1
|
Liu D, Sun W, Han J, Wang C, Chen D, Wu Y, Chang Y, Yang B. Proto-oncogene DEK binds to pre-mRNAs and regulates the alternative splicing of Hippo signaling genes in HeLa cells. Mol Genet Genomics 2025; 300:31. [PMID: 40075046 PMCID: PMC11903584 DOI: 10.1007/s00438-025-02226-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 01/06/2025] [Indexed: 03/14/2025]
Abstract
Our study aimed to explore how DEK, a carcinogenic protein with chromatin architectural function, genome-widely binds to RNA and affects the alternative splicing in cancer cells to decipher its molecular functions. To achieve this goal, cell phenotype experiments, RNA sequencing (RNA-seq), and improved RNA immunoprecipitation sequencing (iRIP-seq) were conducted to identify the function and regulated targets of DEK in HeLa cells. The results showed DEK overexpression promoted cell proliferation and invasion of HeLa cells. Meanwhile, DEK hardly affected transcript level expression of those high expressed genes, but splicing pattern of 411 genes was regulated by DEK in HeLa cells, which were enriched in Hippo signaling pathway. Moreover, DEK broadly bind the RNA of a total of 11, 112 genes, with a biased binding the 5' splice site (5'SS) consensus GGUAA motifs at the CDS and intronic regions. In addition, 297 DEK-binding genes showed different splicing pattern after DEK overexpression in HeLa cells. These genes were enriched in Hippo signaling pathway including CSNK1D. The RT-qPCR and RIP-PCR confirmed that DEK can bind to CSNK1D to regulate its alternative splicing in HeLa cells. In summary, our results indicated DEK could broadly bind and regulate the pre-mRNA splicing process, which provide new insights of mechanisms that DEK functions in various biological processes including cancer.
Collapse
Affiliation(s)
- Dongbo Liu
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Wei Sun
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jing Han
- Cancer Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Cong Wang
- Science Department, Wuhan Ruixing Biotechnology Co. Ltd., Wuhan, Hubei, 430075, China
| | - Dong Chen
- Science Department, Wuhan Ruixing Biotechnology Co. Ltd., Wuhan, Hubei, 430075, China
| | - Yunfei Wu
- Center for Genome Analysis, Wuhan Ruixing Biotechnology Co. Ltd., Wuhan, Hubei, 430075, China
| | - Yongjie Chang
- Center for Genome Analysis, Wuhan Ruixing Biotechnology Co. Ltd., Wuhan, Hubei, 430075, China
| | - Bin Yang
- Department of Thoracic Oncology, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, No. 116 Zhuodaoquan South Rd, Wuhan, Hubei, 430079, China.
| |
Collapse
|
2
|
Hart SA, Hang JF, Chernock RD, Mikula MW, Rooper L, Amin SE, Saluja K, Bishop JA, Chen YH, Cipriani NA, David SN, Dupont WD, Plummer WD, Ferrer KT, Geromes A, Hsieh MS, Hernandez-Prera JC, Kuo YJ, Sasaki E, Shi Q, Truong T, Velez Torres JM, Lewis JS. DEK :: AFF2 Fusion Sinonasal and Skull Base Nonkeratinizing Squamous Cell Carcinoma : A Clinical Outcome Study Compared With Conventional Sinonasal Squamous Cell Carcinoma. Am J Surg Pathol 2025; 49:130-137. [PMID: 39719672 DOI: 10.1097/pas.0000000000002335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
DEK :: AFF2 fusion nonkeratinizing squamous cell carcinoma (NKSCC) is an emerging entity in the sinonasal tract, temporal bone, and skull base. However, the clinical behavior of these tumors has not been well studied. Here, we report the largest cohort of DEK :: AFF2 carcinomas to determine if morphology, mitotic rate, and/or Ki-67 IHC are associated with patient outcomes, including a comparison with high-risk human papillomavirus (HPV)-associated and independent patients. We solicited cases of molecularly or AFF2 immunohistochemistry (IHC) proven DEK :: AFF2 SCC from surgical pathologists to collect patient demographic, clinical, and outcome data. Using representative H&E slides, we characterized the morphology and counted mitoses. Ki-67 immunohistochemistry was performed. We also compared the DEK :: AFF2 survival rates to those in a cohort of AFF2 IHC-negative HPV-associated and HPV-independent SCC. DEK :: AFF2 carcinomas most commonly arose in the nasal cavity (13/30, 43%), and the average number of recurrences was 1.8 (range: 0 to 10). At the last follow-up, most patients were disease free (19/30, 63%) or were alive with disease (9/30, 30%). There was an average mitotic rate of 2 per 2 mm 2 (range: 0 to 9) and Ki-67 proliferation rate of 26% (range: 3% to 60%). Local recurrence was common, but morphology, mitotic activity, and Ki-67 index were not associated with recurrence or survival. On Kaplan-Meier survival analysis, DEK :: AFF2 patients had lower disease-free survival but otherwise had similar outcomes to conventional SCC patients. Our multi-institutional study shows that local recurrence is common in DEK :: AFF2 fusion nonkeratinizing SCC patients, but patients have survival rates similar to conventional SCC. Despite showing a range of different features and proliferation rates, traditional grading by morphology, mitotic rate, and/or Ki-67 activity does not seem to be predictive of outcome.
Collapse
MESH Headings
- Humans
- Female
- Male
- Middle Aged
- Poly-ADP-Ribose Binding Proteins/genetics
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Aged
- Adult
- Paranasal Sinus Neoplasms/pathology
- Paranasal Sinus Neoplasms/mortality
- Paranasal Sinus Neoplasms/chemistry
- Paranasal Sinus Neoplasms/genetics
- Paranasal Sinus Neoplasms/therapy
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/chemistry
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/therapy
- Immunohistochemistry
- Oncogene Proteins/genetics
- Skull Base Neoplasms/pathology
- Skull Base Neoplasms/mortality
- Skull Base Neoplasms/chemistry
- Skull Base Neoplasms/genetics
- Skull Base Neoplasms/therapy
- Chromosomal Proteins, Non-Histone/genetics
- Ki-67 Antigen/analysis
- Mitotic Index
- Aged, 80 and over
- Oncogene Proteins, Fusion/genetics
- Nuclear Proteins/genetics
- Squamous Cell Carcinoma of Head and Neck
Collapse
Affiliation(s)
- Stephanie A Hart
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
| | - Jen-Fan Hang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei
| | | | - Michael W Mikula
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston
| | - Lisa Rooper
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, Houston
| | - Sara E Amin
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX
| | - Karan Saluja
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX
| | - Justin A Bishop
- Department of Pathology, Chang Gung Memorial Hospital, Linkou, Taoyuan City, Taiwan
| | - Yu Hsiu Chen
- Department of Pathology, University of Chicago, Chicago, IL
| | | | | | - William D Dupont
- Department of Pathology and Laboratory, Stroger Hospital of Cook County, Chicago, IL
| | - W Dale Plummer
- Department of Pathology and Laboratory, Stroger Hospital of Cook County, Chicago, IL
| | | | - Ariana Geromes
- Department of Pathology, National Taiwan University Hospital
| | - Min-Shu Hsieh
- Department of Pathology, Lee Moffitt Cancer Center and Research Institute, Tampa
| | | | - Ying-Ju Kuo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei
| | - Eiichi Sasaki
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Qiuying Shi
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Tra Truong
- Department of Laboratory Medicine and Pathobiology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Jaylou M Velez Torres
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - James S Lewis
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ
| |
Collapse
|
3
|
Johnstone M, Leck A, Lange T, Wilcher K, Shephard MS, Paranjpe A, Schutte S, Wells S, Kappes F, Salomonis N, Privette Vinnedge LM. The chromatin remodeler DEK promotes proliferation of mammary epithelium and is associated with H3K27me3 epigenetic modifications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612116. [PMID: 39314335 PMCID: PMC11419013 DOI: 10.1101/2024.09.09.612116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The DEK chromatin remodeling protein was previously shown to confer oncogenic phenotypes to human and mouse mammary epithelial cells using in vitro and knockout mouse models. However, its functional role in normal mammary gland epithelium remained unexplored. We developed two novel mouse models to study the role of Dek in normal mammary gland biology in vivo . Mammary gland-specific Dek over-expression in mice resulted in hyperproliferation of cells that visually resembled alveolar cells, and a transcriptional profile that indicated increased expression of cell cycle, mammary stem/progenitor, and lactation-associated genes. Conversely, Dek knockout mice exhibited an alveologenesis or lactation defect, resulting in dramatically reduced pup survival. Analysis of previously published single-cell RNA-sequencing of mouse mammary glands revealed that Dek is most highly expressed in mammary stem cells and alveolar progenitor cells, and to a lesser extent in basal epithelial cells, supporting the observed phenotypes. Mechanistically, we discovered that Dek is a modifier of Ezh2 methyltransferase activity, upregulating the levels of histone H3 trimethylation on lysine 27 (H3K27me3) to control gene transcription. Combined, this work indicates that Dek promotes proliferation of mammary epithelial cells via cell cycle deregulation. Furthermore, we report a novel function for Dek in alveologenesis and histone H3 K27 trimethylation.
Collapse
|
4
|
Pierzynska-Mach A, Diaspro A, Cella Zanacchi F. Super-resolution microscopy reveals the nanoscale cluster architecture of the DEK protein cancer biomarker. iScience 2023; 26:108277. [PMID: 38026229 PMCID: PMC10660485 DOI: 10.1016/j.isci.2023.108277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 09/02/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
DEK protein, a key chromatin regulator, is strongly overexpressed in various forms of cancer. While conventional microscopy revealed DEK as uniformly distributed within the cell nucleus, advanced super-resolution techniques uncovered cluster-like structures. However, a comprehensive understanding of DEK's cellular distribution and its implications in cancer and cell growth remained elusive. To bridge this gap, we employed single-molecule localization microscopy (SMLM) to dissect DEK's nanoscale organization in both normal-like and aggressive breast cancer cell lines. Our investigation included characteristics such as localizations per cluster, cluster areas, and intra-cluster localization densities (ICLDs). We elucidated how cluster features align with different breast cell types and how chromatin decompaction influences DEK clusters in these contexts. Our results indicate that DEK's intra-cluster localization density and nano-organization remain preserved and not significantly influenced by protein overexpression or chromatin compaction changes. This study advances the understanding of DEK's role in cancer and underscores its stable nanoscale behavior.
Collapse
Affiliation(s)
| | - Alberto Diaspro
- Nanoscopy and NIC@IIT, Istituto Italiano di Tecnologia, 16152 Genoa, Italy
- Department of Physics (DIFILAB), Department of Physics, University of Genoa, 16146 Genoa, Italy
| | - Francesca Cella Zanacchi
- Nanoscopy and NIC@IIT, Istituto Italiano di Tecnologia, 16152 Genoa, Italy
- Physics Department E. Fermi, University of Pisa, 56127 Pisa, Italy
- Centro per l’Integrazione della Strumentazione dell’Università di Pisa (CISUP), University of Pisa, 56127 Pisa, Italy
| |
Collapse
|
5
|
Wilcher KE, Page ERH, Privette Vinnedge LM. The impact of the chromatin binding DEK protein in hematopoiesis and acute myeloid leukemia. Exp Hematol 2023; 123:18-27. [PMID: 37172756 PMCID: PMC10330528 DOI: 10.1016/j.exphem.2023.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/03/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
Hematopoiesis is an exquisitely regulated process of cellular differentiation to create diverse cell types of the blood. Genetic mutations, or aberrant regulation of gene transcription, can interrupt normal hematopoiesis. This can have dire pathological consequences, including acute myeloid leukemia (AML), in which generation of the myeloid lineage of differentiated cells is interrupted. In this literature review, we discuss how the chromatin remodeling DEK protein can control hematopoietic stem cell quiescence, hematopoietic progenitor cell proliferation, and myelopoiesis. We further discuss the oncogenic consequences of the t(6;9) chromosomal translocation, which creates the DEK-NUP214 (aka: DEK-CAN) fusion gene, during the pathogenesis of AML. Combined, the literature indicates that DEK is crucial for maintaining homeostasis of hematopoietic stem and progenitor cells, including myeloid progenitors.
Collapse
Affiliation(s)
- Katherine E Wilcher
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Current: Wright State University Boonshoft School of Medicine, Fairborn, OH
| | - Evan R H Page
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Lisa M Privette Vinnedge
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH.
| |
Collapse
|
6
|
Guo H, Prell M, Königs H, Xu N, Waldmann T, Hermans-Sachweh B, Ferrando-May E, Lüscher B, Kappes F. Bacterial Growth Inhibition Screen (BGIS) identifies a loss-of-function mutant of the DEK oncogene, indicating DNA modulating activities of DEK in chromatin. FEBS Lett 2021; 595:1438-1453. [PMID: 33686684 DOI: 10.1002/1873-3468.14070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
The DEK oncoprotein regulates cellular chromatin function via a number of protein-protein interactions. However, the biological relevance of its unique pseudo-SAP/SAP-box domain, which transmits DNA modulating activities in vitro, remains largely speculative. As hypothesis-driven mutations failed to yield DNA-binding null (DBN) mutants, we combined random mutagenesis with the Bacterial Growth Inhibition Screen (BGIS) to overcome this bottleneck. Re-expression of a DEK-DBN mutant in newly established human DEK knockout cells failed to reduce the increase in nuclear size as compared to wild type, indicating roles for DEK-DNA interactions in cellular chromatin organization. Our results extend the functional roles of DEK in metazoan chromatin and highlight the predictive ability of recombinant protein toxicity in E. coli for unbiased studies of eukaryotic DNA modulating protein domains.
Collapse
Affiliation(s)
- Haihong Guo
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
| | - Malte Prell
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
| | - Hiltrud Königs
- Institute of Pathology, Medical School, RWTH Aachen University, Germany
| | - Nengwei Xu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Dushu Lake Higher Education Town, Suzhou Industrial Park, China
| | - Tanja Waldmann
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Germany
| | | | - Elisa Ferrando-May
- Bioimaging Center, Department of Biology, University of Konstanz, Germany
| | - Bernhard Lüscher
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
| | - Ferdinand Kappes
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Dushu Lake Higher Education Town, Suzhou Industrial Park, China
| |
Collapse
|
7
|
Guo H, Xu N, Prell M, Königs H, Hermanns-Sachweh B, Lüscher B, Kappes F. Bacterial Growth Inhibition Screen (BGIS): harnessing recombinant protein toxicity for rapid and unbiased interrogation of protein function. FEBS Lett 2021; 595:1422-1437. [PMID: 33704777 DOI: 10.1002/1873-3468.14072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/28/2021] [Accepted: 03/02/2021] [Indexed: 12/13/2022]
Abstract
In two proof-of-concept studies, we established and validated the Bacterial Growth Inhibition Screen (BGIS), which explores recombinant protein toxicity in Escherichia coli as a largely overlooked and alternative means for basic characterization of functional eukaryotic protein domains. By applying BGIS, we identified an unrecognized RNA-interacting domain in the DEK oncoprotein (this study) and successfully combined BGIS with random mutagenesis as a screening tool for loss-of-function mutants of the DNA modulating domain of DEK [1]. Collectively, our findings shed new light on the phenomenon of recombinant protein toxicity in E. coli. Given the easy and rapid implementation and wide applicability, BGIS will extend the repertoire of basic methods for the identification, analysis and unbiased manipulation of proteins.
Collapse
Affiliation(s)
- Haihong Guo
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
| | - Nengwei Xu
- Department of Biological Sciences, Suzhou Dushu Lake Science and Education Innovation District, Suzhou Industrial Park, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Malte Prell
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
| | - Hiltrud Königs
- Institute of Pathology, Medical School, RWTH Aachen University, Germany
| | | | - Bernhard Lüscher
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
| | - Ferdinand Kappes
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
- Department of Biological Sciences, Suzhou Dushu Lake Science and Education Innovation District, Suzhou Industrial Park, Xi'an Jiaotong-Liverpool University, Suzhou, China
| |
Collapse
|
8
|
Yi H, Duan H, Shi W, Liu Z, Liu Y. DEK overexpression is predictive of poor prognosis in esophageal squamous cell carcinoma. Arch Med Sci 2021; 17:135-141. [PMID: 33488866 PMCID: PMC7811301 DOI: 10.5114/aoms.2019.84210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/09/2019] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION The DEK gene encodes a nuclear phosphoprotein which is involved in multiple cell metabolic processes, such as DNA damage repair, mRNA splicing, modifying chromatin structure and transcription regulation. DEK has been shown to be overexpressed in various solid human tumors and associated with patient prognosis. In this study, our aim was to investigate DEK protein expression and its relationship with clinicopathological parameters and prognosis in esophageal squamous cell carcinoma (ESCC). MATERIAL AND METHODS Tissue samples were collected from 120 routinely diagnosed ESCC patients who underwent surgical resection at the Zhongshan Hospital, Xiamen University in the period from June 2011 to May 2013. The expression of DEK was determined by immunohistochemistry. RESULTS DEK protein was ubiquitously distributed in the nucleus of ESCC cells, and its positive rate (71.7%) was significantly higher in cancer samples than those of para-carcinoma (21.4%) or normal esophageal (13.9%) tissues (p < 0.001). Similarly, significantly more cells overexpressing DEK were found in ESCC tissues (57.5%) in comparison with para-carcinoma samples (11.4%) and normal esophageal mucosa (0%, p < 0.001). The DEK overexpression rate was significantly different between patients with different tumor-node-metastasis (TNM) stages and differentiation degrees (p < 0.001). ESCC cases with elevated DEK amounts showed reduced disease-free and 5-year survival rates compared with those expressing low DEK amounts (p < 0.001). DEK overexpression was also confirmed to independently predict prognosis in ESCC (HR = 4.121, 95% CI: 1.803-9.42, p = 0.001). CONCLUSIONS DEK expression is positively correlated with reduced survival in ESCC patients. DEK has potential to be an independent biomarker in predicting prognosis of ESCC patients.
Collapse
Affiliation(s)
- Huochun Yi
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Hongbing Duan
- Department of Thoracic, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wensheng Shi
- Center of Clinical Laboratory, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhengjin Liu
- Department of Pathology, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yali Liu
- Department of Thoracic, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
9
|
Wang J, Wen T, Li Z, Che X, Gong L, Jiao Z, Qu X, Liu Y. CD36 upregulates DEK transcription and promotes cell migration and invasion via GSK-3β/β-catenin-mediated epithelial-to-mesenchymal transition in gastric cancer. Aging (Albany NY) 2020; 13:1883-1897. [PMID: 33232276 PMCID: PMC7880392 DOI: 10.18632/aging.103985] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/08/2020] [Indexed: 12/15/2022]
Abstract
Evidence indicates that the lipid scavenger receptor CD36 has pro-metastatic functions in several cancers. Although CD36 expression correlates with an unfavorable prognosis in gastric cancer (GC), its specific contribution to disease onset, progression, and/or metastasis remains unclear. Using bioinformatics analyses, we ascertained that CD36 expression was increased in metastatic GC specimens in The Cancer Genome Atlas and Gene Expression Omnibus databases and correlated with poor prognosis. In addition, higher CD36 expression was associated with lymph node metastasis (p < 0.05) and poor prognosis (p = 0.030) in 79 Chinese GC patients. Basal CD36 expression levels correlated positively with migration, invasion, and expression of epithelial-to-mesenchymal transition (EMT) markers in GC cell lines, a relationship confirmed by knockdown and overexpression experiments. Importantly, analysis of gene expression changes in CD36-knockdown GC cells led us to identify the chromatin-associated protein DEK as a c-Myc target that mediates activation of the GSK-3β/β-catenin signaling pathway to trigger EMT. These findings further our understanding of the mechanisms governing metastatic dissemination of GC cells and suggest the therapeutic potential of strategies targeting CD36.
Collapse
Affiliation(s)
- Jin Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ti Wen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhi Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang 110001, China
| | - Libao Gong
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zihan Jiao
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Province Clinical Research Center for Cancer, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
10
|
Liu T, Zhu G, Yan W, Lv Y, Wang X, Jin G, Cui M, Lin Z, Ren X. Cordycepin Inhibits Cancer Cell Proliferation and Angiogenesis through a DEK Interaction via ERK Signaling in Cholangiocarcinoma. J Pharmacol Exp Ther 2020; 373:279-289. [PMID: 32102917 DOI: 10.1124/jpet.119.263202] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/10/2020] [Indexed: 12/17/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant tumor that arises from the epithelial cells of the bile duct and is notorious for its poor prognosis. The clinical outcome remains disappointing, and thus more effective therapeutic options are urgently required. Cordycepin, a traditional Chinese medicine, provides multiple pharmacological strategies in antitumors, but its mechanisms have not been fully elucidated. In this study, we reported that cordycepin inhibited the viability and proliferation capacity of CCA cells in a time- and dose-dependent manner determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and colony formation assay. Flow cytometry and Hoechst dye showed that cordycepin induced cancer cell apoptosis via extracellular signal-regulated kinase (ERK) 1/2 deactivation. Moreover, cordycepin significantly reduced the angiogenetic capabilities of CCA in vitro as examined by tube formation assay. We also discovered that cordycepin inhibited DEK expression by using Western blot assay. DEK serves as an oncogenic protein that is overexpressed in various gastrointestinal tumors. DEK silencing inhibited CCA cell viability and angiogenesis but not apoptosis induction determined by Western blot and flow cytometry. Furthermore, cordycepin significantly inhibited tumor growth and angiogenic capacities in a xenograft model by downregulating the expression of DEK, phosphorylated ERK1/2 CD31 and von Willebrand factor (vWF). Taken together, we demonstrated that cordycepin inhibited CCA cell proliferation and angiogenesis with a DEK interaction via downregulation in ERK signaling. These data indicate that cordycepin may serve as a novel agent for CCA clinical treatment and prognosis improvement. SIGNIFICANCE STATEMENT: Cordycepin provides multiple strategies in antitumors, but its mechanisms are not fully elucidated, especially on cholangiocarcinoma (CCA). We reported that cordycepin inhibited the viability of CCA cells, induced apoptosis via extracellular signal-regulated kinase 1/2 deactivation and DEK inhibition, and reduced the angiogenetic capabilities of CCA both in vivo and in vitro.
Collapse
Affiliation(s)
- Tesi Liu
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules (Yanbian University), Ministry of Education, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); and Otorhinolaryngology Institute at Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, P.R. China (T.L.)
| | - Guang Zhu
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules (Yanbian University), Ministry of Education, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); and Otorhinolaryngology Institute at Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, P.R. China (T.L.)
| | - Wendi Yan
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules (Yanbian University), Ministry of Education, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); and Otorhinolaryngology Institute at Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, P.R. China (T.L.)
| | - You Lv
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules (Yanbian University), Ministry of Education, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); and Otorhinolaryngology Institute at Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, P.R. China (T.L.)
| | - Xue Wang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules (Yanbian University), Ministry of Education, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); and Otorhinolaryngology Institute at Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, P.R. China (T.L.)
| | - Guang Jin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules (Yanbian University), Ministry of Education, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); and Otorhinolaryngology Institute at Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, P.R. China (T.L.)
| | - Minghua Cui
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules (Yanbian University), Ministry of Education, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); and Otorhinolaryngology Institute at Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, P.R. China (T.L.)
| | - Zhenhua Lin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules (Yanbian University), Ministry of Education, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); and Otorhinolaryngology Institute at Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, P.R. China (T.L.)
| | - Xiangshan Ren
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules (Yanbian University), Ministry of Education, Yanji, China (T.L., G.Z., W.Y., Y.L., X.W., G.J., M.C., Z.L., X.R.); and Otorhinolaryngology Institute at Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, P.R. China (T.L.)
| |
Collapse
|
11
|
Altered miRNA and mRNA Expression in Sika Deer Skeletal Muscle with Age. Genes (Basel) 2020; 11:genes11020172. [PMID: 32041309 PMCID: PMC7073773 DOI: 10.3390/genes11020172] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
Studies of the gene and miRNA expression profiles associated with the postnatal late growth, development, and aging of skeletal muscle are lacking in sika deer. To understand the molecular mechanisms of the growth and development of sika deer skeletal muscle, we used de novo RNA sequencing (RNA-seq) and microRNA sequencing (miRNA-seq) analyses to determine the differentially expressed (DE) unigenes and miRNAs from skeletal muscle tissues at 1, 3, 5, and 10 years in sika deer. A total of 51,716 unigenes, 171 known miRNAs, and 60 novel miRNAs were identified based on four mRNA and small RNA libraries. A total of 2,044 unigenes and 11 miRNAs were differentially expressed between adolescence and juvenile sika deer, 1,946 unigenes and 4 miRNAs were differentially expressed between adult and adolescent sika deer, and 2,209 unigenes and 1 miRNAs were differentially expressed between aged and adult sika deer. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses showed that DE unigenes and miRNA were mainly related to energy and substance metabolism, processes that are closely associate with the growth, development, and aging of skeletal muscle. We also constructed mRNA–mRNA and miRNA–mRNA interaction networks related to the growth, development, and aging of skeletal muscle. The results show that mRNA (Myh1, Myh2, Myh7, ACTN3, etc.) and miRNAs (miR-133a, miR-133c, miR-192, miR-151-3p, etc.) may play important roles in muscle growth and development, and mRNA (WWP1, DEK, UCP3, FUS, etc.) and miRNAs (miR-17-5p, miR-378b, miR-199a-5p, miR-7, etc.) may have key roles in muscle aging. In this study, we determined the dynamic miRNA and unigenes transcriptome in muscle tissue for the first time in sika deer. The age-dependent miRNAs and unigenes identified will offer insights into the molecular mechanism underlying muscle development, growth, and maintenance and will also provide valuable information for sika deer genetic breeding.
Collapse
|
12
|
Jeon S, Song J, Lee D, Kim GT, Park SH, Shin DY, Shin KO, Park K, Shim SM, Park TS. Inhibition of sphingosine 1-phosphate lyase activates human keratinocyte differentiation and attenuates psoriasis in mice. J Lipid Res 2020; 61:20-32. [PMID: 31690639 PMCID: PMC6939600 DOI: 10.1194/jlr.ra119000254] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/29/2019] [Indexed: 11/20/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) lyase is an intracellular enzyme that catalyzes the irreversible degradation of S1P and has been suggested as a therapeutic target for the treatment of psoriasis vulgaris. Because S1P induces differentiation of keratinocytes, we examined whether modulation of S1P lyase and altered intracellular S1P levels regulate proliferation and differentiation of human neonatal epidermal keratinocyte (HEKn) cells. To identify the physiological functions of S1P lyase in skin, we inhibited S1P lyase in HEKn cells with an S1P lyase-specific inhibitor (SLI) and with S1P lyase 1 (SGPL1)-specific siRNA (siSGPL1). In HEKn cells, pharmacological treatment with the SLI caused G1 arrest by upregulation of p21 and p27 and induced keratin 1, an early differentiation marker. Similarly, genetic suppression by siSGPL1 arrested the cell cycle at the G1 phase and activated differentiation. In addition, enzyme suppression by siSGPL1 upregulated keratin 1 and differentiation markers including involucrin and loricrin. When hyperproliferation of HEKn cells was induced by interleukin (IL)-17 and IL-22, pharmacologic inhibition of S1P lyase by SLI decreased proliferation and activated differentiation of HEKn cells simultaneously. In addition, SLI administration ameliorated imiquimod-induced psoriatic symptoms including erythema, scaling, and epidermal thickness in vivo. We thus demonstrated that S1P lyase inhibition reduces cell proliferation and induces keratinocyte differentiation, and that inhibition may attenuate psoriasiform changes. Collectively, these findings suggest that S1P lyase is a modulating factor for proliferation and differentiation, and support its potential as a therapeutic target for psoriasis in human keratinocytes.
Collapse
Affiliation(s)
- Suwon Jeon
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Jaehwi Song
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Dongyup Lee
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Goon-Tae Kim
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Si-Hyun Park
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Dong-Yoon Shin
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Kyong-Oh Shin
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Kyungho Park
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Soon-Mi Shim
- Department of Food Science and Technology, Sejong University, Seoul 05006, Republic of Korea.
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea.
| |
Collapse
|
13
|
DEK terminates diapause by activation of quiescent cells in the crustacean Artemia. Biochem J 2019; 476:1753-1769. [PMID: 31189566 DOI: 10.1042/bcj20190169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 12/24/2022]
Abstract
To cope with harsh environments, the Artemia shrimp produces gastrula embryos in diapause, a state of obligate dormancy, having cellular quiescence and suppressed metabolism. The mechanism behind these cellular events remains largely unknown. Here, we study the regulation of cell quiescence using diapause embryos of Artemia We found that Artemia DEK (Ar-DEK), a nuclear factor protein, was down-regulated in the quiescent cells of diapause embryos and enriched in the activated cells of post-diapause embryos. Knockdown of Ar-DEK induced the production of diapause embryos whereas the control Artemia released free-swimming nuaplii. Our results indicate that Ar-DEK correlated with the termination of cellular quiescence via the increase in euchromatin and decrease in heterochromatin. The phenomena of quiescence have many implications beyond shrimp ecology. In cancer cells, for example, knockdown of DEK also induced a short period of cellular quiescence and increased resistance to environmental stress in MCF-7 and MKN45 cancer cell lines. Analysis of RNA sequences in Artemia and in MCF-7 revealed that the Wnt and AURKA signaling pathways were all down-regulated and the p53 signaling pathway was up-regulated upon inhibition of DEK expression. Our results provide insight into the functions of Ar-DEK in the activation of cellular quiescence during diapause formation in Artemia.
Collapse
|
14
|
Capitano ML, Mor-Vaknin N, Saha AK, Cooper S, Legendre M, Guo H, Contreras-Galindo R, Kappes F, Sartor MA, Lee CT, Huang X, Markovitz DM, Broxmeyer HE. Secreted nuclear protein DEK regulates hematopoiesis through CXCR2 signaling. J Clin Invest 2019; 129:2555-2570. [PMID: 31107242 PMCID: PMC6546479 DOI: 10.1172/jci127460] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
The nuclear protein DEK is an endogenous DNA-binding chromatin factor regulating hematopoiesis. DEK is one of only 2 known secreted nuclear chromatin factors, but whether and how extracellular DEK regulates hematopoiesis is not known. We demonstrated that extracellular DEK greatly enhanced ex vivo expansion of cytokine-stimulated human and mouse hematopoietic stem cells (HSCs) and regulated HSC and hematopoietic progenitor cell (HPC) numbers in vivo and in vitro as determined both phenotypically (by flow cytometry) and functionally (through transplantation and colony formation assays). Recombinant DEK increased long-term HSC numbers and decreased HPC numbers through a mechanism mediated by the CXC chemokine receptor CXCR2 and heparan sulfate proteoglycans (HSPGs) (as determined utilizing Cxcr2-/- mice, blocking CXCR2 antibodies, and 3 different HSPG inhibitors) that was associated with enhanced phosphorylation of ERK1/2, AKT, and p38 MAPK. To determine whether extracellular DEK required nuclear function to regulate hematopoiesis, we utilized 2 mutant forms of DEK: one that lacked its nuclear translocation signal and one that lacked DNA-binding ability. Both altered HSC and HPC numbers in vivo or in vitro, suggesting the nuclear function of DEK is not required. Thus, DEK acts as a hematopoietic cytokine, with the potential for clinical applicability.
Collapse
Affiliation(s)
- Maegan L. Capitano
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nirit Mor-Vaknin
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Anjan K. Saha
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Scott Cooper
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Maureen Legendre
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Haihong Guo
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - Rafael Contreras-Galindo
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Ferdinand Kappes
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University, Suzhou, China
| | - Maureen A. Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher T. Lee
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Xinxin Huang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Disease, University of Michigan, Ann Arbor, Michigan, USA
| | - Hal E. Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
15
|
de Albuquerque Oliveira AC, Kappes F, Martins DBG, de Lima Filho JL. The unique DEK oncoprotein in women's health: A potential novel biomarker. Biomed Pharmacother 2018; 106:142-148. [PMID: 29957464 DOI: 10.1016/j.biopha.2018.06.082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/20/2022] Open
Abstract
Breast and cervical cancer are the first and fourth cancer types with the highest prevalence in women, respectively. The developmental profiles of cancer in women can vary by genetic markers and cellular events. In turn, age and lifestyle influence in the cellular response and also on the cancer progression and relapse. The human DEK protein, a histone chaperone, belongs to a specific subclass of chromatin topology modulators, being involved in the regulation of DNA-dependent processes. These epigenetic mechanisms have dynamic and reversible nature, have been proposed as targets for different treatment approaches, especially in tumor therapy. The expression patterns of DEK vary between healthy and cancer cells. High expression of DEK is associated with poor prognosis in many cancer types, suggesting that DEK takes part in oncogenic activities via different molecular pathways, including inhibition of senescence and apoptosis. The focus of this review was to highlight the role of the DEK protein in these two female cancers.
Collapse
Affiliation(s)
- Ana Cecília de Albuquerque Oliveira
- Molecular Prospecting and Bioinformatics Group - Laboratory of Immunopathology Keizo Asami (LIKA) - Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235 - Cidade Universitária, Recife, PE, Postal Code 50670-901, Brazil
| | - Ferdinand Kappes
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University No 111, Ren Ai Road, Dushu Lake Higher Education Town, Suzhou Industrial Park (SIP), Suzhou, 215123, PR China
| | - Danyelly Bruneska Gondim Martins
- Molecular Prospecting and Bioinformatics Group - Laboratory of Immunopathology Keizo Asami (LIKA) - Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235 - Cidade Universitária, Recife, PE, Postal Code 50670-901, Brazil; Department of Biochemistry - Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235 - Cidade Universitária, Recife, PE, Postal Code 50670-901, Brazil.
| | - José Luiz de Lima Filho
- Molecular Prospecting and Bioinformatics Group - Laboratory of Immunopathology Keizo Asami (LIKA) - Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235 - Cidade Universitária, Recife, PE, Postal Code 50670-901, Brazil; Department of Biochemistry - Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235 - Cidade Universitária, Recife, PE, Postal Code 50670-901, Brazil
| |
Collapse
|
16
|
Matrka MC, Cimperman KA, Haas SR, Guasch G, Ehrman LA, Waclaw RR, Komurov K, Lane A, Wikenheiser-Brokamp KA, Wells SI. Dek overexpression in murine epithelia increases overt esophageal squamous cell carcinoma incidence. PLoS Genet 2018; 14:e1007227. [PMID: 29538372 PMCID: PMC5884580 DOI: 10.1371/journal.pgen.1007227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 04/04/2018] [Accepted: 01/26/2018] [Indexed: 12/12/2022] Open
Abstract
Esophageal cancer occurs as either squamous cell carcinoma (ESCC) or adenocarcinoma. ESCCs comprise almost 90% of cases worldwide, and recur with a less than 15% five-year survival rate despite available treatments. The identification of new ESCC drivers and therapeutic targets is critical for improving outcomes. Here we report that expression of the human DEK oncogene is strongly upregulated in esophageal SCC based on data in the cancer genome atlas (TCGA). DEK is a chromatin-associated protein with important roles in several nuclear processes including gene transcription, epigenetics, and DNA repair. Our previous data have utilized a murine knockout model to demonstrate that Dek expression is required for oral and esophageal SCC growth. Also, DEK overexpression in human keratinocytes, the cell of origin for SCC, was sufficient to cause hyperplasia in 3D organotypic raft cultures that mimic human skin, thus linking high DEK expression in keratinocytes to oncogenic phenotypes. However, the role of DEK over-expression in ESCC development remains unknown in human cells or genetic mouse models. To define the consequences of Dek overexpression in vivo, we generated and validated a tetracycline responsive Dek transgenic mouse model referred to as Bi-L-Dek. Dek overexpression was induced in the basal keratinocytes of stratified squamous epithelium by crossing Bi-L-Dek mice to keratin 5 tetracycline transactivator (K5-tTA) mice. Conditional transgene expression was validated in the resulting Bi-L-Dek_K5-tTA mice and was suppressed with doxycycline treatment in the tetracycline-off system. The mice were subjected to an established HNSCC and esophageal carcinogenesis protocol using the chemical carcinogen 4-nitroquinoline 1-oxide (4NQO). Dek overexpression stimulated gross esophageal tumor development, when compared to doxycycline treated control mice. Furthermore, high Dek expression caused a trend toward esophageal hyperplasia in 4NQO treated mice. Taken together, these data demonstrate that Dek overexpression in the cell of origin for SCC is sufficient to promote esophageal SCC development in vivo.
Collapse
Affiliation(s)
- Marie C. Matrka
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Katherine A. Cimperman
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Sarah R. Haas
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Geraldine Guasch
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS, UMR7258, Institute Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Lisa A. Ehrman
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Ronald R. Waclaw
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Kakajan Komurov
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Adam Lane
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Kathryn A. Wikenheiser-Brokamp
- Division of Pathology & Laboratory Medicine and Perinatal Institute Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center and Department of Pathology & Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | - Susanne I. Wells
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
- Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| |
Collapse
|
17
|
Smith EA, Krumpelbeck EF, Jegga AG, Greis KD, Ali AM, Meetei AR, Wells SI. The nuclear DEK interactome supports multi-functionality. Proteins 2018; 86:88-97. [PMID: 29082557 PMCID: PMC5730476 DOI: 10.1002/prot.25411] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/20/2017] [Accepted: 10/27/2017] [Indexed: 01/01/2023]
Abstract
DEK is an oncoprotein that is overexpressed in many forms of cancer and participates in numerous cellular pathways. Of these different pathways, relevant interacting partners and functions of DEK are well described in regard to the regulation of chromatin structure, epigenetic marks, and transcription. Most of this understanding was derived by investigating DNA-binding and chromatin processing capabilities of the oncoprotein. To facilitate the generation of mechanism-driven hypotheses regarding DEK activities in underexplored areas, we have developed the first DEK interactome model using tandem-affinity purification and mass spectrometry. With this approach, we identify IMPDH2, DDX21, and RPL7a as novel DEK binding partners, hinting at new roles for the oncogene in de novo nucleotide biosynthesis and ribosome formation. Additionally, a hydroxyurea-specific interaction with replication protein A (RPA) was observed, suggesting that a DEK-RPA complex may form in response to DNA replication fork stalling. Taken together, these findings highlight diverse activities for DEK across cellular pathways and support a model wherein this molecule performs a plethora of functions.
Collapse
Affiliation(s)
- Eric A. Smith
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| | - Eric F. Krumpelbeck
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| | - Anil G. Jegga
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45219, USA
| | - Kenneth D. Greis
- Department of Cancer Biology, University of Cincinnati College of Medicine; Cincinnati, OH 45219, USA
| | - Abdullah M. Ali
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| | - Amom R. Meetei
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| | - Susanne I. Wells
- Department of Oncology; Cincinnati Children’s Hospital Medical Center; Cincinnati, OH, 45219; USA
| |
Collapse
|
18
|
Serrano-Lopez J, Nattamai K, Pease NA, Shephard MS, Wellendorf AM, Sertorio M, Smith EA, Geiger H, Wells SI, Cancelas JA, Privette Vinnedge LM. Loss of DEK induces radioresistance of murine restricted hematopoietic progenitors. Exp Hematol 2017; 59:40-50.e3. [PMID: 29288703 DOI: 10.1016/j.exphem.2017.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/15/2017] [Accepted: 12/19/2017] [Indexed: 11/18/2022]
Abstract
Self-renewing hematopoietic stem cells and multipotent progenitor cells are responsible for maintaining hematopoiesis throughout an individual's lifetime. For overall health and survival, it is critical that the genome stability of these cells is maintained and that the cell population is not exhausted. Previous reports have indicated that the DEK protein, a chromatin structural protein that functions in numerous nuclear processes, is required for DNA damage repair in vitro and long-term engraftment of hematopoietic stem cells in vivo. Therefore, we investigated the role of DEK in normal hematopoiesis and response to DNA damaging agents in vivo. Here, we report that hematopoiesis is largely unperturbed in DEK knockout mice compared with wild-type (WT) controls. However, DEK knockout mice have fewer radioprotective units, but increased capacity to survive repeated sublethal doses of radiation exposure compared with WT mice. Furthermore, this increased survival correlated with a sustained quiescent state in which DEK knockout restricted hematopoietic progenitor cells (HPC-1) were nearly three times more likely to be quiescent following irradiation compared with WT cells and were significantly more radioresistant during the early phases of myeloid reconstitution. Together, our studies indicate that DEK functions in the normal hematopoietic stress response to recurrent radiation exposure.
Collapse
Affiliation(s)
- Juana Serrano-Lopez
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kalpana Nattamai
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Nicholas A Pease
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Miranda S Shephard
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ashley M Wellendorf
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Mathieu Sertorio
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Eric A Smith
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Hartmut Geiger
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Susanne I Wells
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jose A Cancelas
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Lisa M Privette Vinnedge
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
19
|
Neuroanatomical Distribution of DEK Protein in Corticolimbic Circuits Associated with Learning and Memory in Adult Male and Female Mice. Neuroscience 2017; 371:254-267. [PMID: 29175155 DOI: 10.1016/j.neuroscience.2017.11.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 02/08/2023]
Abstract
DEK, a chromatin-remodeling gene expressed in most human tissues, is known for its role in cancer biology and autoimmune diseases. DEK depletion in vitro reduces cellular proliferation, induces DNA damage subsequently leading to apoptosis, and down-regulates canonical Wnt/β-catenin signaling, a molecular pathway essential for learning and memory. Despite a recognized role in cancer (non-neuronal) cells, DEK expression and function is not well characterized in the central nervous system. We conducted a gene ontology analysis (ToppGene), using a cancer database to identify genes associated with DEK deficiency, which pinpointed several genes associated with cognitive-related diseases (i.e., Alzheimer's disease, presenile dementia). Based on this information, we examined DEK expression in corticolimbic structures associated with learning and memory in adult male and female mice using immunohistochemistry. DEK was expressed throughout the brain in both sexes, including the medial prefrontal cortex (prelimbic, infralimbic and dorsal peduncular). DEK was also abundant in all amygdalar subdivisions (basolateral, central and medial) and in the hippocampus including the CA1, CA2, CA3, dentate gyrus (DG), ventral subiculum and entorhinal cortex. Of note, compared to males, females had significantly higher DEK immunoreactivity in the CA1, indicating a sex difference in this region. DEK was co-expressed with neuronal and microglial markers in the CA1 and DG, whereas only a small percentage of DEK cells were in apposition to astrocytes in these areas. Given the reported inverse cellular and molecular profiles (e.g., cell survival, Wnt pathway) between cancer and Alzheimer's disease, these findings suggest a potentially important role of DEK in cognition.
Collapse
|
20
|
Nakashima T, Tomita H, Hirata A, Ishida K, Hisamatsu K, Hatano Y, Kanayama T, Niwa A, Noguchi K, Kato K, Miyazaki T, Tanaka T, Shibata T, Hara A. Promotion of cell proliferation by the proto-oncogene DEK enhances oral squamous cell carcinogenesis through field cancerization. Cancer Med 2017; 6:2424-2439. [PMID: 28834425 PMCID: PMC5633549 DOI: 10.1002/cam4.1157] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 12/11/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) develops through a multistep carcinogenic process involving field cancerization. The DEK gene is a proto-oncogene with functions in genetic and epigenetic modifications, and has oncogenic functions, including cellular proliferation, differentiation, and senescence. DEK overexpression is associated with malignancies; however, the functional roles of DEK overexpression are unclear. We demonstrated that DEK-expressing cells were significantly increased in human dysplasia/carcinoma in situ and OSCC. Furthermore, we generated ubiquitous and squamous cell-specific doxycycline (DOX)-inducible Dek mice (iDek and iDek-e mice respectively). Both DOX+ iDek and iDek-e mice did not show differences in the oral mucosa compared with DOX- mice. In the environment exposed to carcinogen, DOX-treated (DOX+) iDek mice showed field cancerization and OSCC development. Microarray analysis revealed that DEK overexpression was mediated by the upregulation of DNA replication- and cell cycle-related genes, particularly those related to the G1 /S transition. Tongue tumors overexpressing DEK showed increased proliferating cell nuclear antigen and elongator complex protein 3 expression. Our data suggest that DEK overexpression enhanced carcinogenesis, including field cancerization, in OSCC by stimulating the G1 /S phase transition and promoting DNA replication, providing important insights into the potential applications of DEK as a target in the treatment and prevention of OSCC.
Collapse
Affiliation(s)
- Takayuki Nakashima
- Department of Tumor PathologyGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
- Department of Oral Maxillofacial SurgeryGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Hiroyuki Tomita
- Department of Tumor PathologyGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Akihiro Hirata
- Division of Animal ExperimentLife Science Research CenterGifu University1‐1 YanagidoGifu501‐1194Japan
| | - Kazuhisa Ishida
- Department of Tumor PathologyGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
- Department of Oral Maxillofacial SurgeryGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Kenji Hisamatsu
- Department of Tumor PathologyGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Yuichiro Hatano
- Department of Tumor PathologyGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Tomohiro Kanayama
- Department of Tumor PathologyGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Ayumi Niwa
- Department of Tumor PathologyGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Kei Noguchi
- Department of Tumor PathologyGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Keizo Kato
- Department of Oral Maxillofacial SurgeryGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Tatsuhiko Miyazaki
- Division of PathologyGifu University Hospital1‐1 YanagidoGifu501‐1194Japan
| | - Takuji Tanaka
- Department of Diagnostic Pathology (DDP) and Research Center of Diagnostic Pathology (RC‐DiP)Gifu Municipal Hospital7‐1 Kashima‐choGifu500‐8513Japan
| | - Toshiyuki Shibata
- Department of Oral Maxillofacial SurgeryGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| | - Akira Hara
- Department of Tumor PathologyGifu University Graduate School of Medicine1‐1 YanagidoGifu501‐1194Japan
| |
Collapse
|
21
|
Xu X, Zou L, Yao Q, Zhang Y, Gan L, Tang L. Silencing DEK downregulates cervical cancer tumorigenesis and metastasis via the DEK/p-Ser9-GSK-3β/p-Tyr216-GSK-3β/β-catenin axis. Oncol Rep 2017; 38:1035-1042. [PMID: 28627610 DOI: 10.3892/or.2017.5721] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 06/06/2017] [Indexed: 11/05/2022] Open
Abstract
Cervical cancer is the second most common gynecological malignancy. The mechanisms of the genesis and progression of cervical cancer are complicated and not thoroughly understood. DEK is reported as an oncogene in various cancers, such as acute myeloid leukemia, bladder cancer, breast cancer and hepatocellular cancer. However, its role in cervical cancer has not been well studied. In our study, we confirmed the DEK protein as an oncoprotein in cervical cancer tissues which is correlated to cervical cancer FIGO staging and tumor type. Moreover, in vitro loss of DEK inhibited cervical cancer cell proliferation, migration and invasion. We proved that silencing DEK downregulated Wnt/β-catenin and MMP-9, and silencing DEK increased GSK-3β activity via regulating its phosphorylation instead of translation. Silencing DEK reduced p-Ser9-GSK-3β and increased p-Tyr216-GSK-3β, which resulted in β-catenin degradation. Finally, the xenograft model in nude mice proved that silencing DEK impaired cervical cancer cell tumorigenicity. This research unveiled the function of DEK in tumorigenesis and metastasis via the DEK/p-Ser9-GSK-3β/p-Tyr216-GSK-3β/β-catenin axis in cervical cancer and gave insights into DEK-targeting therapy for patients suffering from cervical cancer.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Adenocarcinoma/prevention & control
- Adenocarcinoma/secondary
- Animals
- Apoptosis
- Biomarkers, Tumor
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/prevention & control
- Carcinoma, Squamous Cell/secondary
- Case-Control Studies
- Cell Proliferation
- Cell Transformation, Neoplastic
- Chromosomal Proteins, Non-Histone/antagonists & inhibitors
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- Female
- Follow-Up Studies
- Gene Silencing
- Glycogen Synthase Kinase 3 beta/antagonists & inhibitors
- Glycogen Synthase Kinase 3 beta/genetics
- Glycogen Synthase Kinase 3 beta/metabolism
- Humans
- Lymphatic Metastasis
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Neoplasm Invasiveness
- Oncogene Proteins/antagonists & inhibitors
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Phosphorylation
- Poly-ADP-Ribose Binding Proteins/antagonists & inhibitors
- Poly-ADP-Ribose Binding Proteins/genetics
- Poly-ADP-Ribose Binding Proteins/metabolism
- Prognosis
- Tumor Cells, Cultured
- Uterine Cervical Neoplasms/metabolism
- Uterine Cervical Neoplasms/pathology
- Uterine Cervical Neoplasms/prevention & control
- Xenograft Model Antitumor Assays
- beta Catenin/antagonists & inhibitors
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
Affiliation(s)
- Xiaoyang Xu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lin Zou
- Laboratory of Molecular Diagnosis, The Children's Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Qiuhui Yao
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yanbo Zhang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Li Gan
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Liangdan Tang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
22
|
Matrka MC, Watanabe M, Muraleedharan R, Lambert PF, Lane AN, Romick-Rosendale LE, Wells SI. Overexpression of the human DEK oncogene reprograms cellular metabolism and promotes glycolysis. PLoS One 2017; 12:e0177952. [PMID: 28558019 PMCID: PMC5448751 DOI: 10.1371/journal.pone.0177952] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/05/2017] [Indexed: 12/12/2022] Open
Abstract
The DEK oncogene is overexpressed in many human malignancies including at early tumor stages. Our reported in vitro and in vivo models of squamous cell carcinoma have demonstrated that DEK contributes functionally to cellular and tumor survival and to proliferation. However, the underlying molecular mechanisms remain poorly understood. Based on recent RNA sequencing experiments, DEK expression was necessary for the transcription of several metabolic enzymes involved in anabolic pathways. This identified a possible mechanism whereby DEK may drive cellular metabolism to enable cell proliferation. Functional metabolic Seahorse analysis demonstrated increased baseline and maximum extracellular acidification rates, a readout of glycolysis, in DEK-overexpressing keratinocytes and squamous cell carcinoma cells. DEK overexpression also increased the maximum rate of oxygen consumption and therefore increased the potential for oxidative phosphorylation (OxPhos). To detect small metabolites that participate in glycolysis and the tricarboxylic acid cycle (TCA) that supplies substrate for OxPhos, we carried out NMR-based metabolomics studies. We found that high levels of DEK significantly reprogrammed cellular metabolism and altered the abundances of amino acids, TCA cycle intermediates and the glycolytic end products lactate, alanine and NAD+. Taken together, these data support a scenario whereby overexpression of the human DEK oncogene reprograms keratinocyte metabolism to fulfill energy and macromolecule demands required to enable and sustain cancer cell growth.
Collapse
Affiliation(s)
- Marie C. Matrka
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Miki Watanabe
- NMR-Based Metabolomics Core Facility, Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Ranjithmenon Muraleedharan
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Andrew N. Lane
- Center for Environmental Systems Biochemistry, Dept. Toxicology and Cancer Biology and Markey Cancer Center, Lexington, Kentucky, United States of America
| | - Lindsey E. Romick-Rosendale
- NMR-Based Metabolomics Core Facility, Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Susanne I. Wells
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio, United States of America
| |
Collapse
|
23
|
Smith EA, Kumar B, Komurov K, Smith SM, Brown NV, Zhao S, Kumar P, Teknos TN, Wells SI. DEK associates with tumor stage and outcome in HPV16 positive oropharyngeal squamous cell carcinoma. Oncotarget 2017; 8:23414-23426. [PMID: 28423581 PMCID: PMC5410314 DOI: 10.18632/oncotarget.15582] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 02/12/2017] [Indexed: 01/25/2023] Open
Abstract
Oropharyngeal squamous cell carcinomas (OPSCC) are common, have poor outcomes, and comprise two biologically and clinically distinct diseases. While OPSCC that arise from human papillomavirus infections (HPV+) have better overall survival than their HPV- counterparts, the incidence of HPV+ OPSCC is increasing dramatically, affecting younger individuals which are often left with life-long co-morbidities from aggressive treatment. To identify patients which do poorly versus those who might benefit from milder regimens, risk-stratifying biomarkers are now needed within this population. One potential marker is the DEK oncoprotein, whose transcriptional upregulation in most malignancies is associated with chemotherapy resistance, advanced tumor stage, and worse outcomes. Herein, a retrospective case study was performed on DEK protein expression in therapy-naïve surgical resections from 194 OPSCC patients. We found that DEK was associated with advanced tumor stage, increased hazard of death, and interleukin IL6 expression in HPV16+ disease. Surprisingly, DEK levels in HPV16- OPSCC were not associated with advanced tumor stage or increased hazard of death. Overall, these findings mark HPV16- OPSCC as an exceptional malignancy were DEK expression does not correlate with outcome, and support the potential prognostic utility of DEK to identify aggressive HPV16+ disease.
Collapse
Affiliation(s)
- Eric A. Smith
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Bhavna Kumar
- Department of Otolaryngology–Head and Neck Surgery, The Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Kakajan Komurov
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Stephen M. Smith
- Department of Pathology, The Ohio State University, Columbus, OH, 43210, USA
| | - Nicole V. Brown
- Center for Biostatistics, The Ohio State University, Columbus, OH, 43210, USA
| | - Songzhu Zhao
- Center for Biostatistics, The Ohio State University, Columbus, OH, 43210, USA
| | - Pawan Kumar
- Department of Otolaryngology–Head and Neck Surgery, The Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Theodoros N. Teknos
- Department of Otolaryngology–Head and Neck Surgery, The Ohio State University, Columbus, OH, 43210, USA
- The Ohio State University, Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Susanne I. Wells
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| |
Collapse
|
24
|
Riveiro-Falkenbach E, Ruano Y, García-Martín RM, Lora D, Cifdaloz M, Acquadro F, Ballestín C, Ortiz-Romero PL, Soengas MS, Rodríguez-Peralto JL. DEK oncogene is overexpressed during melanoma progression. Pigment Cell Melanoma Res 2017; 30:194-202. [PMID: 27893188 DOI: 10.1111/pcmr.12563] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/24/2016] [Indexed: 12/26/2022]
Abstract
DEK is an oncoprotein involved in a variety of cellular functions, such as DNA repair, replication, and transcriptional control. DEK is preferentially expressed in actively proliferating and malignant cells, including melanoma cell lines in which DEK was previously demonstrated to play a critical role in proliferation and chemoresistance. Still, the impact of this protein in melanoma progression remains unclear. Thus, we performed a comprehensive analysis of DEK expression in different melanocytic tumors. The immunostaining results of 303 tumors demonstrated negligible DEK expression in benign lesions. Conversely, malignant lesions, particularly in metastatic cases, were largely positive for DEK expression, which was partially associated with genomic amplification. Importantly, DEK overexpression was correlated with histological features of aggressiveness in primary tumors and poor prognosis in melanoma patients. In conclusion, our study provides new insight into the involvement of DEK in melanoma progression, as well as proof of concept for its potential application as a marker and therapeutic target of melanoma.
Collapse
Affiliation(s)
- Erica Riveiro-Falkenbach
- Department of Pathology, Hospital Universitario 12 de Octubre, Instituto i+12, Medical School, Universidad Complutense, Madrid, Spain
| | - Yolanda Ruano
- Department of Pathology, Hospital Universitario 12 de Octubre, Instituto i+12, Medical School, Universidad Complutense, Madrid, Spain
| | - Rosa M García-Martín
- Department of Pathology, Hospital Universitario 12 de Octubre, Instituto i+12, Medical School, Universidad Complutense, Madrid, Spain
| | - David Lora
- Clinical Research Unit (CIBERESP), Hospital Universitario 12 de Octubre, Instituto i+12, Madrid, Spain
| | - Metehan Cifdaloz
- Melanoma Laboratory, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Francesco Acquadro
- Molecular Cytogenetics Group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Claudio Ballestín
- Department of Pathology, Hospital Universitario 12 de Octubre, Instituto i+12, Medical School, Universidad Complutense, Madrid, Spain
| | - Pablo L Ortiz-Romero
- Department of Dermatology, Hospital Universitario 12 de Octubre, Instituto i+12, Medical School, Universidad Complutense, Madrid, Spain
| | - María S Soengas
- Melanoma Laboratory, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - José L Rodríguez-Peralto
- Department of Pathology, Hospital Universitario 12 de Octubre, Instituto i+12, Medical School, Universidad Complutense, Madrid, Spain
| |
Collapse
|
25
|
Sun J, Bi F, Yang Y, Zhang Y, Jin A, Li J, Lin Z. DEK protein overexpression predicts poor prognosis in pancreatic ductal adenocarcinoma. Oncol Rep 2017; 37:857-864. [PMID: 27959420 DOI: 10.3892/or.2016.5302] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/25/2016] [Indexed: 11/06/2022] Open
Abstract
DEK, a transcription factor, is involved in mRNA splicing, transcriptional control, cell division and differentiation. Recent studies suggest that DEK overexpression can promote tumorigenesis in a wide range of cancer cell types. However, little is known concerning the status of DEK in pancreatic ductal adenocarcinoma (PDAC). Based on the microarray data from Gene Expression Omnibus (GEO), the expression levels of DEK mRNA in PDAC tissues were significantly higher than levels in the adjacent non-tumor tissues. To explore the clinical features of DEK overexpression in PDAC, 87 PDAC and 52 normal pancreas tissues were selected for immunoenzyme staining of the DEK protein. Localization of the DEK protein was detected in PANC-1 pancreatic cancer cells using immunofluorescence (IF) staining. The correlations between DEK overexpression and the clinical features of PDAC were evaluated using the Chi-squared (χ2) and Fisher's exact tests. The survival rates were calculated by the Kaplan-Meier method, and the relationship between prognostic factors and patient survival was also analyzed by the Cox proportional hazard models. The expression levels of DEK mRNA in PDAC tissues were significantly higher than that in the adjacent non‑tumor tissues. The DEK protein showed a primarily nuclear staining pattern in PDAC. The positive rate of the DEK protein was 52.9% (46/87) in PDAC, which was significantly higher than that in the adjacent normal pancreatic tissues (7.7%, 4/52). DEK overexpression in PDAC was correlated with tumor size, histological grade, tumor‑node‑metastasis (TNM) stage and overall survival (OS) rates. In addition, multivariate analysis demonstrated that DEK overexpression was an independent prognostic factor along with histological grade and TNM stage in patients with PDAC. In conclusion, DEK overexpression is associated with PDAC progression and may be a potential biomarker for poor prognostic evaluation in PDAC.
Collapse
Affiliation(s)
- Jie Sun
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Fangfang Bi
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Yang Yang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Yuan Zhang
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| | - Aihua Jin
- Department of Internal Medicine, Yanbian University Hospital, Yanji, Jilin 133002, P.R. China
| | - Jinzi Li
- Department of Pediatrics, Yanbian University Hospital, Yanji, Jilin 133002, P.R. China
| | - Zhenhua Lin
- Department of Pathology and Cancer Research Center, Yanbian University Medical College, Yanji, Jilin 133002, P.R. China
| |
Collapse
|
26
|
Yu L, Huang X, Zhang W, Zhao H, Wu G, Lv F, Shi L, Teng Y. Critical role of DEK and its regulation in tumorigenesis and metastasis of hepatocellular carcinoma. Oncotarget 2016; 7:26844-26855. [PMID: 27057626 PMCID: PMC5042019 DOI: 10.18632/oncotarget.8565] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/01/2016] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality globally. Therefore, it is quite essential to identify novel HCC-related molecules for the discovery of new prognostic markers and therapeutic targets. As an oncogene, DEK plays an important role in cell processes and participates in a variety of cellular metabolic functions, and its altered expression is associated with several human malignancies. However, the functional significance of DEK and the involved complex biological events in HCC development and progression are poorly understood. Here, combing the results from clinical specimens and cultured cell lines, we uncover a critical oncogenic role of DEK, which is highly expressed in HCC cells. DEK protein encompasses two isoforms (isoforms 1 and 2) and isoform 1 is the most frequently expressed DEK isoform in HCC cells. DEK depletion by using shRNA inhibited the cell proliferation and migration in vitro and suppressed tumorigenesis and metastasis in mouse models. Consistently, DEK overexpression regardless of which isoform produced the opposite effects. Further studies showed that DEK induced cell proliferation through upregulating cell cycle related CDK signaling, and promoted cell migration and EMT, at least in part, through the repression of β-catenin/E-cadherin axis. Interestingly, isoform 1 induced cell proliferation more efficiently than isoform 2, however, no functional differences existed between these two isoforms in cell migration. Together, our study indicates that DEK expression is required for tumorigenesis and metastasis of HCC, providing molecular insights for DEK-related pathogenesis and a basis for developing new strategies against HCC.
Collapse
Affiliation(s)
- Le Yu
- School of Life Sciences, Chongqing University, Chongqing 400044, PR China
| | - Xiaobin Huang
- School of Life Sciences, Chongqing University, Chongqing 400044, PR China
| | - Wenfa Zhang
- School of Life Sciences, Chongqing University, Chongqing 400044, PR China
| | - Huakan Zhao
- School of Life Sciences, Chongqing University, Chongqing 400044, PR China
| | - Gang Wu
- Third Affiliated Hospital, Third Military Medical University, Chongqing 400044, PR China
| | - Fenglin Lv
- School of Life Sciences, Chongqing University, Chongqing 400044, PR China
| | - Lei Shi
- School of Life Sciences, Chongqing University, Chongqing 400044, PR China
| | - Yong Teng
- School of Life Sciences, Chongqing University, Chongqing 400044, PR China
| |
Collapse
|
27
|
Dissecting the Potential Interplay of DEK Functions in Inflammation and Cancer. JOURNAL OF ONCOLOGY 2015; 2015:106517. [PMID: 26425120 PMCID: PMC4575739 DOI: 10.1155/2015/106517] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/05/2015] [Indexed: 12/12/2022]
Abstract
There is a long-standing correlation between inflammation, inflammatory cell signaling pathways, and tumor formation. Understanding the mechanisms behind inflammation-driven tumorigenesis is of great research and clinical importance. Although not entirely understood, these mechanisms include a complex interaction between the immune system and the damaged epithelium that is mediated by an array of molecular signals of inflammation—including reactive oxygen species (ROS), cytokines, and NFκB signaling—that are also oncogenic. Here, we discuss the association of the unique DEK protein with these processes. Specifically, we address the role of DEK in chronic inflammation via viral infections and autoimmune diseases, the overexpression and oncogenic activity of DEK in cancers, and DEK-mediated regulation of NFκB signaling. Combined, evidence suggests that DEK may play a complex, multidimensional role in chronic inflammation and subsequent tumorigenesis.
Collapse
|
28
|
Deutzmann A, Ganz M, Schönenberger F, Vervoorts J, Kappes F, Ferrando-May E. The human oncoprotein and chromatin architectural factor DEK counteracts DNA replication stress. Oncogene 2015; 34:4270-7. [PMID: 25347734 DOI: 10.1038/onc.2014.346] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 09/03/2014] [Accepted: 09/04/2014] [Indexed: 12/16/2022]
Abstract
DNA replication stress is a major source of DNA strand breaks and genomic instability, and a hallmark of precancerous lesions. In these hyperproliferative tissues, activation of the DNA damage response results in apoptosis or senescence preventing or delaying their development to full malignancy. In cells, in which this antitumor barrier is disabled by mutations (for example, in p53), viability and further uncontrolled proliferation depend on factors that help to cope with replication-associated DNA damage. Replication problems preferentially arise in chromatin regions harboring complex DNA structures. DEK is a unique chromatin architectural factor which binds to non-B-form DNA structures, such as cruciform DNA or four-way junctions. It regulates DNA topology and chromatin organization, and is essential for the maintenance of heterochromatin integrity. Since its isolation as part of an oncogenic fusion in a subtype of AML, DEK has been consistently associated with tumor progression and chemoresistance. How DEK promotes cancer, however, is poorly understood. Here we show that DEK facilitates cellular proliferation under conditions of DNA replication stress by promoting replication fork progression. DEK also protects from the transmission of DNA damage to the daughter cell generation. We propose that DEK counteracts replication stress and ensures proliferative advantage by resolving problematic DNA and/or chromatin structures at the replication fork.
Collapse
Affiliation(s)
- A Deutzmann
- Bioimaging Center, Department of Biology, University of Konstanz, Konstanz, Germany
| | - M Ganz
- Bioimaging Center, Department of Biology, University of Konstanz, Konstanz, Germany
| | - F Schönenberger
- Bioimaging Center, Department of Biology, University of Konstanz, Konstanz, Germany
| | - J Vervoorts
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - F Kappes
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany
| | - E Ferrando-May
- Bioimaging Center, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
29
|
Sandén C, Gullberg U. The DEK oncoprotein and its emerging roles in gene regulation. Leukemia 2015; 29:1632-6. [PMID: 25765544 DOI: 10.1038/leu.2015.72] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/08/2015] [Accepted: 03/03/2015] [Indexed: 02/06/2023]
Abstract
The DEK oncogene is highly expressed in cells from most human tissues and overexpressed in a large and growing number of cancers. It also fuses with the NUP214 gene to form the DEK-NUP214 fusion gene in a subset of acute myeloid leukemia. Originally characterized as a member of this translocation, DEK has since been implicated in epigenetic and transcriptional regulation, but its role in these processes is still elusive and intriguingly complex. Similarly multifaceted is its contribution to cellular transformation, affecting multiple cellular processes such as self-renewal, proliferation, differentiation, senescence and apoptosis. Recently, the roles of the DEK and DEK-NUP214 proteins have been elucidated by global analysis of DNA binding and gene expression, as well as multiple functional studies. This review outlines recent advances in the understanding of the basic functions of the DEK protein and its role in leukemogenesis.
Collapse
Affiliation(s)
- C Sandén
- Department of Hematology, Lund University, Lund, Sweden
| | - U Gullberg
- Department of Hematology, Lund University, Lund, Sweden
| |
Collapse
|
30
|
Matrka MC, Hennigan RF, Kappes F, DeLay ML, Lambert PF, Aronow BJ, Wells SI. DEK over-expression promotes mitotic defects and micronucleus formation. Cell Cycle 2015; 14:3939-53. [PMID: 25945971 PMCID: PMC4825741 DOI: 10.1080/15384101.2015.1044177] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/18/2015] [Indexed: 10/23/2022] Open
Abstract
The DEK gene encodes a nuclear protein that binds chromatin and is involved in various fundamental nuclear processes including transcription, RNA splicing, DNA replication and DNA repair. Several cancer types characteristically over-express DEK at the earliest stages of transformation. In order to explore relevant mechanisms whereby DEK supports oncogenicity, we utilized cancer databases to identify gene transcripts whose expression patterns are tightly correlated with that of DEK. We identified an enrichment of genes involved in mitosis and thus investigated the regulation and possible function of DEK in cell division. Immunofluorescence analyses revealed that DEK dissociates from DNA in early prophase and re-associates with DNA during telophase in human keratinocytes. Mitotic cell populations displayed a sharp reduction in DEK protein levels compared to the corresponding interphase population, suggesting DEK may be degraded or otherwise removed from the cell prior to mitosis. Interestingly, DEK overexpression stimulated its own aberrant association with chromatin throughout mitosis. Furthermore, DEK co-localized with anaphase bridges, chromosome fragments, and micronuclei, suggesting a specific association with mitotically defective chromosomes. We found that DEK over-expression in both non-transformed and transformed cells is sufficient to stimulate micronucleus formation. These data support a model wherein normal chromosomal clearance of DEK is required for maintenance of high fidelity cell division and chromosomal integrity. Therefore, the overexpression of DEK and its incomplete removal from mitotic chromosomes promotes genomic instability through the generation of genetically abnormal daughter cells. Consequently, DEK over-expression may be involved in the initial steps of developing oncogenic mutations in cells leading to cancer initiation.
Collapse
Affiliation(s)
- Marie C Matrka
- Cancer and Blood Diseases Institute; Cincinnati Children's Hospital Medical Center and University of Cincinnati; Cincinnati, OH USA
| | - Robert F Hennigan
- Cancer and Blood Diseases Institute; Cincinnati Children's Hospital Medical Center and University of Cincinnati; Cincinnati, OH USA
| | - Ferdinand Kappes
- Department of Biological Sciences; Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu Province, China
- Institute of Biochemistry and Molecular Biology; Medical School; RWTH Aachen University; Aachen, Germany
| | - Monica L DeLay
- Division of Rheumatology; Cincinnati Children's Hospital Medical Center; Cincinnati, OH USA
| | - Paul F Lambert
- McArdle Laboratory for Cancer Research; University of Wisconsin-Madison School of Medicine and Public Health; Madison, WI USA
| | - Bruce J Aronow
- Biomedical Informatics; Cincinnati Children's Hospital Medical Center; Cincinnati, OH USA
| | - Susanne I Wells
- Cancer and Blood Diseases Institute; Cincinnati Children's Hospital Medical Center and University of Cincinnati; Cincinnati, OH USA
| |
Collapse
|
31
|
Privette Vinnedge LM, Benight NM, Wagh PK, Pease NA, Nashu MA, Serrano-Lopez J, Adams AK, Cancelas JA, Waltz SE, Wells SI. The DEK oncogene promotes cellular proliferation through paracrine Wnt signaling in Ron receptor-positive breast cancers. Oncogene 2015; 34:2325-36. [PMID: 24954505 PMCID: PMC4275425 DOI: 10.1038/onc.2014.173] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 04/18/2014] [Accepted: 05/09/2014] [Indexed: 12/12/2022]
Abstract
Disease progression and recurrence are major barriers to survival for breast cancer patients. Understanding the etiology of recurrent or metastatic breast cancer and underlying mechanisms is critical for the development of new treatments and improved survival. Here, we report that two commonly overexpressed breast cancer oncogenes, Ron (Recepteur d'Origine Nantaise) and DEK, cooperate to promote advanced disease through multipronged effects on β-catenin signaling. The Ron receptor is commonly activated in breast cancers, and Ron overexpression in human disease stimulates β-catenin nuclear translocation and is an independent predictor of metastatic dissemination. Dek is a chromatin-associated oncogene whose expression has been linked to cancer through multiple mechanisms, including β-catenin activity. We demonstrate here that Dek is a downstream target of Ron receptor activation in murine and human models. The absence of Dek in the MMTV-Ron mouse model led to a significant delay in tumor development, characterized by decreased cell proliferation, diminished metastasis and fewer cells expressing mammary cancer stem cell markers. Dek complementation of cell lines established from this model was sufficient to promote cellular growth and invasion. Mechanistically, Dek expression stimulated the production and secretion of Wnt ligands to sustain an autocrine/paracrine canonical β-catenin signaling loop. Finally, we show that Dek overexpression promotes tumorigenic phenotypes in immortalized human mammary epithelial MCF10A cells and, in the context of Ron receptor activation, correlates with disease recurrence and metastasis in patients. Overall, our studies demonstrate that DEK overexpression, due in part to Ron receptor activation, drives breast cancer progression through the induction of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
| | - Nancy M. Benight
- Department of Cancer Biology, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH
| | - Purnima K. Wagh
- Department of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Nicholas A. Pease
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Madison A. Nashu
- Department of Cancer Biology, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH
| | - Juana Serrano-Lopez
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- IMIBIC/UCO/University Hospital Reina Sofia, Cordoba, Spain
| | - Allie K. Adams
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Susan E. Waltz
- Department of Cancer Biology, University of Cincinnati College of Medicine Cincinnati, Cincinnati, OH
- Department of Research, Cincinnati Veterans Affairs Medical Center, Cincinnati, OH 45220
| | - Susanne I. Wells
- Division of Oncology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
32
|
Adams AK, Hallenbeck GE, Casper KA, Patil YJ, Wilson KM, Kimple RJ, Lambert PF, Witte DP, Xiao W, Gillison ML, Wikenheiser-Brokamp KA, Wise-Draper TM, Wells SI. DEK promotes HPV-positive and -negative head and neck cancer cell proliferation. Oncogene 2015; 34:868-77. [PMID: 24608431 PMCID: PMC4160430 DOI: 10.1038/onc.2014.15] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/21/2014] [Accepted: 02/03/2014] [Indexed: 02/07/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common malignancy worldwide, and patient outcomes using current treatments remain poor. Tumor development is etiologically associated with tobacco or alcohol use and/or human papillomavirus (HPV) infection. HPV-positive HNSCCs, which frequently harbor wild-type p53, carry a more favorable prognosis and are a biologically distinct subgroup when compared with their HPV-negative counterparts. HPV E7 induces expression of the human DEK gene, both in vitro and in vivo. In keratinocytes, DEK overexpression is sufficient for causing oncogenic phenotypes in the absence of E7. Conversely, DEK loss results in cell death in HPV-positive cervical cancer cells at least in part through p53 activation, and Dek knockout mice are relatively resistant to the development of chemically induced skin papillomas. Despite the established oncogenic role of DEK in HPV-associated cervical cancer cell lines and keratinocytes, a functional role of DEK has not yet been explored in HNSCC. Using an established transgenic mouse model of HPV16 E7-induced HNSCC, we demonstrate that Dek is required for optimal proliferation of E7-transgenic epidermal cells and for the growth of HNSCC tumors. Importantly, these studies also demonstrate that DEK protein is universally upregulated in both HPV-positive and -negative human HNSCC tumors relative to adjacent normal tissue. Furthermore, DEK knockdown inhibited the proliferation of HPV-positive and -negative HNSCC cells, establishing a functional role for DEK in human disease. Mechanistic studies reveal that attenuated HNSCC cell growth in response to DEK loss was associated with reduced expression of the oncogenic p53 family member, ΔNp63. Exogenous ΔNp63 expression rescued the proliferative defect in the absence of DEK, thereby establishing a functional DEK-ΔNp63 oncogenic pathway that promotes HNSCC. Taken together, our data demonstrate that DEK stimulates HNSCC cellular growth and identify ΔNp63 as a novel DEK effector.
Collapse
Affiliation(s)
- Allie K. Adams
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Grace E. Hallenbeck
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Keith A. Casper
- Department of Otolaryngology, Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Yash J. Patil
- Department of Otolaryngology, Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Keith M. Wilson
- Department of Otolaryngology, Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Randall J. Kimple
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - David P. Witte
- Division of Pathology & Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Weihong Xiao
- Viral Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Maura L. Gillison
- Viral Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Kathryn A. Wikenheiser-Brokamp
- Pathology & Laboratory Medicine and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center/University of Cincinnati, Cincinnati, OH, USA
| | - Trisha M. Wise-Draper
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Division of Hematology/Oncology, University Hospital, University of Cincinnati, Cincinnati, OH, USA
| | - Susanne I. Wells
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
33
|
Yi HC, Liu YL, You P, Pan JS, Zhou JY, Liu ZJ, Zhang ZY. Overexpression of DEK gene is correlated with poor prognosis in hepatocellular carcinoma. Mol Med Rep 2015; 11:1318-1323. [PMID: 25351213 DOI: 10.3892/mmr.2014.2781] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 09/09/2014] [Indexed: 11/06/2022] Open
Abstract
The oncogene DEK was originally identified as one of the parts of the DEK‑CAN fusion gene, arising from the translocation (6;9) in a subtype of acute myeloid leukemia. Since then, DEK has been shown to promote tumorigenesis in a variety of cancer cell types through its roles in inhibiting cell differentiation, senescence and apoptosis. Certain studies have established that DEK is dysregulated in several types of cancer, including hepatocellular carcinoma (HCC). However, its clinical significance in human HCC remains unknown. In this study, the expression of DEK mRNA and protein was examined in 55 surgical HCC specimens and matched non‑tumorous tissues. In addition, the correlation between DEK expression and clinicopathological characteristics and prognosis was analyzed. mRNA and protein levels of DEK were found to be significantly overexpressed in the majority of HCC tumors when compared with matched normal hepatic tissues (P<0.05). In addition, the expression pattern of DEK was closely correlated with differentiation status, portal venous invasion and tumor size (P<0.05). Kaplan‑Meier curves demonstrated that patients with higher DEK expression levels had significantly poorer survival than those with lower DEK expression levels (P=0.003). In addition, Cox regression analysis demonstrated that the level of DEK expression may be a valuable prognostic factor (P<0.05). These results suggested that DEK may play a significant role in hepatocyte differentiation and may serve as a useful prognostic marker and biomarker for the staging of HCC.
Collapse
Affiliation(s)
- Huo-Chun Yi
- Center of Clinical Laboratory, Zhongshan Hospital, Medical College of Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Ya-Li Liu
- Department of Thoracic Surgery, Zhongshan Hospital, Medical College of Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Pan You
- Center of Clinical Laboratory, Zhongshan Hospital, Medical College of Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Jin-Shui Pan
- Department of Gastroenterology, Zhongshan Hospital, Medical College of Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Jian-Yan Zhou
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Medical College of Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Zhen-Jin Liu
- Department of Pathology, Zhongshan Hospital, Medical College of Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Zhong-Ying Zhang
- Center of Clinical Laboratory, Zhongshan Hospital, Medical College of Xiamen University, Xiamen, Fujian 361004, P.R. China
| |
Collapse
|
34
|
Lin L, Piao J, Ma Y, Jin T, Quan C, Kong J, Li Y, Lin Z. Mechanisms underlying cancer growth and apoptosis by DEK overexpression in colorectal cancer. PLoS One 2014; 9:e111260. [PMID: 25340858 PMCID: PMC4207817 DOI: 10.1371/journal.pone.0111260] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 09/24/2014] [Indexed: 11/18/2022] Open
Abstract
Our previous study indicated that DEK protein was overexpressed in colorectal carcinoma (CRC) compared with the normal colorectal mucosa. DEK was also significantly correlated with the prognostic characteristics of patients with CRC, demonstrating that DEK played an important role in CRC progression. In this work, we evaluate the effects of DEK on biological behaviors in CRC and explore the related molecular mechanisms. The results showed that DEK was overexpressed in human CRC tissues, and was correlated with the Ki-67 index and the apoptotic index. DEK depletion by RNAi in SW-620 and HCT116 cells significantly decreased cell proliferation, but increased cell apoptosis. Upregulation of DEK was involved in the p53/MDM, Bcl-2 family, and caspase pathways. Our study demonstrates that DEK promotes the growth of CRC, and could be a therapeutic target in CRC.
Collapse
Affiliation(s)
- Lijuan Lin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
- Department of Medical Imaging, College of Medicine, Eastern Liaoning University, Dandong, China
| | - Junjie Piao
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Yibing Ma
- Department of Pathology, Dandong Centre Hospital, Dandong, China
| | - Tiefeng Jin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, Bethune Medical College, Jilin University, Changchun, China
| | - Jienan Kong
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Bethune Medical College, Jilin University, Changchun, China
| | - Zhenhua Lin
- Department of Pathology & Cancer Research Center, Yanbian University Medical College, Yanji, China
| |
Collapse
|
35
|
Sandén C, Järvstråt L, Lennartsson A, Brattås PL, Nilsson B, Gullberg U. The DEK oncoprotein binds to highly and ubiquitously expressed genes with a dual role in their transcriptional regulation. Mol Cancer 2014; 13:215. [PMID: 25216995 PMCID: PMC4175287 DOI: 10.1186/1476-4598-13-215] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The DEK gene is highly expressed in a wide range of cancer cells, and a recurrent translocation partner in acute myeloid leukemia. While DEK has been identified as one of the most abundant proteins in human chromatin, its function and binding properties are not fully understood. METHODS We performed ChIP-seq analysis in the myeloid cell line U937 and coupled it with epigenetic and gene expression analysis to explore the genome-wide binding pattern of DEK and its role in gene regulation. RESULTS We show that DEK preferentially binds to open chromatin, with a low degree of DNA methylation and scarce in the heterochromatin marker H3K9me(3) but rich in the euchromatin marks H3K4me(2/3), H3K27ac and H3K9ac. More specifically, DEK binding is predominantly located at the transcription start sites of highly transcribed genes and a comparative analysis with previously established transcription factor binding patterns shows a similarity with that of RNA polymerase II. Further bioinformatic analysis demonstrates that DEK mainly binds to genes that are ubiquitously expressed across tissues. The functional significance of DEK binding was demonstrated by knockdown of DEK by shRNA, resulting in both significant upregulation and downregulation of DEK-bound genes. CONCLUSIONS We find that DEK binds to transcription start sites with a dual role in activation and repression of highly and ubiquitously expressed genes.
Collapse
Affiliation(s)
- Carl Sandén
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| | - Linnea Järvstråt
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| | - Andreas Lennartsson
- />Center for Biosciences, Department of Biosciences and Nutrition, Karolinska Institute, Novum, 141 83 Huddinge, Sweden
| | - Per Ludvik Brattås
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| | - Björn Nilsson
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| | - Urban Gullberg
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| |
Collapse
|
36
|
Piao J, Shang Y, Liu S, Piao Y, Cui X, Li Y, Lin Z. High expression of DEK predicts poor prognosis of gastric adenocarcinoma. Diagn Pathol 2014; 9:67. [PMID: 24650035 PMCID: PMC3994479 DOI: 10.1186/1746-1596-9-67] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/03/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND DEK, as an oncoprotein, plays an important role in cancer development and progression. This study aimed to investigate the clinicopathological significance of DEK overexpression in patients with gastric cancer. MATERIALS AND METHODS The expression of DEK protein was evaluated by immunohistochemical (IHC) staining of 172 gastric cancer samples with complete clinicopathological features, and the correlation between DEK expression and clinicopathological features was examined. Survival rates were also calculated using the Kaplan-Meier method in gastric cancer patients with complete survival data. RESULTS DEK protein showed a strictly nuclear staining pattern in gastric cancers with IHC and immunofluorescence. The strongly positive rate of DEK protein was 60.5% (104/172) in gastric cancers, which was significantly higher than that in either gastric dysplasia (19.4%, 7/36) or adjacent normal mucosa (0%, 0/27). DEK expression in gastric cancer correlated to tumor size, differentiation, clinical stage, disease-free survival, and overall survival rates. Further analysis showed that patients with early-stage gastric cancer and high DEK expression had shorter disease-free survival and overall survival duration than those with low DEK expression. CONCLUSION High level of DEK protein expression predicts the poor prognosis of patients with gastric cancer. DEK expression might be potentially used as an independent effective biomarker for prognostic evaluation of gastric cancers. VIRTUAL SLIDES The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/5050145571193097.
Collapse
Affiliation(s)
- Junjie Piao
- Department of Pathology, Yanbian University Medical College, No. 977, Gongyuan-Rd, Yanji 133002, China
- Cancer Research Center, Yanbian University, Yanji 133002, China
| | - Yongjun Shang
- Department of Orthopedics, Affiliated Hospital of Chifeng University, Chifeng 024000, China
| | - Shuangping Liu
- Department of Pathology, Yanbian University Medical College, No. 977, Gongyuan-Rd, Yanji 133002, China
- Cancer Research Center, Yanbian University, Yanji 133002, China
| | - Yingshi Piao
- Cancer Research Center, Yanbian University, Yanji 133002, China
| | - Xuelian Cui
- Department of Pathology, Yanbian University Medical College, No. 977, Gongyuan-Rd, Yanji 133002, China
| | - Yuzi Li
- Department of Internal Medicine, Yanbian University Hospital, No. 1327, Juzi-St, Yanji 133000, China
| | - Zhenhua Lin
- Department of Pathology, Yanbian University Medical College, No. 977, Gongyuan-Rd, Yanji 133002, China
- Cancer Research Center, Yanbian University, Yanji 133002, China
| |
Collapse
|
37
|
Zhang Z, Xiao C, Gibson AM, Bass SA, Khurana Hershey GK. EGFR signaling blunts allergen-induced IL-6 production and Th17 responses in the skin and attenuates development and relapse of atopic dermatitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:859-66. [PMID: 24337738 PMCID: PMC3946981 DOI: 10.4049/jimmunol.1301062] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite the important role for epidermal growth factor (EGF) in epithelial homeostasis and wound healing, it has not been investigated in atopic dermatitis (AD). We used AD animal models to explore the role of EGF in AD. In an acute AD model, skin transepidermal water loss was significantly attenuated in EGF-treated mice. Blockade of EGFR signaling genetically or pharmacologically confirms a protective role for EGFR signaling in AD. In a chronic/relapsing AD model, EGF treatment of mice with established AD resulted in an attenuation of AD exacerbation (skin epithelial thickness, cutaneous inflammation, and total and allergen specific IgE) following cutaneous allergen rechallenge. EGF treatment did not alter expression of skin barrier junction proteins or antimicrobial peptides in the AD model. However, EGF treatment attenuated allergen-induced expression of IL-17A, CXCL1, and CXCL2 and neutrophil accumulation in AD skin following cutaneous allergen exposure. IL-17A production was decreased in the in vitro restimulated skin-draining lymph node cells from the EGF-treated mice. Similarly, IL-17A was increased in waved-2 mice skin following allergen exposure. Whereas IL-6 and IL-1β expression was attenuated in the skin of EGF-treated mice, EGF treatment also suppressed allergen-induced IL-6 production by keratinocytes. Given the central role of IL-6 in priming Th17 differentiation in the skin, this effect of EGF on keratinocytes may contribute to the protective roles for EGFR in AD pathogenesis. In conclusion, our study provides evidence for a previously unrecognized protective role for EGF in AD and a new role for EGF in modulating IL-17 responses in the skin.
Collapse
Affiliation(s)
- Zhonghua Zhang
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Chang Xiao
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Aaron M. Gibson
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Stacey A. Bass
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Gurjit K. Khurana Hershey
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| |
Collapse
|
38
|
Reddy D, Bhattacharya S, Gupta S. Histone Chaperones: Functions beyond Nucleosome Deposition. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/abb.2014.56064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
39
|
Broxmeyer HE, Mor-Vaknin N, Kappes F, Legendre M, Saha AK, Ou X, O'Leary H, Capitano M, Cooper S, Markovitz DM. Concise review: role of DEK in stem/progenitor cell biology. Stem Cells 2013; 31:1447-53. [PMID: 23733396 PMCID: PMC3814160 DOI: 10.1002/stem.1443] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 12/19/2022]
Abstract
Understanding the factors that regulate hematopoiesis opens up the possibility of modifying these factors and their actions for clinical benefit. DEK, a non-histone nuclear phosphoprotein initially identified as a putative proto-oncogene, has recently been linked to regulate hematopoiesis. DEK has myelosuppressive activity in vitro on proliferation of human and mouse hematopoietic progenitor cells and enhancing activity on engraftment of long-term marrow repopulating mouse stem cells, has been linked in coordinate regulation with the transcription factor C/EBPα, for differentiation of myeloid cells, and apparently targets a long-term repopulating hematopoietic stem cell for leukemic transformation. This review covers the uniqueness of DEK, what is known about how it now functions as a nuclear protein and also as a secreted molecule that can act in paracrine fashion, and how it may be regulated in part by dipeptidylpeptidase 4, an enzyme known to truncate and modify a number of proteins involved in activities on hematopoietic cells. Examples are provided of possible future areas of investigation needed to better understand how DEK may be regulated and function as a regulator of hematopoiesis, information possibly translatable to other normal and diseased immature cell systems.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Privette Vinnedge LM, Kappes F, Nassar N, Wells SI. Stacking the DEK: from chromatin topology to cancer stem cells. Cell Cycle 2013; 12:51-66. [PMID: 23255114 PMCID: PMC3570517 DOI: 10.4161/cc.23121] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stem cells are essential for development and tissue maintenance and display molecular markers and functions distinct from those of differentiated cell types in a given tissue. Malignant cells that exhibit stem cell-like activities have been detected in many types of cancers and have been implicated in cancer recurrence and drug resistance. Normal stem cells and cancer stem cells have striking commonalities, including shared cell surface markers and signal transduction pathways responsible for regulating quiescence vs. proliferation, self-renewal, pluripotency and differentiation. As the search continues for markers that distinguish between stem cells, progenitor cells and cancer stem cells, growing evidence suggests that a unique chromatin-associated protein called DEK may confer stem cell-like qualities. Here, we briefly describe current knowledge regarding stem and progenitor cells. We then focus on new findings that implicate DEK as a regulator of stem and progenitor cell qualities, potentially through its unusual functions in the regulation of local or global chromatin organization.
Collapse
Affiliation(s)
- Lisa M Privette Vinnedge
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | | | | | | |
Collapse
|
41
|
Wang DM, Liu L, Fan L, Zou ZJ, Zhang LN, Yang S, Li JY, Xu W. Expression level of DEK in chronic lymphocytic leukemia is regulated by fludarabine and Nutlin-3 depending on p53 status. Cancer Biol Ther 2012; 13:1522-8. [PMID: 23052131 PMCID: PMC3542244 DOI: 10.4161/cbt.22252] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human oncogene DEK has been shown to be upregulated in a number of neoplasms. The purpose of this study was to investigate DEK expression level in chronic lymphocytic leukemia (CLL), analyze the correlation between DEK expression and CLL prognostic markers, and characterize the role of DEK in the response to either chemotherapeutic drugs or nongenotoxic activators of the p53 pathway. DEK mRNA was evaluated by real-time quantitative reverse transcriptase-polymerase chain reaction (qPCR), and primary CLL samples were treated in vitro with either fludarabine or Nutlin-3 to explore the interaction of p53 status and DEK mRNA expression. The median expression levels of DEK mRNA were 6.792 × 10 (-2) (1.438 × 10 (-2) -3.201 × 10 (-1) ) in 65 patients with CLL. A marked increase of DEK mRNA expression was observed in the CLL patients with unmutated immunoglobulin heavy chain variable (IGHV) gene (p = 0.025), CD38-positive (p = 0.047), del(17p13) (p = 0.006). Both fludarabine and Nutlin-3 significantly downregulated DEK in the primary CLL cells which were with normal function of p53, or without deletion or mutation of p53 (p = 0.042, p = 0.038; p = 0.021, p = 0.017; p = 0.037, p = 0.017). However, the downregulation of DEK was not observed in the primary CLL cells which were with dysfunction of p53, or with deletion or mutation of p53 (p = 0.834, p = 0.477; p = 0.111, p = 0.378; p = 0.263, p = 0.378). These data show that DEK might be applied for the assessment of prognosis in patients with CLL, and fludarabine and Nutlin-3 regulate DEK expression depended on p53 status.
Collapse
Affiliation(s)
| | | | - Lei Fan
- Department of Hematology; The First Affiliated Hospital of Nanjing Medical University; Jiangsu Province Hospital; Nanjing, PR China
| | - Zhi-Jian Zou
- Department of Hematology; The First Affiliated Hospital of Nanjing Medical University; Jiangsu Province Hospital; Nanjing, PR China
| | - Li-Na Zhang
- Department of Hematology; The First Affiliated Hospital of Nanjing Medical University; Jiangsu Province Hospital; Nanjing, PR China
| | - Shu Yang
- Department of Hematology; The First Affiliated Hospital of Nanjing Medical University; Jiangsu Province Hospital; Nanjing, PR China
| | - Jian-Yong Li
- Department of Hematology; The First Affiliated Hospital of Nanjing Medical University; Jiangsu Province Hospital; Nanjing, PR China
| | - Wei Xu
- Department of Hematology; The First Affiliated Hospital of Nanjing Medical University; Jiangsu Province Hospital; Nanjing, PR China
| |
Collapse
|
42
|
Future directions and treatment strategies for head and neck squamous cell carcinomas. Transl Res 2012; 160:167-77. [PMID: 22683420 PMCID: PMC3423575 DOI: 10.1016/j.trsl.2012.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 01/16/2012] [Accepted: 02/04/2012] [Indexed: 11/23/2022]
Abstract
Head and neck cancer is a devastating disease that afflicts many individuals worldwide. Conventional therapies are successful in only a limited subgroup and often leave the patient with disfigurement and long lasting adverse effects on normal physiologic functions. The field is in dire need of new therapies. Oncolytic viral as well as targeted therapies have shown some success in other malignancies and are attractive for the treatment of head and neck cancer. Recently, it has been shown that a subset of head and neck cancers is human papillomavirus (HPV) positive and that this subset of cancers is biologically distinct and more sensitive to chemoradiation therapies although the underlying mechanism is unclear. However, chemoresistance remains a general problem. One candidate mediator of therapeutic response, which is of interest for the targeting of both HPV-positive and -negative tumors is the human DEK proto-oncogene. DEK is upregulated in numerous tumors including head and neck cancers regardless of their HPV status. Depletion of DEK in tumor cells in culture results in sensitivity to genotoxic agents, particularly in rapidly proliferating cells. This suggests that tumors with high DEK protein expression may be correlated with poor clinical response to clastogenic therapies. Targeting molecules such as DEK in combination with new and/or conventional therapies, holds promise for novel future therapeutics for head and neck cancer.
Collapse
|
43
|
Patel RM, Walters LL, Kappes F, Mehra R, Fullen DR, Markovitz DM, Ma L. DEK expression in Merkel cell carcinoma and small cell carcinoma. J Cutan Pathol 2012; 39:753-7. [PMID: 22765016 DOI: 10.1111/j.1600-0560.2012.01941.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2012] [Indexed: 12/29/2022]
Abstract
BACKGROUND The chromatin architectural factor DEK maps to chromosome 6p and is frequently overexpressed in several neoplasms, including small cell lung carcinoma, where it is associated with poor prognosis, tumor initiation activity and chemoresistance. DEK expression has not been studied in cutaneous Merkel cell carcinoma. METHODS We applied a DEK monoclonal antibody to 15 cases of Merkel cell carcinoma and 12 cases of small cell carcinoma. DEK nuclear immunoreactivity was scored based on percentage (0, negative; 1+, <25%; 2+, 25-50%; 3+, >50%) and intensity (weak, moderate or strong). RESULTS All 15 Merkel cell carcinoma cases (100%) showed diffuse (3+) nuclear positivity (14 strong, 1 weak). Six of 12 small cell carcinoma cases (50%) showed diffuse (3+) and strong nuclear positivity, while one case exhibited focal (1+) weak nuclear positivity. The remaining five cases were negative for DEK expression. CONCLUSIONS Our results suggest that DEK may be involved in the pathogenesis of Merkel cell carcinoma and therefore may provide therapeutic implications for Merkel cell carcinomas. In addition, the difference in DEK expression between Merkel cell carcinoma and small cell carcinoma suggests possible separate tumorigenesis pathways for the two tumors.
Collapse
Affiliation(s)
- Rajiv M Patel
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, MI, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
High-risk human papillomaviruses (HPVs) deregulate epidermal differentiation and cause anogenital and head and neck squamous cell carcinomas (SCCs). The E7 gene is considered the predominant viral oncogene and drives proliferation and genome instability. While the implementation of routine screens has greatly reduced the incidence of cervical cancers which are almost exclusively HPV positive, the proportion of HPV-positive head and neck SCCs is on the rise. High levels of HPV oncogene expression and genome load are linked to disease progression, but genetic risk factors that regulate oncogene abundance and/or genome amplification remain poorly understood. Fanconi anemia (FA) is a genome instability syndrome characterized at least in part by extreme susceptibility to SCCs. FA results from mutations in one of 15 genes in the FA pathway, whose protein products assemble in the nucleus and play important roles in DNA damage repair. We report here that loss of FA pathway components FANCA and FANCD2 stimulates E7 protein accumulation in human keratinocytes and causes increased epithelial proliferation and basal cell layer expansion in the HPV-positive epidermis. Additionally, FANCD2 loss stimulates HPV genome amplification in differentiating cells, demonstrating that the intact FA pathway functions to restrict the HPV life cycle. These findings raise the possibility that FA genes suppress HPV infection and disease and suggest possible mechanism(s) for reported associations of HPV with an FA cohort in Brazil and for allelic variation of FA genes with HPV persistence in the general population.
Collapse
|
45
|
Su PH, Lee IC, Yang SF, Ng YY, Liu CS, Chen JY. Nine genes that may contribute to partial trisomy (6)(p22→pter) and unique presentation of persistent hyperplastic primary vitreous with retinal detachment. Am J Med Genet A 2012; 158A:707-12. [DOI: 10.1002/ajmg.a.33943] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Accepted: 01/22/2011] [Indexed: 11/12/2022]
|
46
|
Liu S, Wang X, Sun F, Kong J, Li Z, Lin Z. DEK overexpression is correlated with the clinical features of breast cancer. Pathol Int 2012; 62:176-81. [PMID: 22360505 DOI: 10.1111/j.1440-1827.2011.02775.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To investigate the clinicopathological significance of DEK overexpression in breast cancers, a total of 196 cases, including 20 of normal tissues, 12 of intraductal hyperplasia, 31 of ductal carcinoma in situ (DCIS) and 133 of invasive ductal carcinoma of the breast, were selected from the Department of Pathology, Yanbian Tumor Hospital for immunohistochemical staining of DEK, estrogen (ER), progesterone (PR) and Ki-67 proteins. In results, DEK protein had higher positivity in DCIS, compared with the adjacent normal breast tissues. Also, DEK protein was strongly positive in invasive ductal carcinoma of the breast on immunohistochemistry, which was significantly higher than normal breast tissues. However, only two (2/12) cases of intraductal hyperplasia of the breast showed positive staining for DEK protein. Additionally, DEK overexpression was significantly correlated with the increased proliferating index of Ki-67. For the histological grade, DEK positive rate was only 39.6% in G1 breast cancers, but significantly higher in G2 (92.3%) and G3 (97.0%) cases (P<0.05). Also, a strongly positive rate of DEK was lower in Stage-0 (21.4%) and Stage-I (40.9%) compared with Stage-IIa (87.5%), Stage-IIb (89.7%) and Stage-IIIa (92.3%) (P<0.05). And DEK protein showed higher expression level in < 3 years disease free survival breast cancers than it did in ≥ 3 years disease free survival cases (P<0.05). However, no statistically difference was found among DEK expression, lymph node metastasis, and ER and PR expressions. In conclusion, DEK overexpression appears to be associated with breast cancer progression and DEK may potentially be used as a breast cancer biomarker for the early diagnosis, prognostic evaluation and therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Shuangping Liu
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | | | | | | | | | | |
Collapse
|
47
|
Sachdeva UM, O'Brien JM. Understanding pRb: toward the necessary development of targeted treatments for retinoblastoma. J Clin Invest 2012; 122:425-34. [PMID: 22293180 DOI: 10.1172/jci57114] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Retinoblastoma is a pediatric retinal tumor initiated by biallelic inactivation of the retinoblastoma gene (RB1). RB1 was the first identified tumor suppressor gene and has defined roles in the regulation of cell cycle progression, DNA replication, and terminal differentiation. However, despite the abundance of work demonstrating the molecular function and identifying binding partners of pRb, the challenge facing molecular biologists and clinical oncologists is how to integrate this vast body of molecular knowledge into the development of targeted therapies for treatment of retinoblastoma. We propose that a more thorough genetic understanding of retinoblastoma would inform targeted treatment decisions and could improve outcomes and quality of life in children affected by this disease.
Collapse
Affiliation(s)
- Uma M Sachdeva
- Scheie Eye Institute, University of Pennsylvania, 51 N. 39th St., Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
48
|
Kavanaugh GM, Wise-Draper TM, Morreale RJ, Morrison MA, Gole B, Schwemberger S, Tichy ED, Lu L, Babcock GF, Wells JM, Drissi R, Bissler JJ, Stambrook PJ, Andreassen PR, Wiesmüller L, Wells SI. The human DEK oncogene regulates DNA damage response signaling and repair. Nucleic Acids Res 2011; 39:7465-76. [PMID: 21653549 PMCID: PMC3177200 DOI: 10.1093/nar/gkr454] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 05/16/2011] [Indexed: 12/04/2022] Open
Abstract
The human DEK gene is frequently overexpressed and sometimes amplified in human cancer. Consistent with oncogenic functions, Dek knockout mice are partially resistant to chemically induced papilloma formation. Additionally, DEK knockdown in vitro sensitizes cancer cells to DNA damaging agents and induces cell death via p53-dependent and -independent mechanisms. Here we report that DEK is important for DNA double-strand break repair. DEK depletion in human cancer cell lines and xenografts was sufficient to induce a DNA damage response as assessed by detection of γH2AX and FANCD2. Phosphorylation of H2AX was accompanied by contrasting activation and suppression, respectively, of the ATM and DNA-PK pathways. Similar DNA damage responses were observed in primary Dek knockout mouse embryonic fibroblasts (MEFs), along with increased levels of DNA damage and exaggerated induction of senescence in response to genotoxic stress. Importantly, Dek knockout MEFs exhibited distinct defects in non-homologous end joining (NHEJ) when compared to their wild-type counterparts. Taken together, the data demonstrate new molecular links between DEK and DNA damage response signaling pathways, and suggest that DEK contributes to DNA repair.
Collapse
Affiliation(s)
- Gina M. Kavanaugh
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Trisha M. Wise-Draper
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Richard J. Morreale
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Monique A. Morrison
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Boris Gole
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sandy Schwemberger
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Elisia D. Tichy
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lu Lu
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - George F. Babcock
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - James M. Wells
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rachid Drissi
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - John J. Bissler
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Peter J. Stambrook
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Paul R. Andreassen
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lisa Wiesmüller
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Susanne I. Wells
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
49
|
Privette Vinnedge LM, McClaine R, Wagh PK, Wikenheiser-Brokamp KA, Waltz SE, Wells SI. The human DEK oncogene stimulates β-catenin signaling, invasion and mammosphere formation in breast cancer. Oncogene 2011; 30:2741-52. [PMID: 21317931 PMCID: PMC3117026 DOI: 10.1038/onc.2011.2] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 01/03/2011] [Accepted: 01/03/2011] [Indexed: 12/16/2022]
Abstract
Breast cancer is a major cause of cancer-related deaths in American women; therefore, the identification of novel breast cancer-related molecules for the discovery of new markers and drug targets remains essential. The human DEK gene, which encodes a chromatin-binding protein and DNA topology regulator, is upregulated in many types of cancer. DEK has been implicated as an oncogene in breast cancer based on mRNA expression studies, but its functional significance in breast cancer growth and progression has not yet been tested directly. We demonstrate that DEK is highly expressed in breast cancer cells compared with normal tissue, and functionally important for cellular growth, invasion and mammosphere formation. DEK overexpression in non-tumorigenic MCF10A cells resulted in increased growth and motility, with a concomitant downregulation of E-cadherin. Conversely, DEK knockdown in MCF7 and MDA-MB-468 breast cancer cells resulted in decreased growth and motility with upregulation of E-cadherin. The use of DEK-proficient and -deficient breast cancer cells in orthotopic xenografts provided further in vivo evidence that DEK contributes to tumor growth. Activation of the β-catenin signaling pathway is important for normal and cancer stem cell character, growth and metastasis. We show that DEK expression stimulated, and DEK knockdown repressed β-catenin nuclear translocation and activity. Importantly, the expression of constitutively active β-catenin rescued breast cancer invasion defects of DEK knockdown cells. Together, our data indicate that DEK expression stimulates the growth, stem cell character and motility of breast cancer cells, and that DEK-dependent cellular invasion occurs at least in part via β-catenin activation.
Collapse
Affiliation(s)
- L M Privette Vinnedge
- Department of Hematology and Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | |
Collapse
|
50
|
Kappes F, Waldmann T, Mathew V, Yu J, Zhang L, Khodadoust MS, Chinnaiyan AM, Luger K, Erhardt S, Schneider R, Markovitz DM. The DEK oncoprotein is a Su(var) that is essential to heterochromatin integrity. Genes Dev 2011; 25:673-8. [PMID: 21460035 PMCID: PMC3070930 DOI: 10.1101/gad.2036411] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Accepted: 02/09/2011] [Indexed: 12/18/2022]
Abstract
Heterochromatin integrity is crucial for genome stability and regulation of gene expression, but the factors involved in mammalian heterochromatin biology are only incompletely understood. Here we identify the oncoprotein DEK, an abundant nuclear protein with a previously enigmatic in vivo function, as a Suppressor of Variegation [Su(var)] that is crucial to global heterochromatin integrity. We show that DEK interacts directly with Heterochromatin Protein 1 α (HP1α) and markedly enhances its binding to trimethylated H3K9 (H3K9me3), which is key for maintaining heterochromatic regions. Loss of Dek in Drosophila leads to a Su(var) phenotype and global reduction in heterochromatin. Thus, these findings show that DEK is a key factor in maintaining the balance between heterochromatin and euchromatin in vivo.
Collapse
Affiliation(s)
- Ferdinand Kappes
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | - Tanja Waldmann
- Max-Planck-Institute for Immunobiology, 79108 Freiburg, Germany
| | - Veena Mathew
- CellNetworks-Cluster of Excellence, ZMBH-DKFZ-Alliance, ZMBH, Heidelberg University, Heidelberg 69120, Germany
| | - Jindan Yu
- Department of Pathology, Michigan Center for Translational Pathology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | - Ling Zhang
- Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Michael S. Khodadoust
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
- Program in Immunology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | - Arul M. Chinnaiyan
- Department of Pathology, Michigan Center for Translational Pathology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | - Karolin Luger
- Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Sylvia Erhardt
- CellNetworks-Cluster of Excellence, ZMBH-DKFZ-Alliance, ZMBH, Heidelberg University, Heidelberg 69120, Germany
| | | | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
- Program in Immunology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
- Cellular and Molecular Biology Program, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| |
Collapse
|