1
|
Hopper MA, Dropik AR, Walker JS, Novak JP, Laverty MS, Manske MK, Wu X, Wenzl K, Krull JE, Sarangi V, Maurer MJ, Yang ZZ, Del Busso MD, Habermann TM, Link BK, Rimsza LM, Witzig TE, Ansell SM, Cerhan JR, Jevremovic D, Novak AJ. DEK regulates B-cell proliferative capacity and is associated with aggressive disease in low-grade B-cell lymphomas. Blood Cancer J 2024; 14:172. [PMID: 39384745 PMCID: PMC11464677 DOI: 10.1038/s41408-024-01145-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 10/11/2024] Open
Abstract
This study sheds light on the pivotal role of the oncoprotein DEK in B-cell lymphoma. We reveal DEK expression correlates with increased tumor proliferation and inferior overall survival in cases diagnosed with low-grade B-cell lymphoma (LGBCL). We also found significant correlation between DEK expression and copy number alterations in LGBCL tumors, highlighting a novel mechanism of LGBCL pathogenesis that warrants additional exploration. To interrogate the mechanistic role of DEK in B-cell lymphoma, we generated a DEK knockout cell line model, which demonstrated DEK depletion caused reduced proliferation and altered expression of key cell cycle and apoptosis-related proteins, including Bcl-2, Bcl-xL, and p53. Notably, DEK depleted cells showed increased sensitivity to apoptosis-inducing agents, including venetoclax and staurosporine, which underscores the therapeutic potential of targeting DEK in B-cell lymphomas. Overall, our study contributes to a better understanding of DEK's role as an oncoprotein in B-cell lymphomas, highlighting its potential as both a promising therapeutic target and a novel biomarker for aggressive LGBCL. Further research elucidating the molecular mechanisms underlying DEK-mediated tumorigenesis could pave the way for improved treatment strategies and better clinical outcomes for patients with B-cell lymphoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaosheng Wu
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Kerstin Wenzl
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Matthew J Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Brian K Link
- Division of Hematology, Oncology, and Bone & Marrow Transplantation, University of Iowa, Iowa City, IA, USA
| | - Lisa M Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, USA
| | | | | | - James R Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Dragan Jevremovic
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Anne J Novak
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Ullah MI, Mikhailova MV, Alkhathami AG, Carbajal NC, Zuta MEC, Rasulova I, Najm MAA, Abosoda M, Alsalamy A, Deorari M. Molecular pathways in the development of HPV-induced oropharyngeal cancer. Cell Commun Signal 2023; 21:351. [PMID: 38098017 PMCID: PMC10722793 DOI: 10.1186/s12964-023-01365-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/25/2023] [Indexed: 12/17/2023] Open
Abstract
Oropharyngeal cancer, a subset of head and neck cancer, is increasingly recognized as a unique clinical entity primarily influenced by high-risk human papillomavirus (HPV) infections, particularly HPV-16. This review delves into the viral life cycle of HPV-16 and its interactions with host cells, with a specific focus on the crucial roles played by the viral oncoproteins E6 and E7. These oncoproteins drive cellular proliferation by targeting critical tumor suppressor proteins like p53 and Rb, resulting in uncontrolled cell growth and genomic instability. Furthermore, the significance of epigenetic modifications induced by HPV-16 and their implications is important for cancer progression. This comprehensive review provides valuable insights into the intricate molecular landscape of HPV-induced oropharyngeal cancer, shedding light on the development of targeted therapies and preventive strategies for this emerging global health concern. Video Abstract.
Collapse
Affiliation(s)
- Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka-72388, Aljouf, Saudi Arabia
| | - Maria V Mikhailova
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| | - Ali G Alkhathami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Nestor Cuba Carbajal
- Doctor en Gestión Pública y Gobernabilidad, Docente en La Universidad Norbert Wiener, Lima, Perú.
| | | | - Irodakhon Rasulova
- School of Humanities, Natural & Social Sciences, New Uzbekistan University, 54 Mustaqillik Ave, 100007, Tashkent, Uzbekistan
- Department of Public Health, Tashkent Pediatric Medical Institute, Bogishamol Street 223, Tashkent, Uzbekistan
| | - Mazin A A Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Nasiriyah, Iraq
| | - Munther Abosoda
- College of Pharmacy, the Islamic University, Najaf, Iraq
- College of Pharmacy, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, the Islamic University of Babylon, Hillah, Iraq
| | - Ali Alsalamy
- College of Pharmacy, Imam Ja'afar Al-Sadiq University, Al-Muthanna, 66002, Iraq
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
3
|
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: 2.0] [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
|
4
|
Greene AN, Solomon MB, Privette Vinnedge LM. Novel molecular mechanisms in Alzheimer's disease: The potential role of DEK in disease pathogenesis. Front Aging Neurosci 2022; 14:1018180. [PMID: 36275000 PMCID: PMC9582447 DOI: 10.3389/fnagi.2022.1018180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease and age-related dementias (AD/ADRD) are debilitating diseases that exact a significant physical, emotional, cognitive, and financial toll on the individual and their social network. While genetic risk factors for early-onset AD have been identified, the molecular and genetic drivers of late-onset AD, the most common subtype, remain a mystery. Current treatment options are limited for the 35 million people in the United States with AD/ADRD. Thus, it is critically important to identify novel molecular mechanisms of dementia-related pathology that may be targets for the development of new interventions. Here, we summarize the overarching concepts regarding AD/ADRD pathogenesis. Then, we highlight one potential molecular driver of AD/ADRD, the chromatin remodeling protein DEK. We discuss in vitro, in vivo, and ex vivo findings, from our group and others, that link DEK loss with the cellular, molecular, and behavioral signatures of AD/ADRD. These include associations between DEK loss and cellular and molecular hallmarks of AD/ADRD, including apoptosis, Tau expression, and Tau hyperphosphorylation. We also briefly discuss work that suggests sex-specific differences in the role of DEK in AD/ADRD pathogenesis. Finally, we discuss future directions for exploiting the DEK protein as a novel player and potential therapeutic target for the treatment of AD/ADRD.
Collapse
Affiliation(s)
- Allie N. Greene
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Matia B. Solomon
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Department of Psychology, University of Cincinnati, Cincinnati, OH, United States
| | - Lisa M. Privette Vinnedge
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
5
|
Greene AN, Nguyen ET, Paranjpe A, Lane A, Privette Vinnedge LM, Solomon MB. In silico gene expression and pathway analysis of DEK in the human brain across the lifespan. Eur J Neurosci 2022; 56:4720-4743. [PMID: 35972263 PMCID: PMC9730547 DOI: 10.1111/ejn.15791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/15/2022] [Accepted: 08/08/2022] [Indexed: 11/30/2022]
Abstract
DEK, a chromatin-remodelling phosphoprotein, is associated with various functions and biological pathways in the periphery, including inflammation, oncogenesis, DNA repair, and transcriptional regulation. We recently identified an association between DEK loss and central nervous system diseases, such as Alzheimer's. To understand DEK's potential role in disease, it is critical to characterize DEK in healthy human brain to distinguish between neural DEK expression and function in healthy versus diseased states like dementia. We utilized two public databases, BrainCloud and Human Brain Transcriptome, and analysed DEK mRNA expression across the lifespan in learning and memory relevant brain regions. Since DEK loss induces phenotypes associated with brain ageing (e.g., DNA damage and apoptosis), we hypothesized that neural DEK expression may be highest during foetal development and lower in elderly individuals. In agreement with this hypothesis, DEK was most prominently expressed during foetal development in all queried forebrain areas, relative to other ages. Consistent with its roles in the periphery, pathways related to DEK in the brain were associated with cellular proliferation, DNA replication and repair, apoptosis, and inflammation. We also found novel neural development-relevant pathways (e.g., synaptic transmission, neurite outgrowth, and myelination) to be enriched from genes correlated with DEK expression. These findings suggest that DEK is important for human brain development. Overall, we highlight age-related changes in neural DEK expression across the human lifespan and illuminate novel biological pathways associated with DEK that are distinct from normal brain ageing. These findings may further our understanding of how DEK impacts brain function and disease susceptibility.
Collapse
Affiliation(s)
- Allie N. Greene
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA 45267
| | | | - Aditi Paranjpe
- Division of Biomedical Informatics, Bioinformatics Collaborative Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Lisa M. Privette Vinnedge
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Matia B. Solomon
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA 45267
- Department of Psychology, University of Cincinnati, Cincinnati, OH 45237
| |
Collapse
|
6
|
Yang YS, Jia XZ, Lu QY, Cai SL, Huang XT, Yang SH, Wood C, Wang YH, Zhou JJ, Chen YD, Yang JS, Yang WJ. Exosomal DEK removes chemoradiotherapy resistance by triggering quiescence exit of breast cancer stem cells. Oncogene 2022; 41:2624-2637. [PMID: 35351996 DOI: 10.1038/s41388-022-02278-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/11/2022] [Indexed: 02/07/2023]
Abstract
Tumor therapeutics often target the primary tumor bulk but fail to eradicate therapy-resistant cancer stem cells (CSCs) in quiescent state. These can then become activated to initiate recurrence and/or metastasis beyond therapy. Here, we identified and isolated chemoradiotherapy-resistant CSCs in quiescent state with high capacity of tumor-initiation and tumorsphere formation from three types of breast tumors in mice. Experiments of knockdown and rescue revealed DEK, a nuclear protein, as essential for CSC activation. Exogenous DEK was then used to trigger quiescence exit of CSCs. ChIP-seq and ATAC-seq showed that DEK directly binds to chromatin, facilitating its genome-wide accessibility. The resulting epigenetic events upregulate the expression of cellular activation-related genes including MYC targets, whereas cellular quiescence-related genes including the p53 signaling pathway are silenced. However, twinned with DEK-induced activation, formerly resistant CSCs were then destroyed by chemotherapy in vitro. In mice, traditional chemoradiotherapy concurrent with the injection of DEK-containing exosomes resulted in eradication of primary tumors together with formerly resistant CSCs without recurrence or metastasis. Our findings advance knowledge of the mechanism of quiescent CSC activation and may provide novel clinical opportunities for removal of quiescence-linked therapy resistance.
Collapse
Affiliation(s)
- Yao-Shun Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xi-Zheng Jia
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qian-Yun Lu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Sun-Li Cai
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xue-Ting Huang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shu-Hua Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chris Wood
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yue-Hong Wang
- Department of Respiratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiao-Jiao Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yi-Ding Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jin-Shu Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wei-Jun Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
7
|
Sharif SB, Zamani N, Chadwick BP. BAZ1B the Protean Protein. Genes (Basel) 2021; 12:genes12101541. [PMID: 34680936 PMCID: PMC8536118 DOI: 10.3390/genes12101541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 02/02/2023] Open
Abstract
The bromodomain adjacent to the zinc finger domain 1B (BAZ1B) or Williams syndrome transcription factor (WSTF) are just two of the names referring the same protein that is encoded by the WBSCR9 gene and is among the 26-28 genes that are lost from one copy of 7q11.23 in Williams syndrome (WS: OMIM 194050). Patients afflicted by this contiguous gene deletion disorder present with a range of symptoms including cardiovascular complications, developmental defects as well as a characteristic cognitive and behavioral profile. Studies in patients with atypical deletions and mouse models support BAZ1B hemizygosity as a contributing factor to some of the phenotypes. Focused analysis on BAZ1B has revealed this to be a versatile nuclear protein with a central role in chromatin remodeling through two distinct complexes as well as being involved in the replication and repair of DNA, transcriptional processes involving RNA Polymerases I, II, and III as well as possessing kinase activity. Here, we provide a comprehensive review to summarize the many aspects of BAZ1B function including its recent link to cancer.
Collapse
Affiliation(s)
- Shahin Behrouz Sharif
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA;
| | - Nina Zamani
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA;
| | - Brian P. Chadwick
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA;
- Correspondence:
| |
Collapse
|
8
|
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: 1.0] [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
|
9
|
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: 1.7] [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
|
10
|
DEK is highly expressed in breast cancer and is associated with malignant phenotype and progression. Oncol Lett 2021; 21:440. [PMID: 33868478 PMCID: PMC8045159 DOI: 10.3892/ol.2021.12701] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/10/2021] [Indexed: 01/21/2023] Open
Abstract
DEK proto-oncogene (DEK) has been demonstrated as an oncogene and is associated with the development of many types of tumor; however, the expression and role of DEK in breast cancer remain unknown. The present study aimed to determine the role of DEK in the progression of breast cancer. The expression of DEK in 110 breast cancer tissues and 50 adjacent normal breast tissues was examined using immunohistochemistry. Furthermore, DEK expression was upregulated by DEK transfection or downregulated by DEK shRNA interference in MCF7 cells. Proliferative and invasive abilities were examined in MCF7 cells using MTT assay, colony-formation assay and transwell invasion assays. The results demonstrated that DEK expression level was significantly increased in breast cancer tissues compared with normal breast tissues. Furthermore, high DEK expression was associated with high histological grade, lymph node metastasis, advanced Tumor-Node-Metastasis stage and high Ki-67 index; however, DEK expression was not associated with the expression level of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. High DEK expression indicated poor prognosis in patients with breast cancer. DEK overexpression upregulated the protein expression of β-catenin and Wnt and increased the proliferative and invasive abilities of breast cancer cells. DEK downregulation had the opposite effect. Taken together, the results from the present study demonstrated that high expression of DEK was common in patients with breast cancer and was associated with progression of the disease and poor prognosis, and that DEK overexpression promoted the proliferative and invasive abilities of breast cancer cells.
Collapse
|
11
|
Mir BA, Rahaman PF, Ahmad A. Viral load and interaction of HPV oncoprotein E6 and E7 with host cellular markers in the progression of cervical cancer. AIMS MOLECULAR SCIENCE 2021. [DOI: 10.3934/molsci.2021014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
<abstract>
<p>Cervical cancer is the sequel of a multi-factorial, long-term unresolved disease that includes genetic, epigenetic, and viral components responsible for its development and progression. It is the second most common cancer of females in India. Human papillomavirus (HPV) is considered the primary causative agent of pre-neoplastic and cancerous lesions and 90% of all cervical carcinomas are linked to high-risk HPV type 16 and type 18. Although most HR-HPV infections are asymptomatic, transient, and self-limiting, the persistent infection with a high risk (HR-HPV) may cause precancerous lesions that can progress to cervical cancer. HPV type 16 is the most common HPV in India associated with more than 75% of cervical cancer, followed by HPV type 18 and other high-risk types. Infection with HPV alone is not sufficient for the development of cervical cancer but there is the involvement of some host genetic factors also that are responsible for the development and progression of cervical cancer. This article briefly reviews molecular pathogenesis, viral load, and the interaction of HPV oncoprotein E6 and E7 with host cellular markers in the progression of cervical cancer.</p>
</abstract>
Collapse
|
12
|
Olmedo-Nieva L, Muñoz-Bello JO, Manzo-Merino J, Lizano M. New insights in Hippo signalling alteration in human papillomavirus-related cancers. Cell Signal 2020; 76:109815. [PMID: 33148514 DOI: 10.1016/j.cellsig.2020.109815] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/20/2020] [Accepted: 10/20/2020] [Indexed: 02/09/2023]
Abstract
The persistent infection with high-risk human papillomavirus (HPV) is an etiologic factor for the development of different types of cancers, mainly attributed to the continuous expression of E6 and E7 HPV oncoproteins, which regulate several cell signalling pathways including the Hippo pathway. It has been demonstrated that E6 proteins promote the increase of the Hippo elements YAP, TAZ and TEAD, at protein level, as well as their transcriptional targets. Also, E6 and E7 oncoproteins promote nuclear YAP localization and a decrease in YAP negative regulators such as MST1, PTPN14 or SOCS6. Interestingly, Hippo signalling components modulate HPV activity, such as TEAD1 and the transcriptional co-factor VGLL1, induce the activation of HPV early and late promoters, while hyperactivation of YAP in specific cells facilitates virus infection by increasing putative HPV receptors and by evading innate immunity. Additionally, alterations in Hippo signalling elements have been found in HPV-related cancers and particularly, the involvement of HPV oncoproteins on the regulation of some of these Hippo components has been also proposed, although the precise mechanisms remain unclear. The present review addresses the recent findings describing the interplay between HPV and Hippo signalling in HPV-related cancers, a fact that highlights the importance of developing more in-depth studies in this field to establish key therapeutic targets.
Collapse
Affiliation(s)
- Leslie Olmedo-Nieva
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico; Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - J Omar Muñoz-Bello
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico; Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Sede sur, Mexico City 14330, Mexico
| | - Joaquín Manzo-Merino
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico; Cátedras CONACyT-Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Marcela Lizano
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico.
| |
Collapse
|
13
|
Ishida K, Nakashima T, Shibata T, Hara A, Tomita H. Role of the DEK oncogene in the development of squamous cell carcinoma. Int J Clin Oncol 2020; 25:1563-1569. [PMID: 32656741 PMCID: PMC7441080 DOI: 10.1007/s10147-020-01735-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/21/2020] [Indexed: 01/21/2023]
Abstract
DEK is a highly conserved nuclear factor that plays an important role in the regulation of multiple cellular processes. DEK was discovered to be an oncogene as a fusion with NUP214 gene, which results in producing DEK-NUP214 proteins, in a subset of patients with acute myeloid leukemia. Subsequently, DEK overexpression was reported in many cancers, thus DEK itself is considered to be an oncoprotein. DEK has been reported to play important roles in the progression of early and late stage squamous cell carcinoma (SCC) and is useful for early diagnosis of the disease. These findings have made DEK an attractive therapeutic target, especially for human papillomavirus (HPV)-associated SCC. However, the mechanism of DEK in SCC remains unclear. In this review, we discuss human DEK oncogene-related SCC.
Collapse
Affiliation(s)
- Kazuhisa Ishida
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
- Department of Oral Maxillofacial Surgery, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Takayuki Nakashima
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
- Department of Oral Maxillofacial Surgery, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Toshiyuki Shibata
- Department of Oral Maxillofacial Surgery, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan.
| |
Collapse
|
14
|
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: 10] [Impact Index Per Article: 2.5] [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
|
15
|
Ganz M, Vogel C, Czada C, Jörke V, Gwosch EC, Kleiner R, Pierzynska-Mach A, Zanacchi FC, Diaspro A, Kappes F, Bürkle A, Ferrando-May E. The oncoprotein DEK affects the outcome of PARP1/2 inhibition during mild replication stress. PLoS One 2019; 14:e0213130. [PMID: 31408463 PMCID: PMC6692024 DOI: 10.1371/journal.pone.0213130] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/03/2019] [Indexed: 01/07/2023] Open
Abstract
DNA replication stress is a major source of genomic instability and is closely linked to tumor formation and progression. Poly(ADP-ribose)polymerases1/2 (PARP1/2) enzymes are activated in response to replication stress resulting in poly(ADP-ribose) (PAR) synthesis. PARylation plays an important role in the remodelling and repair of impaired replication forks, providing a rationale for targeting highly replicative cancer cells with PARP1/2 inhibitors. The human oncoprotein DEK is a unique, non-histone chromatin architectural protein whose deregulated expression is associated with the development of a wide variety of human cancers. Recently, we showed that DEK is a high-affinity target of PARylation and that it promotes the progression of impaired replication forks. Here, we investigated a potential functional link between PAR and DEK in the context of replication stress. Under conditions of mild replication stress induced either by topoisomerase1 inhibition with camptothecin or nucleotide depletion by hydroxyurea, we found that the effect of acute PARP1/2 inhibition on replication fork progression is dependent on DEK expression. Reducing DEK protein levels also overcomes the restart impairment of stalled forks provoked by blocking PARylation. Non-covalent DEK-PAR interaction via the central PAR-binding domain of DEK is crucial for counteracting PARP1/2 inhibition as shown for the formation of RPA positive foci in hydroxyurea treated cells. Finally, we show by iPOND and super resolved microscopy that DEK is not directly associated with the replisome since it binds to DNA at the stage of chromatin formation. Our report sheds new light on the still enigmatic molecular functions of DEK and suggests that DEK expression levels may influence the sensitivity of cancer cells to PARP1/2 inhibitors.
Collapse
Affiliation(s)
- Magdalena Ganz
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz, Germany
| | - Christopher Vogel
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz, Germany
| | - Christina Czada
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz, Germany
| | - Vera Jörke
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz, Germany
| | - Eva Christina Gwosch
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz, Germany
| | - Rebecca Kleiner
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz, Germany
| | - Agnieszka Pierzynska-Mach
- Nanoscopy and NIC@IIT, Istituto Italiano di Tecnologia, Genoa, Italy
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Francesca Cella Zanacchi
- Nanoscopy and NIC@IIT, Istituto Italiano di Tecnologia, Genoa, Italy
- Biophysics Institute (IBF), National Research Council (CNR), Genoa, Italy
| | - Alberto Diaspro
- Nanoscopy and NIC@IIT, Istituto Italiano di Tecnologia, Genoa, Italy
- DIFILAB, Department of Physics, University of Genoa, Genoa, Italy
| | - Ferdinand Kappes
- Xi’an Jiaotong-Liverpool University, Dushu Lake Higher Education Town, Suzhou, China
| | - Alexander Bürkle
- Department of Biology, Molecular Toxicology Group, University of Konstanz, Konstanz, Germany
| | - Elisa Ferrando-May
- Department of Biology, Bioimaging Center, University of Konstanz, Konstanz, Germany
| |
Collapse
|
16
|
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: 2] [Impact Index Per Article: 0.4] [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
|
17
|
Wen Y, Xu HN, Privette Vinnedge L, Feng M, Li LZ. Optical Redox Imaging Detects the Effects of DEK Oncogene Knockdown on the Redox State of MDA-MB-231 Breast Cancer Cells. Mol Imaging Biol 2019; 21:410-416. [PMID: 30758703 PMCID: PMC6684344 DOI: 10.1007/s11307-019-01321-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Optical redox imaging (ORI), based on collecting the endogenous fluorescence of reduced nicotinamide adenine dinucleotide (NADH) and oxidized flavoproteins (Fp) containing a redox cofactor flavin adenine dinucleotide (FAD), provides sensitive indicators of cellular metabolism and redox status. ORI indices (such as NADH, FAD, and their ratio) have been under investigation as potential progression/prognosis biomarkers for cancer. Higher FAD redox ratio (i.e., FAD/(FAD + NADH)) has been associated with higher invasive/metastatic potential in tumor xenografts and cultured cells. This study is to examine whether ORI indices can respond to the modulation of oncogene DEK activities that change cancer cell invasive/metastatic potential. PROCEDURES Using lentiviral shRNA, DEK gene expression was efficiently knocked down in MDA-MB-231 breast cancer cells (DEKsh). These DEKsh cells, along with scrambled shRNA-transduced control cells (NTsh), were imaged with a fluorescence microscope. In vitro invasive potential of the DEKsh cells and NTsh cells was also measured in parallel using the transwell assay. RESULTS FAD and FAD redox ratios in polyclonal cells with DEKsh were significantly lower than that in NTsh control cells. Consistently, the DEKsh cells demonstrated decreased invasive potential than their non-knockdown counterparts NTsh cells. CONCLUSIONS This study provides direct evidence that oncogene activities could mediate ORI-detected cellular redox state.
Collapse
Affiliation(s)
- Yu Wen
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - He N Xu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lisa Privette Vinnedge
- Cincinnati Children's Hospital Medical Center, Cancer and Blood Diseases Institute, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Min Feng
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Z Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center and Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
18
|
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: 0.5] [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
|
19
|
Zhou QC, Deng XF, Yang J, Jiang H, Qiao MX, Liu HH, Qian Z, Hou LL, Hu HG. Oncogene DEK is highly expressed in lung cancerous tissues and positively regulates cell proliferation as well as invasion. Oncol Lett 2018; 15:8573-8581. [PMID: 29844811 PMCID: PMC5958825 DOI: 10.3892/ol.2018.8436] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 03/16/2018] [Indexed: 11/06/2022] Open
Abstract
DEK is a protein ubiquitously expressed in multicellular organisms as well as certain unicellular organisms. It is associated with the regulation of cell proliferation, differentiation, migration, apoptosis, senescence, self-renewal and DNA repairing. In tumor cells it is associated with the carcinogenesis process, however there have been few previous studies into the expression of DEK in lung cancer. In the present study the expression level of DEK mRNA and protein was detected in lung cancer tissues and non-cancerous counterparts by performing reverse transcription-quantitative polymerase chain reaction and immunohistochemical staining. It was revealed that the expression of DEK was increased in lung cancer tissues compared with normal tissue. Knock-down and over-expression of DEK in A549 cells were performed to determine the role of DEK in tumor formation. An MTT assay, colony formation assay and Matrigel invasion assay demonstrated that DEK positively regulated cell proliferation and invasion. These results suggest that DEK is highly expressed in lung cancer tissues and positively regulates cell proliferation and invasion.
Collapse
Affiliation(s)
- Qian-Cheng Zhou
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Xue-Feng Deng
- Department of Cardio-Thoracic Surgery, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, P.R. China
| | - Juan Yang
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Hong Jiang
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Ming-Xu Qiao
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Huan-Huan Liu
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Zhen Qian
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Ling-Ling Hou
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Hong-Gang Hu
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| |
Collapse
|
20
|
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: 1.8] [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
|
21
|
Martinez-Useros J, Moreno I, Fernandez-Aceñero MJ, Rodriguez-Remirez M, Borrero-Palacios A, Cebrian A, Gomez del Pulgar T, del Puerto-Nevado L, Li W, Puime-Otin A, Perez N, Soengas MS, Garcia-Foncillas J. The potential predictive value of DEK expression for neoadjuvant chemoradiotherapy response in locally advanced rectal cancer. BMC Cancer 2018; 18:144. [PMID: 29409457 PMCID: PMC5801838 DOI: 10.1186/s12885-018-4048-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 01/24/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Limited data are available regarding the ability of biomarkers to predict complete pathological response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. Complete response translates to better patient survival. DEK is a transcription factor involved not only in development and progression of different types of cancer, but is also associated with treatment response. This study aims to analyze the role of DEK in complete pathological response following chemoradiotherapy for locally advanced rectal cancer. METHODS Pre-treated tumour samples from 74 locally advanced rectal-cancer patients who received chemoradiation therapy prior to total mesorectal excision were recruited for construction of a tissue microarray. DEK immunoreactivity from all samples was quantified by immunohistochemistry. Then, association between positive stained tumour cells and pathologic response to neoadjuvant treatment was measured to determine optimal predictive power. RESULTS DEK expression was limited to tumour cells located in the rectum. Interestingly, high percentage of tumour cells with DEK positiveness was statistically associated with complete pathological response to neoadjuvant treatment based on radiotherapy and fluoropyrimidine-based chemotherapy and a marked trend toward significance between DEK positiveness and absence of treatment toxicity. Further analysis revealed an association between DEK and the pro-apoptotic factor P38 in the pre-treated rectal cancer biopsies. CONCLUSIONS These data suggest DEK as a potential biomarker of complete pathological response to treatment in locally advanced rectal cancer.
Collapse
Affiliation(s)
- J. Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - I. Moreno
- Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | | | - M. Rodriguez-Remirez
- Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - A. Borrero-Palacios
- Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - A. Cebrian
- Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - T. Gomez del Pulgar
- Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - L. del Puerto-Nevado
- Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - W. Li
- Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - A. Puime-Otin
- Department of Pathology, University Hospital “Fundación Jiménez Díaz”-UAM, Madrid, Spain
| | - N. Perez
- Department of Pathology, University Hospital “Fundación Jiménez Díaz”-UAM, Madrid, Spain
| | - M. S. Soengas
- Melanoma Research Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - J. Garcia-Foncillas
- Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| |
Collapse
|
22
|
Abstract
Histone chaperones are indispensable regulators of chromatin structure and function. Recent work has shown that they are frequently mis-regulated in cancer, which can have profound consequences on tumor growth and survival. Here, we focus on chaperones for the essential H3 histone variants H3.3 and CENP-A, specifically HIRA, DAXX/ATRX, DEK, and HJURP. This review summarizes recent studies elucidating their roles in regulating chromatin and discusses how cancer-specific chromatin interactions can be exploited to target cancer cells.
Collapse
Affiliation(s)
- Jonathan Nye
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniël P Melters
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yamini Dalal
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
23
|
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: 2.5] [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
|
24
|
Wise-Draper T, Sendilnathan A, Palackdharry S, Pease N, Qualtieri J, Butler R, Sadraei NH, Morris JC, Patil Y, Wilson K, Mark J, Casper K, Takiar V, Lane A, Privette Vinnedge L. Decreased plasma DEK Oncogene Levels Correlate with p16-Negative Disease and Advanced Tumor Stage in a Case-Control Study of Patients with Head and Neck Squamous Cell Carcinoma. Transl Oncol 2017; 11:168-174. [PMID: 29289845 PMCID: PMC6002348 DOI: 10.1016/j.tranon.2017.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 12/17/2022] Open
Abstract
Head and neck cancer (HNC) remains the sixth most common malignancy worldwide and survival upon recurrence and/or metastasis remains poor. HNSCC has traditionally been associated with alcohol and nicotine use, but more recently the Human Papilloma Virus (HPV) has emerged as a favorable prognostic risk factor for oropharyngeal HNSCC. However, further stratification with additional biomarkers to predict patient outcome continues to be essential. One candidate biomarker is the DEK oncogenic protein, which was previously detected in the urine of patients with bladder cancer and is known to be secreted by immune cells such as macrophages. Here, we investigated if DEK could be detected in human plasma and if DEK levels correlated with clinical and pathological variables of HNSCC. Plasma was separated from the peripheral blood of newly diagnosed, untreated HNSCC patients or age-matched normal healthy controls and analyzed for DEK protein using ELISA. Plasma concentrations of DEK protein were lower in p16-negative tumors compared to both normal controls and patients with p16-positive tumors. Patients with lower plasma concentrations of DEK were also more likely to have late stage tumors and a lower white blood cell count. Contrary to previously published work demonstrating a poor prognosis with high intratumoral DEK levels, we show for the first time that decreased concentrations of DEK in patient plasma correlates with poor prognostic factors, including HPV-negative status as determined by negative p16 expression and advanced tumor stage.
Collapse
Affiliation(s)
- Trisha Wise-Draper
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267.
| | - Arun Sendilnathan
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267
| | - Sarah Palackdharry
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267
| | - Nicholas Pease
- Department of Bioengineering, University of Washington, Seattle, WA 98105
| | - Julianne Qualtieri
- Department of Pathology, University of Cincinnati, Cincinnati, OH, 45267
| | - Randall Butler
- Department of Pathology, University of Cincinnati, Cincinnati, OH, 45267
| | - Nooshin Hashemi Sadraei
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267
| | - John C Morris
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267
| | - Yash Patil
- Department of Otolaryngology- Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, 45267
| | - Keith Wilson
- Department of Otolaryngology- Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, 45267
| | - Jonathan Mark
- Department of Otolaryngology- Head and Neck Surgery, University of Cincinnati, Cincinnati, OH, 45267
| | - Keith Casper
- Department of Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Vinita Takiar
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, 45267
| | - Adam Lane
- Cancer and Blood Diseases Institute, Cincinnati Children's Medical Center, Cincinnati, OH, 45229
| | - Lisa Privette Vinnedge
- Cancer and Blood Diseases Institute, Cincinnati Children's Medical Center, Cincinnati, OH, 45229.
| |
Collapse
|
25
|
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: 1.1] [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
|
26
|
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: 3.9] [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
|
27
|
Çalışkaner ZO, Çakar T, Özçelik E, Özdilek A, Kim AS, Doğan Ö, Bosompem A, Grosveld G, Saka B, Kandilci A. DEK protein level is a biomarker of CD138positive normal and malignant plasma cells. PLoS One 2017; 12:e0178025. [PMID: 28558048 PMCID: PMC5448761 DOI: 10.1371/journal.pone.0178025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/11/2017] [Indexed: 11/22/2022] Open
Abstract
Overexpression of DEK oncogene is associated with increased proliferation of carcinoma cells and it is observed in several solid tumors due to the amplification of the 6p22.3 chromosomal region where DEK locates. Although the same chromosomal amplification occurs in multiple myeloma (MM), a plasma cell neoplasm, whether the expression and the copy number of the DEK gene are affected in MM remains elusive. We show that despite the increased copy number in CD138positive MM cells (4 out of 41 MM samples), DEK mRNA expression was down-regulated compared with that in CD138negative bone marrow (BM) cells of the same patients (P<0.0001). DEK protein was not detectable by immunohistochemistry (IHC) in CD138positive normal plasma cells or in malignant plasma cells of MM patients (n = 56) whereas it was widely expressed in normal and neoplastic B-cells. Stable knockdown or overexpression of DEK in CD138positive MM cell lines did not affect the proliferation and viability of the cells profoundly in the presence or absence of chemotherapeutic agent melphalan whereas knockdown of DEK moderately but significantly increased the expression level of CD138 (p<0.01). Decreased DEK expression in plasma cells suggests a potential role of this gene in plasma cell development and lack of detectable DEK protein by IHC could be used as a biomarker for normal and malignant plasma cells.
Collapse
Affiliation(s)
- Zihni Onur Çalışkaner
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Türkan Çakar
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Emrah Özçelik
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Ahmet Özdilek
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze, Kocaeli, Turkey
| | - Annette S. Kim
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Öner Doğan
- Department of Pathology, Istanbul University, Istanbul Medical Faculty, Istanbul, Turkey
| | - Amma Bosompem
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Gerard Grosveld
- Department of Genetics, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Bülent Saka
- Department of Internal Medicine, Istanbul University, Istanbul Medical Faculty, Istanbul, Turkey
| | - Ayten Kandilci
- Department of Molecular Biology and Genetics, Gebze Technical University, Gebze, Kocaeli, Turkey
| |
Collapse
|
28
|
Smith EA, Gole B, Willis NA, Soria R, Starnes LM, Krumpelbeck EF, Jegga AG, Ali AM, Guo H, Meetei AR, Andreassen PR, Kappes F, Vinnedge LMP, Daniel JA, Scully R, Wiesmüller L, Wells SI. DEK is required for homologous recombination repair of DNA breaks. Sci Rep 2017; 7:44662. [PMID: 28317934 PMCID: PMC5357905 DOI: 10.1038/srep44662] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/13/2017] [Indexed: 12/16/2022] Open
Abstract
DEK is a highly conserved chromatin-bound protein whose upregulation across cancer types correlates with genotoxic therapy resistance. Loss of DEK induces genome instability and sensitizes cells to DNA double strand breaks (DSBs), suggesting defects in DNA repair. While these DEK-deficiency phenotypes were thought to arise from a moderate attenuation of non-homologous end joining (NHEJ) repair, the role of DEK in DNA repair remains incompletely understood. We present new evidence demonstrating the observed decrease in NHEJ is insufficient to impact immunoglobulin class switching in DEK knockout mice. Furthermore, DEK knockout cells were sensitive to apoptosis with NHEJ inhibition. Thus, we hypothesized DEK plays additional roles in homologous recombination (HR). Using episomal and integrated reporters, we demonstrate that HR repair of conventional DSBs is severely compromised in DEK-deficient cells. To define responsible mechanisms, we tested the role of DEK in the HR repair cascade. DEK-deficient cells were impaired for γH2AX phosphorylation and attenuated for RAD51 filament formation. Additionally, DEK formed a complex with RAD51, but not BRCA1, suggesting a potential role regarding RAD51 filament formation, stability, or function. These findings define DEK as an important and multifunctional mediator of HR, and establish a synthetic lethal relationship between DEK loss and NHEJ inhibition.
Collapse
Affiliation(s)
- Eric A. Smith
- Division of Oncology; Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Boris Gole
- Department of Obstetrics and Gynecology; Ulm University, Ulm, 89075, Germany
| | - Nicholas A. Willis
- Department of Medicine, Division of Hematology-Oncology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Rebeca Soria
- Chromatin Structure and Function Group, The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Linda M. Starnes
- Chromatin Structure and Function Group, The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Eric F. Krumpelbeck
- Division of Oncology; Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Anil G. Jegga
- Division of Oncology; Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Abdullah M. Ali
- Division of Experimental Hematology and Cancer Biology; Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Haihong Guo
- Institute of Biochemistry and Molecular Biology; Medical School, RWTH Aachen University, Aachen, 52074, Germany
| | - Amom R. Meetei
- Division of Experimental Hematology and Cancer Biology; Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Paul R. Andreassen
- Division of Experimental Hematology and Cancer Biology; Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Ferdinand Kappes
- Institute of Biochemistry and Molecular Biology; Medical School, RWTH Aachen University, Aachen, 52074, Germany
| | | | - Jeremy A. Daniel
- Chromatin Structure and Function Group, The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Ralph Scully
- Department of Medicine, Division of Hematology-Oncology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology; Ulm University, Ulm, 89075, Germany
| | - Susanne I. Wells
- Division of Oncology; Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| |
Collapse
|
29
|
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: 2.7] [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
|
30
|
Role of WDHD1 in Human Papillomavirus-Mediated Oncogenesis Identified by Transcriptional Profiling of E7-Expressing Cells. J Virol 2016; 90:6071-6084. [PMID: 27099318 DOI: 10.1128/jvi.00513-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 04/16/2016] [Indexed: 01/04/2023] Open
Abstract
UNLABELLED The E7 oncoprotein of the high-risk human papillomavirus (HPV) plays a major role in HPV-induced carcinogenesis. E7 abrogates the G1 cell cycle checkpoint and induces genomic instability, but the mechanism is not fully understood. In this study, we performed RNA sequencing (RNA-seq) to characterize the transcriptional profile of keratinocytes expressing HPV 16 (HPV-16) E7. At the transcriptome level, 236 genes were differentially expressed between E7 and vector control cells. A subset of the differentially expressed genes, most of them novel to E7-expressing cells, was further confirmed by real-time PCR. Of interest, the activities of multiple transcription factors were altered in E7-expressing cells. Through bioinformatics analysis, pathways altered in E7-expressing cells were investigated. The upregulated genes were enriched in cell cycle and DNA replication, as well as in the DNA metabolic process, transcription, DNA damage, DNA repair, and nucleotide metabolism. Specifically, we focused our studies on the gene encoding WDHD1 (WD repeat and high mobility group [HMG]-box DNA-binding protein), one of the genes that was upregulated in E7-expressing cells. WDHD1 is a component of the replisome that regulates DNA replication. Recent studies suggest that WDHD1 may also function as a DNA replication initiation factor as well as a G1 checkpoint regulator. We found that in E7-expressing cells, the steady-state level of WDHD1 protein was increased along with the half-life. Moreover, downregulation of WDHD1 reduced E7-induced G1 checkpoint abrogation and rereplication, demonstrating a novel function for WDHD1. These studies shed light on mechanisms by which HPV induces genomic instability and have therapeutic implications. IMPORTANCE The high-risk HPV types induce cervical cancer and encode an E7 oncoprotein that plays a major role in HPV-induced carcinogenesis. However, the mechanism by which E7 induces carcinogenesis is not fully understood; specific anti-HPV agents are not available. In this study, we performed RNA-seq to characterize transcriptional profiling of keratinocytes expressing HPV-16 E7 and identified more than 200 genes that were differentially expressed between E7 and vector control cells. Through bioinformatics analysis, pathways altered in E7-expressing cells were identified. Significantly, the WDHD1 gene, one of the genes that is upregulated in E7-expressing cells, was found to play an important role in E7-induced G1 checkpoint abrogation and rereplication. These studies shed light on mechanisms by which HPV induces genomic instability and have therapeutic implications.
Collapse
|
31
|
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; 7:26844-55. [PMID: 27057626 PMCID: PMC5042019 DOI: 10.18632/oncotarget.8565] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
32
|
Zhang Y, Liu J, Wang S, Luo X, Li Y, Lv Z, Zhu J, Lin J, Ding L, Ye Q. The DEK oncogene activates VEGF expression and promotes tumor angiogenesis and growth in HIF-1α-dependent and -independent manners. Oncotarget 2016; 7:23740-56. [PMID: 26988756 PMCID: PMC5029660 DOI: 10.18632/oncotarget.8060] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/29/2016] [Indexed: 11/25/2022] Open
Abstract
The DEK oncogene is overexpressed in various cancers and overexpression of DEK correlates with poor clinical outcome. Vascular endothelial growth factor (VEGF) is the most important regulator of tumor angiogenesis, a process essential for tumor growth and metastasis. However, whether DEK enhances tumor angiogenesis remains unclear. Here, we show that DEK is a key regulator of VEGF expression and tumor angiogenesis. Using chromatin immunoprecipitation assay, we found that DEK promoted VEGF transcription in breast cancer cells (MCF7, ZR75-1 and MDA-MB-231) by directly binding to putative DEK-responsive element (DRE) of the VEGF promoter and indirectly binding to hypoxia response element (HRE) upstream of the DRE through its interaction with the transcription factor hypoxia-inducible factor 1α (HIF-1α), a master regulator of tumor angiogenesis and growth. DEK is responsible for recruitment of HIF-1α and the histone acetyltransferase p300 to the VEGF promoter. DEK-enhanced VEGF increases vascular endothelial cell proliferation, migration and tube formation as well as angiogenesis in the chick chorioallantoic membrane. DEK promotes tumor angiogenesis and growth in nude mice in HIF-1α-dependent and -independent manners. Immunohistochemical staining showed that DEK expression positively correlates with the expression of VEGF and microvessel number in 58 breast cancer patients. Our data establish DEK as a sequence-specific binding transcription factor, a novel coactivator for HIF-1α in regulation of VEGF transcription and a novel promoter of angiogenesis.
Collapse
MESH Headings
- Animals
- Apoptosis
- Biomarkers, Tumor
- Breast Neoplasms/blood supply
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Proliferation
- Chick Embryo
- Chorioallantoic Membrane/metabolism
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Mice
- Mice, Nude
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Poly-ADP-Ribose Binding Proteins/genetics
- Poly-ADP-Ribose Binding Proteins/metabolism
- Response Elements
- Signal Transduction
- Tumor Cells, Cultured
- Vascular Endothelial Growth Factor A/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yanan Zhang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Liaoning, People's Republic of China
| | - Jie Liu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Shibin Wang
- First Affiliated Hospital, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xiaoli Luo
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Yang Li
- First Affiliated Hospital, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhaohui Lv
- Department of Endocrinology, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, People's Republic of China
| | - Jie Zhu
- Department of Endocrinology, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, People's Republic of China
| | - Jing Lin
- First Affiliated Hospital, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lihua Ding
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, People's Republic of China
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Liaoning, People's Republic of China
| |
Collapse
|
33
|
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: 3.2] [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
|
34
|
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: 27] [Impact Index Per Article: 3.0] [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
|
35
|
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: 3.1] [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
|
36
|
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: 2.0] [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
|
37
|
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: 5.8] [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
|
38
|
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: 37] [Impact Index Per Article: 4.1] [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
|
39
|
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; 11:1318-23. [PMID: 25351213 DOI: 10.3892/mmr.2014.2781] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [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
|
40
|
Logan GE, Mor-Vaknin N, Braunschweig T, Jost E, Schmidt PV, Markovitz DM, Mills KI, Kappes F, Percy MJ. DEK oncogene expression during normal hematopoiesis and in Acute Myeloid Leukemia (AML). Blood Cells Mol Dis 2015; 54:123-31. [PMID: 25128083 DOI: 10.1016/j.bcmd.2014.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 12/24/2022]
Abstract
DEK is important in regulating cellular processes including proliferation, differentiation and maintenance of stem cell phenotype. The translocation t(6;9) in Acute Myeloid Leukemia (AML), which fuses DEK with NUP214, confers a poor prognosis and a higher risk of relapse. The over-expression of DEK in AML has been reported, but different studies have shown diminished levels in pediatric and promyelocytic leukemias. This study has characterized DEK expression, in silico, using a large multi-center cohort of leukemic and normal control cases. Overall, DEK was under-expressed in AML compared to normal bone marrow (NBM). Studying specific subtypes of AML confirmed either no significant change or a significant reduction in DEK expression compared to NBM. Importantly, the similarity of DEK expression between AML and NBM was confirmed using immunohistochemistry analysis of tissue mircorarrays. In addition, stratification of AML patients based on median DEK expression levels indicated that DEK showed no effect on the overall survival of patients. DEK expression during normal hematopoiesis did reveal a relationship with specific cell types implicating a distinct function during myeloid differentiation. Whilst DEK may play a potential role in hematopoiesis, it remains to be established whether it is important for leukemagenesis, except when involved in the t(6;9) translocation.
Collapse
MESH Headings
- Animals
- Chromosomal Proteins, Non-Histone/biosynthesis
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomes, Human, Pair 6/genetics
- Chromosomes, Human, Pair 9/genetics
- Cohort Studies
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Databases, Genetic
- Disease-Free Survival
- Gene Expression Regulation, Leukemic
- Hematopoiesis
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Multicenter Studies as Topic
- Oncogene Proteins/biosynthesis
- Oncogene Proteins/genetics
- Poly-ADP-Ribose Binding Proteins
- Survival Rate
- Translocation, Genetic
Collapse
Affiliation(s)
- Gemma E Logan
- Centre for Cancer Research and Cell Biology (CCRCB), Queen's University Belfast, Belfast, United Kingdom.
| | - Nirit Mor-Vaknin
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA.
| | - Till Braunschweig
- Institute of Pathology, Medical School, RWTH Aachen University, Aachen, Germany.
| | - Edgar Jost
- Clinic for Oncology, Hematology and Stem Cell Transplantation, Medical School, RWTH Aachen University, Aachen, Germany.
| | - Pia Verena Schmidt
- Clinic for Oncology, Hematology and Stem Cell Transplantation, Medical School, RWTH Aachen University, Aachen, Germany.
| | - David M Markovitz
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA.
| | - Ken I Mills
- Centre for Cancer Research and Cell Biology (CCRCB), Queen's University Belfast, Belfast, United Kingdom.
| | - Ferdinand Kappes
- Institute of Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen, Germany.
| | - Melanie J Percy
- Centre for Cancer Research and Cell Biology (CCRCB), Queen's University Belfast, Belfast, United Kingdom; Haematology Department, Belfast City Hospital, Belfast Health and Social Care Trust, Belfast, United Kingdom.
| |
Collapse
|
41
|
Martinez-Useros J, Rodriguez-Remirez M, Borrero-Palacios A, Moreno I, Cebrian A, Gomez del Pulgar T, del Puerto-Nevado L, Vega-Bravo R, Puime-Otin A, Perez N, Zazo S, Senin C, Fernandez-Aceñero MJ, Soengas MS, Rojo F, Garcia-Foncillas J. DEK is a potential marker for aggressive phenotype and irinotecan-based therapy response in metastatic colorectal cancer. BMC Cancer 2014; 14:965. [PMID: 25515240 PMCID: PMC4300837 DOI: 10.1186/1471-2407-14-965] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 12/11/2014] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND DEK is a transcription factor involved in stabilization of heterochromatin and cruciform structures. It plays an important role in development and progression of different types of cancer. This study aims to analyze the role of DEK in metastatic colorectal cancer. METHODS Baseline DEK expression was firstly quantified in 9 colorectal cell lines and normal mucosa by WB. SiRNA-mediated DEK inhibition was carried out for transient DEK silencing in DLD1 and SW620 to dissect its role in colorectal cancer aggressiveness. Irinotecan response assays were performed with SN38 over 24 hours and apoptosis was quantified by flow cytometry. Ex-vivo assay was carried out with 3 fresh tumour tissues taken from surgical resection and treated with SN38 for 24 hours. DEK expression was determined by immunohistochemistry in 67 formalin-fixed paraffin-embedded tumour samples from metastatic colorectal cancer patients treated with irinotecan-based therapy as first-line treatment. RESULTS The DEK oncogene is overexpressed in all colorectal cancer cell lines. Knock-down of DEK on DLD1 and SW620 cell lines decreased cell migration and increased irinotecan-induced apoptosis. In addition, low DEK expression level predicted irinotecan-based chemotherapy response in metastatic colorectal cancer patients with KRAS wild-type. CONCLUSIONS These data suggest DEK overexpression as a crucial event for the emergence of an aggressive phenotype in colorectal cancer and its potential role as biomarker for irinotecan response in those patients with KRAS wild-type status.
Collapse
Affiliation(s)
- Javier Martinez-Useros
- />Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - Maria Rodriguez-Remirez
- />Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - Aurea Borrero-Palacios
- />Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - Irene Moreno
- />Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - Arancha Cebrian
- />Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - Teresa Gomez del Pulgar
- />Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - Laura del Puerto-Nevado
- />Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| | - Ricardo Vega-Bravo
- />Department of Pathology, University Hospital “Fundación Jiménez Díaz”-UAM, Madrid, Spain
| | - Alberto Puime-Otin
- />Department of Pathology, University Hospital “Fundación Jiménez Díaz”-UAM, Madrid, Spain
| | - Nuria Perez
- />Department of Pathology, University Hospital “Fundación Jiménez Díaz”-UAM, Madrid, Spain
| | - Sandra Zazo
- />Department of Pathology, University Hospital “Fundación Jiménez Díaz”-UAM, Madrid, Spain
| | - Clara Senin
- />Department of Oncology, Vigo Hospital, Vigo, Spain
| | | | - Maria S Soengas
- />Melanoma Research Group, Spanish National Cancer Research Centre, Madrid, Spain
| | - Federico Rojo
- />Department of Pathology, University Hospital “Fundación Jiménez Díaz”-UAM, Madrid, Spain
| | - Jesus Garcia-Foncillas
- />Translational Oncology Division, OncoHealth Institute, Health Research Institute - University Hospital “Fundación Jiménez Díaz”-UAM, Av. Reyes Católicos 2, 28040 Madrid, Spain
| |
Collapse
|
42
|
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: 1.9] [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
|
43
|
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: 2.6] [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
|
44
|
Organista-Nava J, Gómez-Gómez Y, Gariglio P. Embryonic stem cell-specific signature in cervical cancer. Tumour Biol 2013; 35:1727-38. [PMID: 24163107 DOI: 10.1007/s13277-013-1321-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 10/14/2013] [Indexed: 10/26/2022] Open
Abstract
The wide range of invasive and noninvasive lesion phenotypes associated with high-risk human papillomavirus (HR-HPV) infection in cervical cancer (CC) indicates that not only the virus but also specific cervical epithelial cells in the transformation zone (TZ), such as stem cells (SCs), play an important part in the development of cervical neoplasia. In this review, we focused in an expression signature that is specific to embryonic SCs and to poorly differentiated cervical malignant tumors and we hypothesize that this expression signature may play an important role to promote cell growth, survival, colony formation, lack of adhesion, as well as cell invasion and migration in CC.
Collapse
Affiliation(s)
- Jorge Organista-Nava
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México (UNAM), México, DF, México,
| | | | | |
Collapse
|
45
|
Kaewprag J, Umnajvijit W, Ngamkham J, Ponglikitmongkol M. HPV16 oncoproteins promote cervical cancer invasiveness by upregulating specific matrix metalloproteinases. PLoS One 2013; 8:e71611. [PMID: 23967226 PMCID: PMC3742460 DOI: 10.1371/journal.pone.0071611] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 07/01/2013] [Indexed: 02/07/2023] Open
Abstract
Production of matrix metalloproteinases (MMPs) for degradation of extracellular matrix is a vital step in cancer metastasis. We investigated the effects of HPV16 oncoproteins (16E6, 16E6*I and 16E7), either individually or combined, on the transcription of 7 MMPs implicated in cervical cancer invasiveness. The levels of 7 MMPs reported to be increased in cervical cancer were determined in C33A stably expressing different HPV16 oncoproteins using quantitative RT-PCR and compared with invasion ability of cell lines using in vitro invasion and wound healing assays. Overexpression of MMP-2 and MT1-MMP was detected in HPV16E6E7 expressing cells which correlated with increased cell invasion. Combination of HPV oncoproteins always showed greater effects than its individual form. Inhibition of cell invasion using a specific MMP-2 inhibitor, OA-Hy, and anti-MT1-MMP antibody confirmed that invasion in these cells was dependent on both MMP-2 and MT1-MMP expression. Depletion of HPV16E6E7 by shRNA-mediated knock-down experiments resulted in decreased MMP-2 and MT1-MMP expression levels as well as reduced invasion ability which strongly suggested specific effects of HPV oncoproteins on both MMPs and on cell invasion. Immunohistochemistry study in invasive cervical cancers confirmed the enhanced in vivo expression of these two MMPs in HPV16-infected cells. In addition, possible sites required by HPV16E6E7 on the MMP-2 and MT1-MMP promoters were investigated and PEA3 (at -552/-540 for MMP-2, -303 for MT1-MMP) and Sp1 (at -91 for MMP-2, -102 for MT1-MMP) binding sites were shown to be essential for mediating their transactivation activity. In conclusion, our study demonstrated that HPV16E6 and E7 oncoproteins cooperate in promoting cervical cancer invasiveness by specifically upregulating MMP-2 and MT1-MMP transcription in a similar manner.
Collapse
Affiliation(s)
- Jittranan Kaewprag
- Molecular Medicine Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Wareerat Umnajvijit
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | | |
Collapse
|
46
|
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: 2.3] [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
|
47
|
The rate of spontaneous mutations in human myeloid cells. Mutat Res 2013; 749:49-57. [PMID: 23748046 DOI: 10.1016/j.mrfmmm.2013.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 05/28/2013] [Accepted: 05/29/2013] [Indexed: 01/09/2023]
Abstract
The mutation rate (μ) is likely to be a key parameter in leukemogenesis, but historically, it has been difficult to measure in humans. The PIG-A gene has some advantages for the detection of spontaneous mutations because it is X-linked, and therefore only one mutation is required to disrupt its function. Furthermore, the PIG-A-null phenotype is readily detected by flow cytometry. Using PIG-A, we have now provided the first in vitro measurement of μ in myeloid cells, using cultures of CD34+ cells that are transduced with either the AML-ETO or the MLL-AF9 fusion genes and expanded with cytokines. For the AML-ETO cultures, the median μ value was ∼9.4×10(-7) (range ∼3.6-23×10(-7)) per cell division. In contrast, few spontaneous mutations were observed in the MLL-AF9 cultures. Knockdown of p53 or introduction of mutant NRAS or FLT3 alleles did not have much of an effect on μ. Based on these data, we provide a model to predict whether hypermutability must occur in the process of leukemogenesis.
Collapse
|
48
|
Abstract
E7 is an accessory protein that is not encoded by all papillomaviruses. The E7 amino terminus contains two regions of similarity to conserved regions 1 and 2 of the adenovirus E1A protein, which are also conserved in the simian vacuolating virus 40 large tumor antigen. The E7 carboxyl terminus consists of a zinc-binding motif, which is related to similar motifs in E6 proteins. E7 proteins play a central role in the human papillomavirus life cycle, reprogramming the cellular environment to be conducive to viral replication. E7 proteins encoded by the cancer-associated alpha human papillomaviruses have potent transforming activities, which together with E6, are necessary but not sufficient to render their host squamous epithelial cell tumorigenic. This article strives to provide a comprehensive summary of the published research studies on human papillomavirus E7 proteins.
Collapse
|
49
|
Saha AK, Kappes F, Mundade A, Deutzmann A, Rosmarin DM, Legendre M, Chatain N, Al-Obaidi Z, Adams BS, Ploegh HL, Ferrando-May E, Mor-Vaknin N, Markovitz DM. Intercellular trafficking of the nuclear oncoprotein DEK. Proc Natl Acad Sci U S A 2013; 110:6847-52. [PMID: 23569252 PMCID: PMC3637753 DOI: 10.1073/pnas.1220751110] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
DEK is a biochemically distinct, conserved nonhistone protein that is vital to global heterochromatin integrity. In addition, DEK can be secreted and function as a chemotactic, proinflammatory factor. Here we show that exogenous DEK can penetrate cells, translocate to the nucleus, and there carry out its endogenous nuclear functions. Strikingly, adjacent cells can take up DEK secreted from synovial macrophages. DEK internalization is a heparan sulfate-dependent process, and cellular uptake of DEK into DEK knockdown cells corrects global heterochromatin depletion and DNA repair deficits, the phenotypic aberrations characteristic of these cells. These findings thus unify the extracellular and intracellular activities of DEK, and suggest that this paracrine loop involving DEK plays a role in chromatin biology.
Collapse
Affiliation(s)
- Anjan K. Saha
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Ferdinand Kappes
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen 52074, Germany
| | - Amruta Mundade
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Anja Deutzmann
- Department of Biology, University of Konstanz, Konstanz 78457, Germany
| | - David M. Rosmarin
- Whitehead Institute, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Maureen Legendre
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Nicolas Chatain
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Aachen 52074, Germany
| | - Zeina Al-Obaidi
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Barbara S. Adams
- Department of Pediatrics, Division of Rheumatology, University of Michigan, Ann Arbor, MI 48109; and
| | - Hidde L. Ploegh
- Whitehead Institute, Massachusetts Institute of Technology, Cambridge, MA 02142
| | | | - Nirit Mor-Vaknin
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI 48109
| | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI 48109
- Programs in Immunology, Cellular and Molecular Biology, and Cancer Biology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
50
|
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: 4.5] [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
|