aneuploidy generates proteotoxic stress and DNA damage concurrently with p53-mediated post-mitotic apoptosis in SAC-impaired cells. Nat Commun 2015; 6:7668. [PMID: 26144554 PMCID: PMC4506520 DOI: 10.1038/ncomms8668] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 06/01/2015] [Indexed: 01/14/2023] Open
Abstract
The molecular mechanism responsible that determines cell fate after mitotic slippage is unclear. Here we investigate the post-mitotic effects of different mitotic aberrations—misaligned chromosomes produced by CENP-E inhibition and monopolar spindles resulting from Eg5 inhibition. Eg5 inhibition in cells with an impaired spindle assembly checkpoint (SAC) induces polyploidy through cytokinesis failure without a strong anti-proliferative effect. In contrast, CENP-E inhibition causes p53-mediated post-mitotic apoptosis triggered by chromosome missegregation. Pharmacological studies reveal that aneuploidy caused by the CENP-E inhibitor, Compound-A, in SAC-attenuated cells causes substantial proteotoxic stress and DNA damage. Polyploidy caused by the Eg5 inhibitor does not produce this effect. Furthermore, p53-mediated post-mitotic apoptosis is accompanied by aneuploidy-associated DNA damage response and unfolded protein response activation. Because Compound-A causes p53 accumulation and antitumour activity in an SAC-impaired xenograft model, CENP-E inhibitors could be potential anticancer drugs effective against SAC-impaired tumours. CENP-E regulates chromosome alignment during mitosis to distribute chromosomes equally into daughter cells. Here, the authors show that CENP-E inhibition causes p53-mediated post-mitotic apoptosis in tumours where the spindle assembly checkpoint is compromised, suggesting that CENP-E is a therapeutic target for these cancers.
Collapse
Affiliation(s)
- Akihiro Ohashi
- Oncology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Momoko Ohori
- Oncology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Kenichi Iwai
- Oncology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Yusuke Nakayama
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Tadahiro Nambu
- Oncology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Daisuke Morishita
- Oncology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Tomohiro Kawamoto
- Biomolecular Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Maki Miyamoto
- DMPK Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Takaharu Hirayama
- Oncology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Masanori Okaniwa
- Oncology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Hiroshi Banno
- Oncology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Tomoyasu Ishikawa
- Oncology Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Hitoshi Kandori
- Drug Safety Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| | - Kentaro Iwata
- DMPK Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa 251-8555, Japan
| |
Collapse
|
144
|
Krem MM, Press OW, Horwitz MS, Tidwell T. Mechanisms and clinical applications of chromosomal instability in lymphoid malignancy. Br J Haematol 2015; 171:13-28. [PMID: 26018193 DOI: 10.1111/bjh.13507] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Lymphocytes are unique among cells in that they undergo programmed DNA breaks and translocations, but that special property predisposes them to chromosomal instability (CIN), a cardinal feature of neoplastic lymphoid cells that manifests as whole chromosome- or translocation-based aneuploidy. In several lymphoid malignancies translocations may be the defining or diagnostic markers of the diseases. CIN is a cornerstone of the mutational architecture supporting lymphoid neoplasia, though it is perhaps one of the least understood components of malignant transformation in terms of its molecular mechanisms. CIN is associated with prognosis and response to treatment, making it a key area for impacting treatment outcomes and predicting prognoses. Here we will review the types and mechanisms of CIN found in Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma and the lymphoid leukaemias, with emphasis placed on pathogenic mutations affecting DNA recombination, replication and repair; telomere function; and mitotic regulation of spindle attachment, centrosome function, and chromosomal segregation. We will discuss the means by which chromosome-level genetic aberrations may give rise to multiple pathogenic mutations required for carcinogenesis and conclude with a discussion of the clinical applications of CIN and aneuploidy to diagnosis, prognosis and therapy.
Collapse
Affiliation(s)
- Maxwell M Krem
- Department of Medicine and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Oliver W Press
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Marshall S Horwitz
- Department of Pathology and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Timothy Tidwell
- Department of Pathology and Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
145
|
How C, Bruce J, So J, Pintilie M, Haibe-Kains B, Hui A, Clarke BA, Hedley DW, Hill RP, Milosevic M, Fyles A, Liu FF. Chromosomal instability as a prognostic marker in cervical cancer. BMC Cancer 2015; 15:361. [PMID: 25944123 PMCID: PMC4433070 DOI: 10.1186/s12885-015-1372-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/27/2015] [Indexed: 01/10/2023] Open
Abstract
Background Cervical cancer is the third most common cancer in women globally, and despite treatment, distant metastasis and nodal recurrence will still develop in approximately 30% of patients. The ability to predict which patients are likely to experience distant relapse would allow clinicians to better tailor treatment. Previous studies have investigated the role of chromosomal instability (CIN) in cancer, which can promote tumour initiation and growth; a hallmark of human malignancies. In this study, we sought to examine the published CIN70 gene signature in a cohort of cervical cancer patients treated at the Princess Margaret (PM) Cancer Centre and an independent cohort of The Cancer Genome Atlas (TCGA) cervical cancer patients, to determine if this CIN signature associated with patient outcome. Methods Cervical cancer samples were collected from 79 patients, treated between 2000–2007 at the PM, prior to undergoing curative chemo-radiation. Total RNA was extracted from each patient sample and analyzed using the GeneChip Human Genome U133 Plus 2.0 array (Affymetrix). Results High CIN70 scores were significantly related to increased chromosomal alterations in TCGA cervical cancer patients, including a higher percentage of genome altered and a higher number of copy number alterations. In addition, this same CIN70 signature was shown to be predictive of para-aortic nodal relapse in the PM Cancer Centre cohort. Conclusions These findings demonstrate that chromosomal instability plays an important role in cervical cancer, and is significantly associated with patient outcome. For the first time, this CIN70 gene signature provided prognostic value for patients with cervical cancer.
Collapse
Affiliation(s)
- Christine How
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Jeff Bruce
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Jonathan So
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| | - Melania Pintilie
- Division of Biostatistics, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Medical Biophysics Department, University of Toronto, Toronto, ON, Canada.
| | - Angela Hui
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Blaise A Clarke
- Department of Pathology, University Health Network, Toronto, ON, Canada.
| | - David W Hedley
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
| | - Richard P Hill
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Michael Milosevic
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| | - Anthony Fyles
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| | - Fei-Fei Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
146
|
Thompson LL, McManus KJ. A novel multiplexed, image-based approach to detect phenotypes that underlie chromosome instability in human cells. PLoS One 2015; 10:e0123200. [PMID: 25893404 PMCID: PMC4404342 DOI: 10.1371/journal.pone.0123200] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/19/2015] [Indexed: 11/30/2022] Open
Abstract
Chromosome instability (CIN) is characterized by a progressive change in chromosome numbers. It is a characteristic common to virtually all tumor types, and is commonly observed in highly aggressive and drug resistant tumors. Despite this information, the majority of human CIN genes have yet to be elucidated. In this study, we developed and validated a multiplexed, image-based screen capable of detecting three different phenotypes associated with CIN. Large-scale chromosome content changes were detected by quantifying changes in nuclear volumes following RNAi-based gene silencing. Using a DsRED-LacI reporter system to fluorescently label chromosome 11 within a human fibrosarcoma cell line, we were able to detect deviations from the expected number of two foci per nucleus (one focus/labelled chromosome) that occurred following CIN gene silencing. Finally, micronucleus enumeration was performed, as an increase in micronucleus formation is a classic hallmark of CIN. To validate the ability of each assay to detect phenotypes that underlie CIN, we silenced the established CIN gene, SMC1A. Following SMC1A silencing we detected an increase in nuclear volumes, a decrease in the number of nuclei harboring two DsRED-LacI foci, and an increase in micronucleus formation relative to controls (untreated and siGAPDH). Similar results were obtained in an unrelated human fibroblast cell line. The results of this study indicate that each assay is capable of detecting CIN-associated phenotypes, and can be utilized in future experiments to uncover novel human CIN genes, which will provide novel insight into the pathogenesis of cancer.
Collapse
Affiliation(s)
- Laura L. Thompson
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- Manitoba Institute of Cell Biology, Winnipeg, Manitoba, Canada
| | - Kirk J. McManus
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
- Manitoba Institute of Cell Biology, Winnipeg, Manitoba, Canada
- * E-mail:
| |
Collapse
|
147
|
Skp2-macroH2A1-CDK8 axis orchestrates G2/M transition and tumorigenesis. Nat Commun 2015; 6:6641. [PMID: 25818643 PMCID: PMC4500169 DOI: 10.1038/ncomms7641] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/13/2015] [Indexed: 12/14/2022] Open
Abstract
Understanding the mechanism by which cell growth, migration, polyploidy, and tumorigenesis are regulated may provide important therapeutic strategies for cancer therapy. Here we identify the Skp2-macroH2A1 (mH2A1)-cyclin-dependent kinase 8 (CDK8) axis as a critical pathway for these processes, and deregulation of this pathway is associated with human breast cancer progression and patient survival outcome. We showed that mH2A1 is a new substrate of Skp2 SCF complex whose degradation by Skp2 promotes CDK8 gene and protein expression. Strikingly, breast tumour suppression on Skp2 deficiency can be rescued by mH2A1 knockdown or CDK8 restoration using mouse tumour models. We further show that CDK8 regulates p27 protein expression by facilitating Skp2-mediated p27 ubiquitination and degradation. Our study establishes a critical role of Skp2-mH2A1-CDK8 axis in breast cancer development and targeting this pathway offers a promising strategy for breast cancer therapy.
Collapse
|
148
|
Chen K, Xia G, Zhang C, Sun Y. Correlation between smoking history and molecular pathways in sporadic colorectal cancer: a meta-analysis. Int J Clin Exp Med 2015; 8:3241-3257. [PMID: 26064214 PMCID: PMC4443048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/20/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Epidemiological studies have shown that smoking increases the risk for colorectal cancer (CRC). Evidence of the guiding significance of smoking history for molecular classification and molecular targeted anti-tumor therapy is not well established. AIMS To provide indirectly evidence, we conducted a systematic meta-analysis of association between smoking history and different molecular classification. METHODS We searched in multiple databases up to January 2014, and identified 27 eligible studies. All studies were divided into seven groups based on different molecular alteration categories, which are MSI, CIMP, and three molecular pathway-associated gene alterations (APC, KRAS, P53, BRAF mutation, and APC methylation). Crude odds ratios (ORs) and their 95% confidence intervals (CIs) were calculated to evaluate the association. RESULTS Smoking showed a significantly positive correlation with P53 mutation (exons 4 to 8), BRAF (codon 600) mutation, MSI positivity, and CIMP positivity, with ORs of 1.25 (95% CI: 1.07-1.45), 1.41 (95% CI: 1.18-1.68), 1.28 (95% CI: 1.12-1.47), and 1.23 (95% CI: 1.01-1.50), respectively. However, smoking was not positively correlated with APC (mutation cluster region) and KRAS (codons 12 and 13) mutation in sporadic CRC patients. CONCLUSIONS These findings suggested smoking history occurred with P53 mutation, BRAF mutation, MSI positivity, and CIMP positivity in sporadic CRCs; and could guide those specifically therapeutic designs when molecular classification with genetic test was infeasible. More associated studies should be conducted for strengthening and renewing the current result.
Collapse
Affiliation(s)
- Ke Chen
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University of MedicineShanghai 200025, China
| | - Guanggai Xia
- Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou 510089, China
| | - Changhua Zhang
- Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou 510089, China
| | - Yunwei Sun
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University of MedicineShanghai 200025, China
| |
Collapse
|
149
|
Bochtler T, Fröhling S, Krämer A. Role of chromosomal aberrations in clonal diversity and progression of acute myeloid leukemia. Leukemia 2015; 29:1243-52. [PMID: 25673237 DOI: 10.1038/leu.2015.32] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 11/24/2014] [Accepted: 12/18/2014] [Indexed: 12/20/2022]
Abstract
Genetic abnormalities are a hallmark of cancer. Hereby, cytogenetic aberrations and small-scale abnormalities, such as single-nucleotide variations and insertion/deletion mutations, have emerged as two alternative modes of genetic diversification. Both mechanisms are at work in acute myeloid leukemia (AML), in which conventional karyotyping and molecular studies demonstrate that gene mutations occur predominantly in cytogenetically normal AML, whereas chromosomal changes are a driving force of development and progression of disease in aberrant karyotype AML. All steps of disease evolution in AML, ranging from the transformation of preleukemic clones into overt leukemia to the expansion and recurrence of malignant clones, are paralleled by clonal evolution at either the gene mutation or chromosome aberration level. Preleukemic conditions, such as Fanconi anemia and Bloom syndrome, demonstrate that the acquisition of chromosomal aberrations can contribute to leukemic transformation. Similar to what has been shown at the mutational level, expansion and recurrence of AML clones goes along with increasing genetic diversification. Hereby, cytogenetically more evolved subclones are at a proliferative advantage and outgrow ancestor clones or have evolved toward a more aggressive behavior with additional newly acquired aberrations as compared with the initial leukemic clone, respectively.
Collapse
Affiliation(s)
- T Bochtler
- 1] Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany [2] Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - S Fröhling
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Krämer
- 1] Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany [2] Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
150
|
Role of NEK2A in human cancer and its therapeutic potentials. BIOMED RESEARCH INTERNATIONAL 2015; 2015:862461. [PMID: 25705694 PMCID: PMC4330945 DOI: 10.1155/2015/862461] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/14/2014] [Indexed: 02/08/2023]
Abstract
Chromosome instability (CIN) has been identified as a common feature of most human cancers. A number of centrosomal kinases are thought to cause CIN in cancer cells. Part of those centrosomal kinases exhibit elevated expression in a wide variety of tumours and cancer cell lines. Additionally, critical roles in many aspects of cancer cell growth, proliferation, metastasis, and drug resistance have been assigned to some of these centrosomal kinases, such as polo-like kinase 1 (PLk1) and Aurora-A kinase. Recent studies from our group and others revealed that a centrosomal kinase, Never in Mitosis (NIMA) Related Kinase 2A (NEK2A), is frequently upregulated in multiple types of human cancers. Uncontrolled activity of NEK2A activates several oncogenic pathways and ABC transporters, thereby leading to CIN, cancer cell proliferation, metastasis, and enhanced drug resistance. In this paper, we highlight recent findings on the aberrant expression and functional significance of NEK2A in human cancers and emphasize their significance for therapeutic potentials.
Collapse
|