1
|
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: 0.3] [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
|
2
|
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: 0.3] [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
|
3
|
Liu B, Sun Y, Zhang Y, Xing Y, Suo J. DEK modulates both expression and alternative splicing of cancer‑related genes. Oncol Rep 2022; 47:111. [PMID: 35475534 PMCID: PMC9073418 DOI: 10.3892/or.2022.8322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/11/2022] [Indexed: 11/05/2022] Open
Abstract
DEK is known to be a potential proto‑oncogene and is highly expressed in gastric cancer (GC); thus, DEK is considered to contribute to the malignant progression of GC. DEK is an RNA‑binding protein involved in transcription, DNA repair, and selection of splicing sites during mRNA processing; however, its precise function remains elusive due to the lack of clarification of the overall profiles of gene transcription and post‑transcriptional splicing that are regulated by DEK. We performed our original whole‑genomic RNA‑Seq data to analyze the global transcription and alternative splicing profiles in a human GC cell line by comparing DEK siRNA‑treated and control conditions, dissecting both differential gene expression and potential alternative splicing events regulated by DEK. The siRNA‑mediated knockdown of DEK in a GC cell line led to significant changes in gene expression of multiple cancer‑related genes including both oncogenes and tumor suppressors. Moreover, it was revealed that DEK regulated a number of alternative splicing in genes which were significantly enriched in various cancer‑related pathways including apoptosis and cell cycle processes. This study clarified for the first time that DEK has a regulatory effect on the alternative splicing, as well as on the expression, of numerous cancer‑related genes, which is consistent with the role of DEK as a possible oncogene. Our results further expand the importance and feasibility of DEK as a clinical therapeutic target for human malignancies including GC.
Collapse
Affiliation(s)
- Bin Liu
- Department of Gastrocolorectal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuanlin Sun
- Department of Gastrocolorectal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yang Zhang
- Department of Gastrocolorectal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yanpeng Xing
- Department of Gastrocolorectal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jian Suo
- Department of Gastrocolorectal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
4
|
Chen Z, Huo D, Li L, Liu Z, Li Z, Xu S, Huang Y, Wu W, Zhou C, Liu Y, Kuang M, Wu F, Li H, Qian P, Song G, Wu X, Chen J, Hou Y. Nuclear DEK preserves hematopoietic stem cells potential via NCoR1/HDAC3-Akt1/2-mTOR axis. J Exp Med 2021; 218:e20201974. [PMID: 33755722 PMCID: PMC7992411 DOI: 10.1084/jem.20201974] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/16/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022] Open
Abstract
The oncogene DEK is found fused with the NUP214 gene creating oncoprotein DEK-NUP214 that induces acute myeloid leukemia (AML) in patients, and secreted DEK protein functions as a hematopoietic cytokine to regulate hematopoiesis; however, the intrinsic role of nuclear DEK in hematopoietic stem cells (HSCs) remains largely unknown. Here, we show that HSCs lacking DEK display defects in long-term self-renew capacity, eventually resulting in impaired hematopoiesis. DEK deficiency reduces quiescence and accelerates mitochondrial metabolism in HSCs, in part, dependent upon activating mTOR signaling. At the molecular level, DEK recruits the corepressor NCoR1 to repress acetylation of histone 3 at lysine 27 (H3K27ac) and restricts the chromatin accessibility of HSCs, governing the expression of quiescence-associated genes (e.g., Akt1/2, Ccnb2, and p21). Inhibition of mTOR activity largely restores the maintenance and potential of Dek-cKO HSCs. These findings highlight the crucial role of nuclear DEK in preserving HSC potential, uncovering a new link between chromatin remodelers and HSC homeostasis, and have clinical implications.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dawei Huo
- Department of Cell Biology, Tianjin Medical University, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin, China
| | - Lei Li
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhilong Liu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhigang Li
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shuangnian Xu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongxiu Huang
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Weiru Wu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chengfang Zhou
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuanyuan Liu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Mei Kuang
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Feng Wu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Li
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pengxu Qian
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Xudong Wu
- Department of Cell Biology, Tianjin Medical University, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin, China
| | - Jieping Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu Hou
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
5
|
Guo H, Prell M, Königs H, Xu N, Waldmann T, Hermans-Sachweh B, Ferrando-May E, Lüscher B, Kappes F. Bacterial Growth Inhibition Screen (BGIS) identifies a loss-of-function mutant of the DEK oncogene, indicating DNA modulating activities of DEK in chromatin. FEBS Lett 2021; 595:1438-1453. [PMID: 33686684 DOI: 10.1002/1873-3468.14070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
Abstract
The DEK oncoprotein regulates cellular chromatin function via a number of protein-protein interactions. However, the biological relevance of its unique pseudo-SAP/SAP-box domain, which transmits DNA modulating activities in vitro, remains largely speculative. As hypothesis-driven mutations failed to yield DNA-binding null (DBN) mutants, we combined random mutagenesis with the Bacterial Growth Inhibition Screen (BGIS) to overcome this bottleneck. Re-expression of a DEK-DBN mutant in newly established human DEK knockout cells failed to reduce the increase in nuclear size as compared to wild type, indicating roles for DEK-DNA interactions in cellular chromatin organization. Our results extend the functional roles of DEK in metazoan chromatin and highlight the predictive ability of recombinant protein toxicity in E. coli for unbiased studies of eukaryotic DNA modulating protein domains.
Collapse
Affiliation(s)
- Haihong Guo
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
| | - Malte Prell
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
| | - Hiltrud Königs
- Institute of Pathology, Medical School, RWTH Aachen University, Germany
| | - Nengwei Xu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Dushu Lake Higher Education Town, Suzhou Industrial Park, China
| | - Tanja Waldmann
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Germany
| | | | - Elisa Ferrando-May
- Bioimaging Center, Department of Biology, University of Konstanz, Germany
| | - Bernhard Lüscher
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
| | - Ferdinand Kappes
- Institute for Biochemistry and Molecular Biology, Medical School, RWTH Aachen University, Germany
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Dushu Lake Higher Education Town, Suzhou Industrial Park, China
| |
Collapse
|
6
|
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: 1.0] [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
|
7
|
Saikia S, Bordoloi M, Sarmah R, Kolita B. Antiviral compound screening, peptide designing, and protein network construction of influenza a virus (strain a/Puerto Rico/8/1934 H1N1). Drug Dev Res 2018; 80:106-124. [PMID: 30276835 DOI: 10.1002/ddr.21475] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 11/11/2022]
Abstract
Plant-based antiviral therapy is the current need for holistic health care management, which can be achieved through screening of phytochemicals and designing of antiviral peptides. There exist certain host's factors which are directly involved for rapid viral replication causing worldwide pandemic. A total of 177 phytochemicals from Ocimum sanctum (L.), Tinospora cordifolia (Thunb.) Miers, Cinnamomum camphora (L.) J. Presl., Allium sativum (L.), Curcuma longa (L.), and Aloe vera (L.) Burm. f. were evaluated for their affinity to all viral proteins of H1N1. Applying drug filters and keeping the threshold of such filters relative to the standards, 82 compounds were found suitable for further analysis. Consensus scoring system was used for screening top ligands from 82 compounds, which screened the top 12 compounds. Highly conserved regions (>80%) which were hydrophilic, flexible, antigenic, and also charged were screened out as potent antiviral peptides. The viral proteins were taken as the targets for the modeled peptides for protein-protein docking. Further, host-pathogen interacting network was constructed to unveil host factors involved in viral replication, from which unique protein clusters representing their involvement in viral reproduction were selected through mapping with pathway databases. Twelve compounds and five peptides were found to be highly effective against all the proteins of H1N1. Based on the uniqueness, 13 clusters of proteins were obtained which are engaged in cellular process, namely, viral reproduction, fructose-6-phosphate metabolism, nitrogen compound metabolism, biosynthesis, cellular process, oligodendrocyte development, localization, multiorganism process, primary metabolism, response to unfolded protein, metabolism, and response to protein and catabolism.
Collapse
Affiliation(s)
- Surovi Saikia
- Chemical Sciences & Technology (Natural Products Chemistry) Division, CSIR North East Institute of Science & Technology, Jorhat, Assam, India
| | - Manobjyoti Bordoloi
- Chemical Sciences & Technology (Natural Products Chemistry) Division, CSIR North East Institute of Science & Technology, Jorhat, Assam, India
| | - Rajeev Sarmah
- Allied Health Sciences, Assam Down Town University, Panikhaiti, Guwahati, Assam, India
| | - Bhaskor Kolita
- Chemical Sciences & Technology (Natural Products Chemistry) Division, CSIR North East Institute of Science & Technology, Jorhat, Assam, India
| |
Collapse
|
8
|
Mor‐Vaknin N, Rivas M, Legendre M, Mohan S, Yuanfan Y, Mau T, Johnson A, Huang B, Zhao L, Kimura Y, Spalding SJ, Morris PW, Gottlieb BS, Onel K, Olson JC, Edelheit BS, Shishov M, Jung LK, Cassidy EA, Prahalad S, Passo MH, Beukelman T, Mehta J, Giannini EH, Adams BS, Lovell DJ, Markovitz DM. High Levels of DEK Autoantibodies in Sera of Patients With Polyarticular Juvenile Idiopathic Arthritis and With Early Disease Flares Following Cessation of Anti-Tumor Necrosis Factor Therapy. Arthritis Rheumatol 2018; 70:594-605. [PMID: 29287303 PMCID: PMC5876119 DOI: 10.1002/art.40404] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 12/18/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The nuclear oncoprotein DEK is an autoantigen associated with juvenile idiopathic arthritis (JIA), especially the oligoarticular subtype. DEK is a secreted chemotactic factor. Abundant levels of DEK and DEK autoantibodies are found in inflamed synovium in JIA. We undertook this study to further characterize the nature of DEK autoantibodies in screening serum samples from 2 different cohorts that consisted mostly of patients with JIA. METHODS DEK autoantibody levels were analyzed in sera from 33 JIA patients, 13 patients with other inflammatory conditions, and 11 healthy controls, as well as in 89 serum samples from JIA patients receiving anti-tumor necrosis factor (anti-TNF) therapy. Recombinant His-tagged full-length DEK protein (1-375 amino acids [aa]) and the 187-375-aa and 1-350-aa His-tagged DEK fragments made in a baculovirus system were used for enzyme-linked immunosorbent assay (ELISA) and immunoblotting. The C-terminal 25-aa fragment of DEK was expressed in a glutathione S-transferase-tagged vector. ELISA results were calculated as area under the curve by the trapezoidal rule. RESULTS DEK autoantibody levels were significantly higher in patients with polyarticular JIA than in those with oligoarticular JIA, and were higher in patients with polyarticular JIA who had more active disease after cessation of anti-TNF therapy. Immunoblotting against the C-terminal 25-aa fragment of DEK confirmed that this section of the DEK molecule is the most immunogenic domain. CONCLUSION DEK autoantibody levels are higher in patients with polyarticular JIA than in those with oligoarticular JIA, and higher in patients who have disease flares after cessation of anti-TNF therapy. The C-terminal 25-aa fragment is the most immunogenic portion of DEK. These findings are significant with respect to the nature of DEK autoantibodies, their contribution to JIA pathogenesis, and their implications for JIA management.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Anne Johnson
- Cincinnati Children's Hospital Medical CenterCincinnatiOhio
| | - Bin Huang
- Cincinnati Children's Hospital Medical Center and University of Cincinnati School of MedicineCincinnatiOhio
| | | | - Yukiko Kimura
- Joseph M. Sanzari Children's HospitalHackensack University Medical CenterHackensackNew Jersey
| | | | | | - Beth S. Gottlieb
- Cohen Children's Medical Center, Northwell HealthHofstra Norwell School of MedicineHempsteadNew York
| | - Karen Onel
- Joseph M. Sanzari Children's HospitalHackensack University Medical CenterHackensackNew Jersey
| | | | | | | | | | | | | | | | | | - Jay Mehta
- Children's Hospital at Montefiore/Albert Einstein College of MedicineBronxNew York
| | | | | | | | | |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem/progenitor cell fate decision during hematopoiesis is regulated by intracellular and extracellular signals such as transcription factors, growth factors, and cell-to-cell interactions. In this review, we explore the function of DEK, a nuclear phosphoprotein, on gene regulation. We also examine how DEK is secreted and internalized by cells, and discuss how both endogenous and extracellular DEK regulates hematopoiesis. Finally, we explore what currently is known about the regulation of DEK during inflammation. RECENT FINDINGS DEK negatively regulates the proliferation of early myeloid progenitor cells but has a positive effect on the differentiation of mature myeloid cells. Inflammation regulates intracellular DEK concentrations with inflammatory stimuli enhancing DEK expression. Inflammation-induced nuclear factor-kappa B activation is regulated by DEK, resulting in changes in the production of other inflammatory molecules such as IL-8. Inflammatory stimuli in turn regulates DEK secretion by cells of hematopoietic origin. However, how inflammation-induced expression and secretion of DEK regulates hematopoiesis remains unknown. SUMMARY Understanding how DEK regulates hematopoiesis under both homeostatic and inflammatory conditions may lead to a better understanding of the biology of HSCs and HPCs. Furthering our knowledge of the regulation of hematopoiesis will ultimately lead to new therapeutics that may increase the efficacy of hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Maegan L Capitano
- Indiana University School of Medicine, Department of Microbiology and Immunology, Indianapolis, Indiana, USA
| | | |
Collapse
|
10
|
Port SA, Mendes A, Valkova C, Spillner C, Fahrenkrog B, Kaether C, Kehlenbach RH. The Oncogenic Fusion Proteins SET-Nup214 and Sequestosome-1 (SQSTM1)-Nup214 Form Dynamic Nuclear Bodies and Differentially Affect Nuclear Protein and Poly(A)+ RNA Export. J Biol Chem 2016; 291:23068-23083. [PMID: 27613868 PMCID: PMC5087727 DOI: 10.1074/jbc.m116.735340] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/31/2016] [Indexed: 01/09/2023] Open
Abstract
Genetic rearrangements are a hallmark of several forms of leukemia and can lead to oncogenic fusion proteins. One example of an affected chromosomal region is the gene coding for Nup214, a nucleoporin that localizes to the cytoplasmic side of the nuclear pore complex (NPC). We investigated two such fusion proteins, SET-Nup214 and SQSTM1 (sequestosome)-Nup214, both containing C-terminal portions of Nup214. SET-Nup214 nuclear bodies containing the nuclear export receptor CRM1 were observed in the leukemia cell lines LOUCY and MEGAL. Overexpression of SET-Nup214 in HeLa cells leads to the formation of similar nuclear bodies that recruit CRM1, export cargo proteins, and certain nucleoporins and concomitantly affect nuclear protein and poly(A)+ RNA export. SQSTM1-Nup214, although mostly cytoplasmic, also forms nuclear bodies and inhibits nuclear protein but not poly(A)+ RNA export. The interaction of the fusion proteins with CRM1 is RanGTP-dependent, as shown in co-immunoprecipitation experiments and binding assays. Further analysis revealed that the Nup214 parts mediate the inhibition of nuclear export, whereas the SET or SQSTM1 part determines the localization of the fusion protein and therefore the extent of the effect. SET-Nup214 nuclear bodies are highly mobile structures, which are in equilibrium with the nucleoplasm in interphase and disassemble during mitosis or upon treatment of cells with the CRM1-inhibitor leptomycin B. Strikingly, we found that nucleoporins can be released from nuclear bodies and reintegrated into existing NPC. Our results point to nuclear bodies as a means of preventing the formation of potentially insoluble and harmful protein aggregates that also may serve as storage compartments for nuclear transport factors.
Collapse
Affiliation(s)
- Sarah A Port
- From the Department of Molecular Biology, Faculty of Medicine and the Göttingen Center for Molecular Biosciences (GZMB), Georg August University, Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Adélia Mendes
- the Institute of Molecular Biology and Medicine, Université Libre de Bruxelles, 6041 Charleroi, Belgium, and
| | - Christina Valkova
- the Leibniz Institute on Aging,Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Christiane Spillner
- From the Department of Molecular Biology, Faculty of Medicine and the Göttingen Center for Molecular Biosciences (GZMB), Georg August University, Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Birthe Fahrenkrog
- the Institute of Molecular Biology and Medicine, Université Libre de Bruxelles, 6041 Charleroi, Belgium, and
| | - Christoph Kaether
- the Leibniz Institute on Aging,Fritz Lipmann Institute, Beutenbergstrasse 11, 07745 Jena, Germany
| | - Ralph H Kehlenbach
- From the Department of Molecular Biology, Faculty of Medicine and the Göttingen Center for Molecular Biosciences (GZMB), Georg August University, Göttingen, Humboldtallee 23, 37073 Göttingen, Germany,
| |
Collapse
|
11
|
Qiao MX, Li C, Zhang AQ, Hou LL, Yang J, Hu HG. Regulation of DEK expression by AP-2α and methylation level of DEK promoter in hepatocellular carcinoma. Oncol Rep 2016; 36:2382-90. [PMID: 27499261 DOI: 10.3892/or.2016.4984] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 06/23/2016] [Indexed: 11/06/2022] Open
Abstract
DEK is overexpressed in multiple invasive tumors. However, the transcriptional regulatory mechanism of DEK remains unclear. In the present study, progressive-type truncation assay indicated that CpG2-2 (-167 bp/+35 bp) was the DEK core promoter, whose methylation inhibited DEK expression. Bisulfite genomic sequencing analysis indicated that the methylation levels of the DEK promoter in normal hepatic cells and tissues were higher than those in hepatocellular carcinoma (HCC) cells. TFSEARCH result revealed transcription factor binding sites in CpG2-2. Among the sites, the AP-2α binding site showed the most significant methylation difference; hence, AP-2α is a key transcription factor that regulates DEK expression. Point or deletion mutation of the AP-2α binding site significantly reduced the promoter activity. Chromatin immunoprecipitation assay demonstrated the binding of AP-2α to the core promoter. Furthermore, knock down of endogenous AP-2α downregulated DEK expression, whereas overexpression of AP-2α upregulated DEK expression. Thus, AP-2α is an important transcription factor of DEK expression, which is correlated with the methylation level of the DEK core promoter in HCC.
Collapse
Affiliation(s)
- Ming-Xu Qiao
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Chun Li
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Ai-Qun Zhang
- Institute of Hepatobiliary Surgery, PLA General Hospital, Beijing 100853, P.R. China
| | - Ling-Ling Hou
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing 100044, P.R. China
| | - Juan Yang
- 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
|
12
|
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: 2.7] [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
|
13
|
Lohmann F, Dangeti M, Soni S, Chen X, Planutis A, Baron MH, Choi K, Bieker JJ. The DEK Oncoprotein Is a Critical Component of the EKLF/KLF1 Enhancer in Erythroid Cells. Mol Cell Biol 2015; 35:3726-38. [PMID: 26303528 PMCID: PMC4589598 DOI: 10.1128/mcb.00382-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 05/06/2015] [Accepted: 08/17/2015] [Indexed: 02/07/2023] Open
Abstract
Understanding how transcriptional regulators are themselves controlled is important in attaining a complete picture of the intracellular effects that follow signaling cascades during early development and cell-restricted differentiation. We have addressed this issue by focusing on the regulation of EKLF/KLF1, a zinc finger transcription factor that plays a necessary role in the global regulation of erythroid gene expression. Using biochemical affinity purification, we have identified the DEK oncoprotein as a critical factor that interacts with an essential upstream enhancer element of the EKLF promoter and exerts a positive effect on EKLF levels. This element also binds a core set of erythroid transcription factors, suggesting that DEK is part of a tissue-restricted enhanceosome that contains BMP4-dependent and -independent components. Together with local enrichment of properly coded histones and an open chromatin domain, optimal transcriptional activation of the EKLF locus can be established.
Collapse
Affiliation(s)
- Felix Lohmann
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Mohan Dangeti
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Shefali Soni
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Xiaoyong Chen
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Antanas Planutis
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA
| | - Margaret H Baron
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA Department of Medicine, Mount Sinai School of Medicine, New York, New York, USA
| | - Kyunghee Choi
- Department of Pathology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - James J Bieker
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, New York, USA Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York, USA Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| |
Collapse
|
14
|
Dissecting the Potential Interplay of DEK Functions in Inflammation and Cancer. JOURNAL OF ONCOLOGY 2015; 2015:106517. [PMID: 26425120 PMCID: PMC4575739 DOI: 10.1155/2015/106517] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/05/2015] [Indexed: 12/12/2022]
Abstract
There is a long-standing correlation between inflammation, inflammatory cell signaling pathways, and tumor formation. Understanding the mechanisms behind inflammation-driven tumorigenesis is of great research and clinical importance. Although not entirely understood, these mechanisms include a complex interaction between the immune system and the damaged epithelium that is mediated by an array of molecular signals of inflammation—including reactive oxygen species (ROS), cytokines, and NFκB signaling—that are also oncogenic. Here, we discuss the association of the unique DEK protein with these processes. Specifically, we address the role of DEK in chronic inflammation via viral infections and autoimmune diseases, the overexpression and oncogenic activity of DEK in cancers, and DEK-mediated regulation of NFκB signaling. Combined, evidence suggests that DEK may play a complex, multidimensional role in chronic inflammation and subsequent tumorigenesis.
Collapse
|
15
|
Sandén C, Gullberg U. The DEK oncoprotein and its emerging roles in gene regulation. Leukemia 2015; 29:1632-6. [PMID: 25765544 DOI: 10.1038/leu.2015.72] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/08/2015] [Accepted: 03/03/2015] [Indexed: 02/06/2023]
Abstract
The DEK oncogene is highly expressed in cells from most human tissues and overexpressed in a large and growing number of cancers. It also fuses with the NUP214 gene to form the DEK-NUP214 fusion gene in a subset of acute myeloid leukemia. Originally characterized as a member of this translocation, DEK has since been implicated in epigenetic and transcriptional regulation, but its role in these processes is still elusive and intriguingly complex. Similarly multifaceted is its contribution to cellular transformation, affecting multiple cellular processes such as self-renewal, proliferation, differentiation, senescence and apoptosis. Recently, the roles of the DEK and DEK-NUP214 proteins have been elucidated by global analysis of DNA binding and gene expression, as well as multiple functional studies. This review outlines recent advances in the understanding of the basic functions of the DEK protein and its role in leukemogenesis.
Collapse
Affiliation(s)
- C Sandén
- Department of Hematology, Lund University, Lund, Sweden
| | - U Gullberg
- Department of Hematology, Lund University, Lund, Sweden
| |
Collapse
|
16
|
Uppal T, Banerjee S, Sun Z, Verma SC, Robertson ES. KSHV LANA--the master regulator of KSHV latency. Viruses 2014; 6:4961-98. [PMID: 25514370 PMCID: PMC4276939 DOI: 10.3390/v6124961] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 12/16/2022] Open
Abstract
Kaposi's sarcoma associated herpesvirus (KSHV), like other human herpes viruses, establishes a biphasic life cycle referred to as dormant or latent, and productive or lytic phases. The latent phase is characterized by the persistence of viral episomes in a highly ordered chromatin structure and with the expression of a limited number of viral genes. Latency Associated Nuclear Antigen (LANA) is among the most abundantly expressed proteins during latency and is required for various nuclear functions including the recruitment of cellular machineries for viral DNA replication and segregation of the replicated genomes to daughter cells. LANA achieves these functions by recruiting cellular proteins including replication factors, chromatin modifying enzymes and cellular mitotic apparatus assembly. LANA directly binds to the terminal repeat region of the viral genome and associates with nucleosomal proteins to tether to the host chromosome. Binding of LANA to TR recruits the replication machinery, thereby initiating DNA replication within the TR. However, other regions of the viral genome can also initiate replication as determined by Single Molecule Analysis of the Replicated DNA (SMARD) approach. Recent, next generation sequence analysis of the viral transcriptome shows the expression of additional genes during latent phase. Here, we discuss the newly annotated latent genes and the role of major latent proteins in KSHV biology.
Collapse
Affiliation(s)
- Timsy Uppal
- Department of Microbiology and Immunology, University of Nevada, Reno, School of Medicine, 1664 N Virginia Street, MS 320, Reno, NV 89557, USA.
| | - Sagarika Banerjee
- Department of Microbiology and the Tumor Virology Program of the Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 201E Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104, USA.
| | - Zhiguo Sun
- Department of Microbiology and the Tumor Virology Program of the Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 201E Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104, USA.
| | - Subhash C Verma
- Department of Microbiology and Immunology, University of Nevada, Reno, School of Medicine, 1664 N Virginia Street, MS 320, Reno, NV 89557, USA.
| | - Erle S Robertson
- Department of Microbiology and the Tumor Virology Program of the Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, 201E Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104, USA.
| |
Collapse
|
17
|
Waidmann S, Kusenda B, Mayerhofer J, Mechtler K, Jonak C. A DEK domain-containing protein modulates chromatin structure and function in Arabidopsis. THE PLANT CELL 2014; 26:4328-44. [PMID: 25387881 PMCID: PMC4277211 DOI: 10.1105/tpc.114.129254] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 10/01/2014] [Accepted: 10/22/2014] [Indexed: 05/19/2023]
Abstract
Chromatin is a major determinant in the regulation of virtually all DNA-dependent processes. Chromatin architectural proteins interact with nucleosomes to modulate chromatin accessibility and higher-order chromatin structure. The evolutionarily conserved DEK domain-containing protein is implicated in important chromatin-related processes in animals, but little is known about its DNA targets and protein interaction partners. In plants, the role of DEK has remained elusive. In this work, we identified DEK3 as a chromatin-associated protein in Arabidopsis thaliana. DEK3 specifically binds histones H3 and H4. Purification of other proteins associated with nuclear DEK3 also established DNA topoisomerase 1α and proteins of the cohesion complex as in vivo interaction partners. Genome-wide mapping of DEK3 binding sites by chromatin immunoprecipitation followed by deep sequencing revealed enrichment of DEK3 at protein-coding genes throughout the genome. Using DEK3 knockout and overexpressor lines, we show that DEK3 affects nucleosome occupancy and chromatin accessibility and modulates the expression of DEK3 target genes. Furthermore, functional levels of DEK3 are crucial for stress tolerance. Overall, data indicate that DEK3 contributes to modulation of Arabidopsis chromatin structure and function.
Collapse
Affiliation(s)
- Sascha Waidmann
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, Vienna Biocenter, 1030 Vienna, Austria
| | - Branislav Kusenda
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, Vienna Biocenter, 1030 Vienna, Austria
| | - Juliane Mayerhofer
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, Vienna Biocenter, 1030 Vienna, Austria
| | - Karl Mechtler
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Claudia Jonak
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, Vienna Biocenter, 1030 Vienna, Austria
| |
Collapse
|
18
|
Sandén C, Järvstråt L, Lennartsson A, Brattås PL, Nilsson B, Gullberg U. The DEK oncoprotein binds to highly and ubiquitously expressed genes with a dual role in their transcriptional regulation. Mol Cancer 2014; 13:215. [PMID: 25216995 PMCID: PMC4175287 DOI: 10.1186/1476-4598-13-215] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The DEK gene is highly expressed in a wide range of cancer cells, and a recurrent translocation partner in acute myeloid leukemia. While DEK has been identified as one of the most abundant proteins in human chromatin, its function and binding properties are not fully understood. METHODS We performed ChIP-seq analysis in the myeloid cell line U937 and coupled it with epigenetic and gene expression analysis to explore the genome-wide binding pattern of DEK and its role in gene regulation. RESULTS We show that DEK preferentially binds to open chromatin, with a low degree of DNA methylation and scarce in the heterochromatin marker H3K9me(3) but rich in the euchromatin marks H3K4me(2/3), H3K27ac and H3K9ac. More specifically, DEK binding is predominantly located at the transcription start sites of highly transcribed genes and a comparative analysis with previously established transcription factor binding patterns shows a similarity with that of RNA polymerase II. Further bioinformatic analysis demonstrates that DEK mainly binds to genes that are ubiquitously expressed across tissues. The functional significance of DEK binding was demonstrated by knockdown of DEK by shRNA, resulting in both significant upregulation and downregulation of DEK-bound genes. CONCLUSIONS We find that DEK binds to transcription start sites with a dual role in activation and repression of highly and ubiquitously expressed genes.
Collapse
Affiliation(s)
- Carl Sandén
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| | - Linnea Järvstråt
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| | - Andreas Lennartsson
- />Center for Biosciences, Department of Biosciences and Nutrition, Karolinska Institute, Novum, 141 83 Huddinge, Sweden
| | - Per Ludvik Brattås
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| | - Björn Nilsson
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| | - Urban Gullberg
- />Department of Hematology, Lund University, BMC B13, Klinikgatan 26, 221 84 Lund, Sweden
| |
Collapse
|
19
|
Shmaryahu A, Carrasco M, Valenzuela PD. Prediction of Bacterial microRNAs and possible targets in human cell transcriptome. J Microbiol 2014; 52:482-9. [DOI: 10.1007/s12275-014-3658-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 01/23/2014] [Accepted: 01/24/2014] [Indexed: 11/24/2022]
|
20
|
Piao J, Shang Y, Liu S, Piao Y, Cui X, Li Y, Lin Z. High expression of DEK predicts poor prognosis of gastric adenocarcinoma. Diagn Pathol 2014; 9:67. [PMID: 24650035 PMCID: PMC3994479 DOI: 10.1186/1746-1596-9-67] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/03/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND DEK, as an oncoprotein, plays an important role in cancer development and progression. This study aimed to investigate the clinicopathological significance of DEK overexpression in patients with gastric cancer. MATERIALS AND METHODS The expression of DEK protein was evaluated by immunohistochemical (IHC) staining of 172 gastric cancer samples with complete clinicopathological features, and the correlation between DEK expression and clinicopathological features was examined. Survival rates were also calculated using the Kaplan-Meier method in gastric cancer patients with complete survival data. RESULTS DEK protein showed a strictly nuclear staining pattern in gastric cancers with IHC and immunofluorescence. The strongly positive rate of DEK protein was 60.5% (104/172) in gastric cancers, which was significantly higher than that in either gastric dysplasia (19.4%, 7/36) or adjacent normal mucosa (0%, 0/27). DEK expression in gastric cancer correlated to tumor size, differentiation, clinical stage, disease-free survival, and overall survival rates. Further analysis showed that patients with early-stage gastric cancer and high DEK expression had shorter disease-free survival and overall survival duration than those with low DEK expression. CONCLUSION High level of DEK protein expression predicts the poor prognosis of patients with gastric cancer. DEK expression might be potentially used as an independent effective biomarker for prognostic evaluation of gastric cancers. VIRTUAL SLIDES The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/5050145571193097.
Collapse
Affiliation(s)
- Junjie Piao
- Department of Pathology, Yanbian University Medical College, No. 977, Gongyuan-Rd, Yanji 133002, China
- Cancer Research Center, Yanbian University, Yanji 133002, China
| | - Yongjun Shang
- Department of Orthopedics, Affiliated Hospital of Chifeng University, Chifeng 024000, China
| | - Shuangping Liu
- Department of Pathology, Yanbian University Medical College, No. 977, Gongyuan-Rd, Yanji 133002, China
- Cancer Research Center, Yanbian University, Yanji 133002, China
| | - Yingshi Piao
- Cancer Research Center, Yanbian University, Yanji 133002, China
| | - Xuelian Cui
- Department of Pathology, Yanbian University Medical College, No. 977, Gongyuan-Rd, Yanji 133002, China
| | - Yuzi Li
- Department of Internal Medicine, Yanbian University Hospital, No. 1327, Juzi-St, Yanji 133000, China
| | - Zhenhua Lin
- Department of Pathology, Yanbian University Medical College, No. 977, Gongyuan-Rd, Yanji 133002, China
- Cancer Research Center, Yanbian University, Yanji 133002, China
| |
Collapse
|
21
|
Broxmeyer HE, Mor-Vaknin N, Kappes F, Legendre M, Saha AK, Ou X, O'Leary H, Capitano M, Cooper S, Markovitz DM. Concise review: role of DEK in stem/progenitor cell biology. Stem Cells 2013; 31:1447-53. [PMID: 23733396 PMCID: PMC3814160 DOI: 10.1002/stem.1443] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 12/19/2022]
Abstract
Understanding the factors that regulate hematopoiesis opens up the possibility of modifying these factors and their actions for clinical benefit. DEK, a non-histone nuclear phosphoprotein initially identified as a putative proto-oncogene, has recently been linked to regulate hematopoiesis. DEK has myelosuppressive activity in vitro on proliferation of human and mouse hematopoietic progenitor cells and enhancing activity on engraftment of long-term marrow repopulating mouse stem cells, has been linked in coordinate regulation with the transcription factor C/EBPα, for differentiation of myeloid cells, and apparently targets a long-term repopulating hematopoietic stem cell for leukemic transformation. This review covers the uniqueness of DEK, what is known about how it now functions as a nuclear protein and also as a secreted molecule that can act in paracrine fashion, and how it may be regulated in part by dipeptidylpeptidase 4, an enzyme known to truncate and modify a number of proteins involved in activities on hematopoietic cells. Examples are provided of possible future areas of investigation needed to better understand how DEK may be regulated and function as a regulator of hematopoiesis, information possibly translatable to other normal and diseased immature cell systems.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
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: 3.2] [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
|
23
|
Privette Vinnedge LM, Kappes F, Nassar N, Wells SI. Stacking the DEK: from chromatin topology to cancer stem cells. Cell Cycle 2013; 12:51-66. [PMID: 23255114 PMCID: PMC3570517 DOI: 10.4161/cc.23121] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stem cells are essential for development and tissue maintenance and display molecular markers and functions distinct from those of differentiated cell types in a given tissue. Malignant cells that exhibit stem cell-like activities have been detected in many types of cancers and have been implicated in cancer recurrence and drug resistance. Normal stem cells and cancer stem cells have striking commonalities, including shared cell surface markers and signal transduction pathways responsible for regulating quiescence vs. proliferation, self-renewal, pluripotency and differentiation. As the search continues for markers that distinguish between stem cells, progenitor cells and cancer stem cells, growing evidence suggests that a unique chromatin-associated protein called DEK may confer stem cell-like qualities. Here, we briefly describe current knowledge regarding stem and progenitor cells. We then focus on new findings that implicate DEK as a regulator of stem and progenitor cell qualities, potentially through its unusual functions in the regulation of local or global chromatin organization.
Collapse
Affiliation(s)
- Lisa M Privette Vinnedge
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | | | | | | |
Collapse
|
24
|
Kavanaugh GM, Wise-Draper TM, Morreale RJ, Morrison MA, Gole B, Schwemberger S, Tichy ED, Lu L, Babcock GF, Wells JM, Drissi R, Bissler JJ, Stambrook PJ, Andreassen PR, Wiesmüller L, Wells SI. The human DEK oncogene regulates DNA damage response signaling and repair. Nucleic Acids Res 2011; 39:7465-76. [PMID: 21653549 PMCID: PMC3177200 DOI: 10.1093/nar/gkr454] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 05/16/2011] [Indexed: 12/04/2022] Open
Abstract
The human DEK gene is frequently overexpressed and sometimes amplified in human cancer. Consistent with oncogenic functions, Dek knockout mice are partially resistant to chemically induced papilloma formation. Additionally, DEK knockdown in vitro sensitizes cancer cells to DNA damaging agents and induces cell death via p53-dependent and -independent mechanisms. Here we report that DEK is important for DNA double-strand break repair. DEK depletion in human cancer cell lines and xenografts was sufficient to induce a DNA damage response as assessed by detection of γH2AX and FANCD2. Phosphorylation of H2AX was accompanied by contrasting activation and suppression, respectively, of the ATM and DNA-PK pathways. Similar DNA damage responses were observed in primary Dek knockout mouse embryonic fibroblasts (MEFs), along with increased levels of DNA damage and exaggerated induction of senescence in response to genotoxic stress. Importantly, Dek knockout MEFs exhibited distinct defects in non-homologous end joining (NHEJ) when compared to their wild-type counterparts. Taken together, the data demonstrate new molecular links between DEK and DNA damage response signaling pathways, and suggest that DEK contributes to DNA repair.
Collapse
Affiliation(s)
- Gina M. Kavanaugh
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Trisha M. Wise-Draper
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Richard J. Morreale
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Monique A. Morrison
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Boris Gole
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sandy Schwemberger
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Elisia D. Tichy
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lu Lu
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - George F. Babcock
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - James M. Wells
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rachid Drissi
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - John J. Bissler
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Peter J. Stambrook
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Paul R. Andreassen
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lisa Wiesmüller
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Susanne I. Wells
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
25
|
Kappes F, Khodadoust MS, Yu L, Kim DSL, Fullen DR, Markovitz DM, Ma L. DEK expression in melanocytic lesions. Hum Pathol 2011; 42:932-8. [PMID: 21316078 PMCID: PMC3162348 DOI: 10.1016/j.humpath.2010.10.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 10/14/2010] [Accepted: 10/20/2010] [Indexed: 12/14/2022]
Abstract
The diagnosis of malignant melanoma presents a clinical challenge and relies principally on histopathological evaluation. Previous studies have indicated that increased expression of the DEK oncogene, a chromatin-bound factor, could contribute to the development of melanoma and may be a frequent event in melanoma progression. Here, we investigated DEK expression by immunohistochemistry in a total of 147 melanocytic lesions, including ordinary nevi, dysplastic nevi, Spitz nevi, melanoma in situ, primary invasive melanomas, and metastatic melanomas. Most benign nevi (ordinary, dysplastic, and Spitz nevi) were negative or exhibited weak staining for DEK, with only 4 of 49 cases showing strong staining. Similar to benign nevi, melanoma in situ also demonstrated low levels of DEK expression. In contrast, the expression of DEK in primary invasive melanomas was significantly higher than benign nevi (P < .0001). Moreover, DEK expression was significantly increased in deep melanomas (Breslow depth >1 mm) and metastatic melanomas as compared with superficial melanomas (Breslow depth ≤1 mm) (P < .05). Our findings indicate that DEK overexpression may be a frequent event in invasive melanomas, and further augmentation of DEK expression may be associated with the acquisition of ominous features such as deep dermal invasion and metastasis. These data suggest a role of DEK in melanoma progression.
Collapse
Affiliation(s)
- Ferdinand Kappes
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Michael S Khodadoust
- Program in Immunology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Limin Yu
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - David SL Kim
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Douglas R Fullen
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - David M Markovitz
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, MI, USA
- Program in Immunology, University of Michigan Medical Center, Ann Arbor, MI, USA
- Program in Cellular & Molecular Biology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Linglei Ma
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
26
|
Mor-Vaknin N, Kappes F, Dick AE, Legendre M, Damoc C, Teitz-Tennenbaum S, Kwok R, Ferrando-May E, Adams BS, Markovitz DM. DEK in the synovium of patients with juvenile idiopathic arthritis: characterization of DEK antibodies and posttranslational modification of the DEK autoantigen. ARTHRITIS AND RHEUMATISM 2011; 63:556-67. [PMID: 21280010 PMCID: PMC3117121 DOI: 10.1002/art.30138] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE DEK is a nuclear phosphoprotein and autoantigen in a subset of children with juvenile idiopathic arthritis (JIA). Autoantibodies to DEK are also found in a broad spectrum of disorders associated with abnormal immune activation. We previously demonstrated that DEK is secreted by macrophages, is released by apoptotic T cells, and attracts leukocytes. Since DEK has been identified in the synovial fluid (SF) of patients with JIA, this study was undertaken to investigate how DEK protein and/or autoantibodies may contribute to the pathogenesis of JIA. METHODS DEK autoantibodies, immune complexes (ICs), and synovial macrophages were purified from the SF of patients with JIA. DEK autoantibodies and ICs were purified by affinity-column chromatography and analyzed by 2-dimensional gel electrophoresis, immunoblotting, and enzyme-linked immunosorbent assay. DEK in supernatants and exosomes was purified by serial centrifugation and immunoprecipitation with magnetic beads, and posttranslational modifications of DEK were identified by nano-liquid chromatography tandem mass spectrometry (nano-LC-MS/MS). RESULTS DEK autoantibodies and protein were found in the SF of patients with JIA. Secretion of DEK by synovial macrophages was observed both in a free form and via exosomes. DEK autoantibodies (IgG2) may activate the complement cascade, primarily recognize the C-terminal portion of DEK protein, and exhibit higher affinity for acetylated DEK. Consistent with these observations, DEK underwent acetylation on an unprecedented number of lysine residues, as demonstrated by nano-LC-MS/MS. CONCLUSION These results indicate that DEK can contribute directly to joint inflammation in JIA by generating ICs through high-affinity interaction between DEK and DEK autoantibodies, a process enhanced by acetylation of DEK in the inflamed joint.
Collapse
|
27
|
Fahrer J, Popp O, Malanga M, Beneke S, Markovitz DM, Ferrando-May E, Bürkle A, Kappes F. High-affinity interaction of poly(ADP-ribose) and the human DEK oncoprotein depends upon chain length. Biochemistry 2010; 49:7119-30. [PMID: 20669926 PMCID: PMC2929705 DOI: 10.1021/bi1004365] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is a molecular DNA damage sensor that catalyzes the synthesis of the complex biopolymer poly(ADP-ribose) (PAR) under consumption of NAD(+). PAR engages in fundamental cellular processes such as DNA metabolism and transcription and interacts noncovalently with specific binding proteins involved in DNA repair and regulation of chromatin structure. A factor implicated in DNA repair and chromatin organization is the DEK oncoprotein, an abundant and conserved constituent of metazoan chromatin, and the only member of its protein class. We have recently demonstrated that DEK, under stress conditions, is covalently modified with PAR by PARP-1, leading to a partial release of DEK into the cytoplasm. Additionally, we have also observed a noncovalent interaction between DEK and PAR, which we detail here. Using sequence alignment, we identify three functional PAR-binding sites in the DEK primary sequence and confirm their functionality in PAR binding studies. Furthermore, we show that the noncovalent binding to DEK is dependent on PAR chain length as revealed by an overlay blot technique and a PAR electrophoretic mobility shift assay. Intriguingly, DEK promotes the formation of a defined complex with a 54mer PAR (K(D) = 6 x 10(-8) M), whereas no specific interaction is detected with a short PAR chain (18mer). In stark contrast to covalent poly(ADP-ribosyl)ation of DEK, the noncovalent interaction does not affect the overall ability of DEK to bind to DNA. Instead the noncovalent interaction interferes with subsequent DNA-dependent multimerization activities of DEK, as seen in South-Western, electrophoretic mobility shift, topology, and aggregation assays. In particular, noncovalent attachment of PAR to DEK promotes the formation of DEK-DEK complexes by competing with DNA binding. This was seen by the reduced affinity of PAR-bound DEK for DNA templates in solution. Taken together, our findings deepen the molecular understanding of the DEK-PAR interplay and support the existence of a cellular "PAR code" represented by PAR chain length.
Collapse
Affiliation(s)
- Jörg Fahrer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Oliver Popp
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Maria Malanga
- Department of Structural and Functional Biology, University Federico II of Naples, Naples, Italy
| | - Sascha Beneke
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany
| | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Cellular & Molecular Biology Program, University of Michigan Medical Center, Ann Arbor, Michigan, USA
- Program in Immunology, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Elisa Ferrando-May
- Bioimaging Center, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Ferdinand Kappes
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
28
|
Shibata T, Kokubu A, Miyamoto M, Hosoda F, Gotoh M, Tsuta K, Asamura H, Matsuno Y, Kondo T, Imoto I, Inazawa J, Hirohashi S. DEK oncoprotein regulates transcriptional modifiers and sustains tumor initiation activity in high-grade neuroendocrine carcinoma of the lung. Oncogene 2010; 29:4671-81. [PMID: 20543864 DOI: 10.1038/onc.2010.217] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Revised: 04/26/2010] [Accepted: 05/02/2010] [Indexed: 01/06/2023]
Abstract
Lung cancer shows diverse histological subtypes. Large-cell neuroendocrine cell carcinoma and small-cell lung carcinoma show similar histological features and clinical behaviors, and can be classified as high-grade neuroendocrine carcinoma (HGNEC) of the lung. Here we elucidated the molecular classification of pulmonary endocrine tumors by copy-number profiling. We compared alterations of copy number with the clinical outcome of HGNEC and identified a chromosomal gain of the DEK oncogene locus (6p22.3) that was significantly associated with poor prognosis. We further confirmed that DEK overexpression was associated with poor prognosis in a larger set of HGNEC. Downregulation of DEK by small hairpin RNA led to a marked reduction of in vitro colony formation, in vivo tumorigenicity and chemo-resistance, and was associated with loss of lung cancer stem cell markers. Gene expression profiling revealed that DEK downregulation was associated with altered expression of transcriptional regulators, which specifically include known targets of interchromosomal translocations in hematopoietic tumors, and knockdown of these epigenetic modifiers affected colony formation activity. Our study showed that DEK overexpression, partly through an increase in its gene dose, mediates the activity of global transcriptional regulators and is associated with tumor initiation activity and poor prognosis in HGNEC.
Collapse
MESH Headings
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/metabolism
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Small Cell/genetics
- Carcinoma, Small Cell/metabolism
- Carcinoma, Small Cell/pathology
- Cell Growth Processes/genetics
- Cell Movement/genetics
- Chromosomal Proteins, Non-Histone/biosynthesis
- Chromosomal Proteins, Non-Histone/genetics
- Cluster Analysis
- Down-Regulation
- Gene Dosage
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Neoplastic Stem Cells/pathology
- Oncogene Proteins/biosynthesis
- Oncogene Proteins/genetics
- Poly-ADP-Ribose Binding Proteins
- Prognosis
- RNA, Small Interfering/genetics
- Transcription, Genetic
Collapse
Affiliation(s)
- T Shibata
- Cancer Genomics Project, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Ageberg M, Drott K, Olofsson T, Gullberg U, Lindmark A. Identification of a novel and myeloid specific role of the leukemia-associated fusion protein DEK-NUP214 leading to increased protein synthesis. Genes Chromosomes Cancer 2008; 47:276-87. [PMID: 18181180 DOI: 10.1002/gcc.20531] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
MESH Headings
- Apoptosis
- Blotting, Western
- Eukaryotic Initiation Factor-4E/metabolism
- Flow Cytometry
- Gene Expression Regulation, Leukemic/physiology
- Genes, Reporter
- Genetic Vectors
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Luciferases/metabolism
- Neoplasm Proteins/biosynthesis
- Oncogene Proteins, Fusion/physiology
- Phosphorylation
- Protein Biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Deletion
- Transcription, Genetic
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Malin Ageberg
- Division of Hematology and Transfusion Medicine, Faculty of Medicine, Lund University, Lund, Sweden.
| | | | | | | | | |
Collapse
|
30
|
Nagpal JK, Das BR. Identification of differentially expressed genes in tobacco chewing-mediated oral cancer by differential display-polymerase chain reaction. Eur J Clin Invest 2007; 37:658-64. [PMID: 17635577 DOI: 10.1111/j.1365-2362.2007.01841.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Identification of changes in gene expression that occur in oral squamous cell carcinoma (OSCC), after sufficient characterization, may yield novel molecular markers that may be useful in the diagnosis and disease management of oral cancer. MATERIALS AND METHODS We used differential display-polymerase chain reaction (DD-PCR) to study critically the global gene expression profile of the oral tumour versus normal epithelium. The differential expression of fished out cDNA were confirmed by Northern blot and reverse transcription-PCR. The differentially expressed cDNA were cloned, sequenced and matched for homology in the GenBank database. RESULTS We identified 13 cDNA that showed differential expression. Out of these we selected four cDNA showing consistent reproducibility. One of the cDNA expressed exclusively in tumour had a homology to DEK, a putative oncogene, and is linked to leukaemia, various cancers, HIV infection and several autoimmune disorders. Another cDNA expressed only in tumour had homology to sorcin protein. Sorcin is a 22-kDa calcium-binding protein and is associated with drug resistance in various cell lines. Apparently, sorcin expression might be responsible for drug resistance of OSCC and poor prognosis. Another cDNA showing 10 times overexpression in cheek tumour as compared to normal had homology to CDK6 gene. Hence, it seems from our results that CDK6 is dysregulated during oral carcinogenesis. The fourth cDNA was overexpressed in normal as compared to cheek tumour, but did not show any match in BLAST search. CONCLUSIONS We conclude that there is an enormous significance of these differentially expressed cDNA in oral cancer progression as they can serve as cancer markers to be used for diagnosis and therapeutic intervention.
Collapse
Affiliation(s)
- J K Nagpal
- Institute of Life Sciences, Bhubaneswar, India
| | | |
Collapse
|
31
|
Serum from mice immunized in the context of Treg inhibition identifies DEK as a neuroblastoma tumor antigen. BMC Immunol 2007; 8:4. [PMID: 17397536 PMCID: PMC1852119 DOI: 10.1186/1471-2172-8-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Accepted: 03/30/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We have developed a cell-based vaccine that features the expression of both CD80 and CD86 on the surface of a murine neuroblastoma cell line. The cellular immunity induced by this vaccine is enhanced by treatment with antibody that interferes with T-regulatory cell (Treg) function and we report here that immunization combined with interfering with Treg function also produces a profound serological effect. Serum from mice immunized with our cell-based vaccine in the context of Treg blockade was used to screen a cDNA expression library constructed from the parental neuroblastoma tumor cell line, AGN2a. RESULTS Serum from mice vaccinated in the context of Treg blockade identified a number of potentially oncogenic transcripts that may serve as important immune targets in a tumor-derived cDNA library screen. This novel approach identified far more candidates than could be seen with serum derived from vaccine-treated only, Treg-depleted only, or tumor-bearing mice. The most commonly identified tumor-associated antigen, using serum from immunized and Treg-depleted mice, was the DEK oncogene. Altered expression of the DEK oncogene has been implicated in a number of human cancers. Importantly, we were able to demonstrate that the DEK oncogene also induces a T cell response. CONCLUSION The use of post-vaccine immune serum in this report differs from previous approaches where serum collected at the time of cancer onset or diagnosis and was used for tumor antigen identification. We hypothesize that the use of diagnostic serum samples may be inadequate for the clinical translation of this approach, and that identification of protective immunogenic tumor antigens may require the use of serum from post-treatment or vaccinated subjects. The identification of DEK as a tumor-associated antigen capable of eliciting a T cell response validates our experimental approach and argues for the antigens we have identified here to be evaluated as targets of effector immunity and as vaccine candidates.
Collapse
|
32
|
Napetschnig J, Blobel G, Hoelz A. Crystal structure of the N-terminal domain of the human protooncogene Nup214/CAN. Proc Natl Acad Sci U S A 2007; 104:1783-8. [PMID: 17264208 PMCID: PMC1794303 DOI: 10.1073/pnas.0610828104] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2006] [Indexed: 12/31/2022] Open
Abstract
The mammalian nuclear pore complex (NPC) is an approximately 120-MDa proteinaceous assembly consisting of approximately 30 proteins and is the sole gate in the nuclear envelope. The human protooncogene Nup214 was first identified as a target for chromosomal translocation involved in leukemogenesis. Nup214 is located on the cytoplasmic face of the NPC and is implicated in anchoring the cytoplasmic filaments of the NPC and recruiting the RNA helicase Ddx19. Here, we present the crystal structure of the human Nup214 N-terminal domain at 1.65-A resolution. The structure reveals a seven-bladed beta-propeller followed by a 30-residue C-terminal extended peptide segment, which folds back onto the beta-propeller and binds to its bottom face. The beta-propeller repeats lack any recognizable sequence motif and are distinguished by extensive insertions between the canonical beta-strands. We propose a mechanism by which the C-terminal peptide extension is involved in NPC assembly.
Collapse
Affiliation(s)
- Johanna Napetschnig
- Laboratory of Cell Biology and Howard Hughes Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10021
| | - Günter Blobel
- Laboratory of Cell Biology and Howard Hughes Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10021
| | - André Hoelz
- Laboratory of Cell Biology and Howard Hughes Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10021
| |
Collapse
|
33
|
Mor-Vaknin N, Punturieri A, Sitwala K, Faulkner N, Legendre M, Khodadoust MS, Kappes F, Ruth JH, Koch A, Glass D, Petruzzelli L, Adams BS, Markovitz DM. The DEK nuclear autoantigen is a secreted chemotactic factor. Mol Cell Biol 2006; 26:9484-96. [PMID: 17030615 PMCID: PMC1698538 DOI: 10.1128/mcb.01030-06] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Revised: 07/13/2006] [Accepted: 09/15/2006] [Indexed: 12/17/2022] Open
Abstract
The nuclear DNA-binding protein DEK is an autoantigen that has been implicated in the regulation of transcription, chromatin architecture, and mRNA processing. We demonstrate here that DEK is actively secreted by macrophages and is also found in synovial fluid samples from patients with juvenile arthritis. Secretion of DEK is modulated by casein kinase 2, stimulated by interleukin-8, and inhibited by dexamethasone and cyclosporine A, consistent with a role as a proinflammatory molecule. DEK is secreted in both a free form and in exosomes, vesicular structures in which transcription-modulating factors such as DEK have not previously been found. Furthermore, DEK functions as a chemotactic factor, attracting neutrophils, CD8+ T lymphocytes, and natural killer cells. Therefore, the DEK autoantigen, previously described as a strictly nuclear protein, is secreted and can act as an extracellular chemoattractant, suggesting a direct role for DEK in inflammation.
Collapse
Affiliation(s)
- Nirit Mor-Vaknin
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, MI 48109-0640, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wise-Draper TM, Allen HV, Jones EE, Habash KB, Matsuo H, Wells SI. Apoptosis inhibition by the human DEK oncoprotein involves interference with p53 functions. Mol Cell Biol 2006; 26:7506-19. [PMID: 16894028 PMCID: PMC1636856 DOI: 10.1128/mcb.00430-06] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2006] [Revised: 04/26/2006] [Accepted: 07/26/2006] [Indexed: 11/20/2022] Open
Abstract
The DEK proto-oncogene has been associated with human carcinogenesis-either as a fusion with the CAN nucleoporin protein or when transcriptionally upregulated. Mechanisms of intracellular DEK functions, however, have remained relatively unexplored. We have recently demonstrated that DEK expression is induced by the high-risk human papillomavirus (HPV) E7 protein in a manner which is dependent upon retinoblastoma protein function and have implicated DEK in the inhibition of cellular senescence. Additionally, overexpression of DEK resulted in significant life span extension of primary human keratinocytes. In order to determine whether DEK expression is required for cellular proliferation and/or survival, we monitored cellular responses to the knockdown of DEK in cancer and primary cells. The results indicate that DEK expression protects both HPV-positive cancer and primary human cells from apoptotic cell death. Cell death in response to DEK depletion was accompanied by increased protein stability and transcriptional activity of the p53 tumor suppressor and consequent upregulation of known p53 target genes such as p21CIP and Bax. Consistent with a possible role for p53 in DEK-mediated cell death inhibition, the p53-negative human osteosarcoma cell line SAOS-2 was resistant to the knockdown of DEK. Finally, expression of a dominant negative p53 miniprotein inhibited DEK RNA interference-induced p53 transcriptional induction, as well as cell death, thus directly implicating p53 activation in the observed apoptotic phenotype. These findings suggest a novel role for DEK in cellular survival, involving the destabilization of p53 in a manner which is likely to contribute to human carcinogenesis.
Collapse
Affiliation(s)
- Trisha M Wise-Draper
- Division of Pediatric Hematology/Oncology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA
| | | | | | | | | | | |
Collapse
|
35
|
Sammons M, Wan SS, Vogel NL, Mientjes EJ, Grosveld G, Ashburner BP. Negative regulation of the RelA/p65 transactivation function by the product of the DEK proto-oncogene. J Biol Chem 2006; 281:26802-12. [PMID: 16829531 DOI: 10.1074/jbc.m600915200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NF-kappaB-mediated transcriptional activation is controlled at several levels including interaction with coregulatory proteins. To identify new proteins capable of modulating NF-kappaB-mediated activation, a cytoplasmic two-hybrid screen was performed using the p65 C-terminal transactivation domain as bait and identified the product of the DEK proto-oncogene. DEK is a ubiquitous nuclear protein that has been implicated in several types of cancer and autoimmune diseases. DEK appears to function in several nuclear processes including transcriptional repression and modulation of chromatin structure. Our data indicate that DEK functions as a transcriptional corepressor to repress NF-kappaB activity. DEK expression blocked p65-mediated activation of an NF-kappaB-dependent reporter gene and also inhibited TNFalpha-induced activation of the reporter gene. Chromatin Immunoprecipitation (ChIP) assays showed that DEK associates with the promoters of the NF-kappaB-regulated cIAP2 and IL-8 genes in untreated cells and dissociates from these promoters upon NF-kappaB binding in response to TNFalpha treatment. Moreover, the expression levels of an NF-kappaB-dependent reporter gene as well as the NF-kappaB-regulated Mcp-1 and IkappaBalpha genes is increased in DEK-/- cells compared with wild-type cells. ChIP assays on these promoters show enhanced and prolonged binding of p65 and increased recruitment of the P/CAF coactivator. Overall, these data provide further evidence that DEK functions to negatively regulate transcription.
Collapse
Affiliation(s)
- Morgan Sammons
- Department of Biological Sciences and Undergraduate Honors Program, University of Toledo, Toledo, Ohio 43606, and Department of Genetics and Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | | | |
Collapse
|
36
|
Wise-Draper TM, Allen HV, Thobe MN, Jones EE, Habash KB, Münger K, Wells SI. The human DEK proto-oncogene is a senescence inhibitor and an upregulated target of high-risk human papillomavirus E7. J Virol 2005; 79:14309-17. [PMID: 16254365 PMCID: PMC1280217 DOI: 10.1128/jvi.79.22.14309-14317.2005] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2005] [Accepted: 08/19/2005] [Indexed: 12/24/2022] Open
Abstract
The human DEK proto-oncogene is a nucleic acid binding protein with suspected roles in human carcinogenesis, autoimmune disease, and viral infection. Intracellular DEK functions, however, are poorly understood. In papillomavirus-positive cervical cancer cells, downregulation of viral E6/E7 oncogene expression results in cellular senescence. We report here the specific repression of DEK message and protein levels in senescing human papillomavirus type 16- (HPV16-) and HPV18-positive cancer cell lines as well as in primary cells undergoing replicative senescence. Cervical cancer cell senescence was partially overcome by DEK overexpression, and DEK overexpression was sufficient for extending the life span of primary keratinocytes, supporting critical roles for this molecule as a senescence regulator. In order to determine whether DEK is a bona fide HPV oncogene target in primary cells, DEK expression was monitored in human keratinocytes transduced with HPV E6 and/or E7. The results identify high-risk HPV E7 as a positive DEK regulator, an activity that is not shared by low-risk HPV E7 protein. Experiments in mouse embryo fibroblasts recapitulated the observed E7-mediated DEK induction and demonstrated that both basal and E7-induced regulation of DEK expression are controlled by the retinoblastoma protein family. Taken together, our results suggest that DEK upregulation may be a common event in human carcinogenesis and may reflect its senescence inhibitory function.
Collapse
Affiliation(s)
- Trisha M Wise-Draper
- Division of Hematology/Oncology, Children's Hospital Medical Center and The University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Cleary J, Sitwala KV, Khodadoust MS, Kwok RPS, Mor-Vaknin N, Cebrat M, Cole PA, Markovitz DM. p300/CBP-associated factor drives DEK into interchromatin granule clusters. J Biol Chem 2005; 280:31760-7. [PMID: 15987677 DOI: 10.1074/jbc.m500884200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
DEK is a mammalian protein that has been implicated in the pathogenesis of autoimmune diseases and cancer, including acute myeloid leukemia, melanoma, glioblastoma, hepatocellular carcinoma, and bladder cancer. In addition, DEK appears to participate in multiple cellular processes, including transcriptional repression, mRNA processing, and chromatin remodeling. Sub-nuclear distribution of this protein, with the attendant functional ramifications, has remained a controversial topic. Here we report that DEK undergoes acetylation in vivo at lysine residues within the first 70 N-terminal amino acids. Acetylation of DEK decreases its affinity for DNA elements within the promoter, which is consistent with the involvement of DEK in transcriptional repression. Furthermore, deacetylase inhibition results in accumulation of DEK within interchromatin granule clusters (IGCs), sub-nuclear structures that contain RNA processing factors. Overexpression of P/CAF acetylase drives DEK into IGCs, and addition of a newly developed, synthetic, cell-permeable P/CAF inhibitor blocks this movement. To our knowledge, this is the first reported example of acetylation playing a direct role in relocation of a protein to IGCs, and this may explain how DEK can function in multiple pathways that take place in distinct sub-nuclear compartments. These findings also suggest that DEK-associated malignancies and autoimmune diseases might be amenable to treatment with agents that alter acetylation.
Collapse
Affiliation(s)
- Joanne Cleary
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, Michigan 48109-0640, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Namiki T, Yanagawa S, Izumo T, Ishikawa M, Tachibana M, Kawakami Y, Yokozeki H, Nishioka K, Kaneko Y. Genomic alterations in primary cutaneous melanomas detected by metaphase comparative genomic hybridization with laser capture or manual microdissection: 6p gains may predict poor outcome. ACTA ACUST UNITED AC 2005; 157:1-11. [PMID: 15676140 DOI: 10.1016/j.cancergencyto.2004.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Revised: 05/31/2004] [Accepted: 06/04/2004] [Indexed: 11/15/2022]
Abstract
To clarify the correlation of genomic alterations with clinical and histological features, we performed metaphase comparative genomic hybridization analysis on 20 primary cutaneous melanomas, which were obtained by laser capture or manual microdissection, and 16 melanoma cell lines. There were no differences in the average number of aberrations between acral melanomas (AM) and non-AM, although gains of 5q and 11q13 were more frequent (P=0.05) and 10q loss was less frequent (P=0.01) in AM than in non-AM. Although tumor thickness is considered a measurable estimate of clinical expression, there were no differences in the average number of aberrations among 4 groups, classified by thickness of the tumor. While the majority of aberrations were equally distributed among the 4 groups, 6p gains were found only in the thickest tumors. Patients with 6p or 1q gains had a lower overall survival rate than those without them (P=0.0002 or P=0.013). While gains of 1q, 2q, 3p, 3q, 7q, 20p, and 20q were more frequent in the cell lines than in the primary tumors (P<0.01), losses of 6q, 9p, 10p, and 10q were equally found in both cell lines and primary tumors. The present study showed that chromosomal aberrations had already occurred in the thinner tumors, and that 6p and 1q gains may be a prognostic factor.
Collapse
Affiliation(s)
- Takeshi Namiki
- Research Institute for Clinical Oncology and Department of Pathology, Saitama Cancer Center, 818 Komuro, Ina, Saitama, 362-0806 Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Waldmann T, Scholten I, Kappes F, Hu HG, Knippers R. The DEK protein--an abundant and ubiquitous constituent of mammalian chromatin. Gene 2004; 343:1-9. [PMID: 15563827 DOI: 10.1016/j.gene.2004.08.029] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Revised: 08/09/2004] [Accepted: 08/25/2004] [Indexed: 11/21/2022]
Abstract
The protein DEK is an abundant and ubiquitous chromatin protein in multicellular organisms (not in yeast). It is expressed in more than a million copies/nucleus of rapidly proliferating mammalian cells. DEK has two DNA binding modules of which one includes a SAP box, a sequence motif that DEK shares with a number of other chromatin proteins. DEK has no apparent affinity to specific DNA sequences, but preferentially binds to superhelical and cruciform DNA, and induces positive supercoils into closed circular DNA. The available evidence strongly suggests that DEK could function as an architectural protein in chromatin comparable to the better known classic architectural chromatin proteins, the high-mobility group or HMG proteins.
Collapse
Affiliation(s)
- Tanja Waldmann
- University of Konstanz, Department of Biology, 78457 Konstanz, Germany.
| | | | | | | | | |
Collapse
|
40
|
Lemieux AM, Paré ME, Audet B, Legault E, Lefort S, Boucher N, Landry S, van Opijnen T, Berkhout B, Naghavi MH, Tremblay MJ, Barbeau B. T-cell activation leads to poor activation of the HIV-1 clade E long terminal repeat and weak association of nuclear factor-kappaB and NFAT with its enhancer region. J Biol Chem 2004; 279:52949-60. [PMID: 15466412 DOI: 10.1074/jbc.m409896200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The enhancer region in the human immunodeficiency virus type 1 (HIV-1) 5'-long terminal repeat (LTR) is very important for viral transcription. This promoter sequence binds both nuclear factor-kappaB and NFAT, two important modulators of HIV-1 gene expression. Previous studies have indicated that the enhancer regions of the different HIV-1 clade LTRs differ in their number of NF-kappaB-binding sites. In this study, we have compared the activation potential of the different HIV-1 clade and HIV-2 LTRs and assessed their interaction with NFAT and NF-kappaB. In T-cell lines and primary CD4(+) T-cells, the results showed that the HIV-1 clade E LTR (with a single NF-kappaB-binding site) was the weakest LTR regardless of the tested activators, whereas the HIV-2 LTR was the most responsive LTR. The clade E enhancer region was also demonstrated to be the weakest enhancer region in transfection experiments with luciferase reporter-based vectors. Electrophoretic mobility shift assays with extracts from activated CD4(+) T-cells indicated that, although NF-kappaB and NFAT bound all enhancers, HIV-1 clade E and HIV-2 LTR enhancers were poor binding targets for these two factors. Weak NFAT binding to clade E enhancers was also confirmed using NFAT1-expressing 293T cells in competition experiments. We have also shown the absence of interaction of NF-kappaB or NFAT with the third NF-kappaB repeat present in clade C. However, the clade C enhancer bound NFAT more efficiently than all other enhancer regions tested. Our results hence demonstrate for the first time that differences in the binding of NF-kappaB and NFAT to the enhancer regions could be responsible for some of the observed variation in HIV-1 clade LTR activation, whereas HIV-2 LTR activation seems mostly independent of these interactions.
Collapse
Affiliation(s)
- Anne-Marie Lemieux
- Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Pavillon CHUL, 2705 Blvd. Laurier, Sainte-Foy, Quebec G1V 4G2, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kappes F, Damoc C, Knippers R, Przybylski M, Pinna LA, Gruss C. Phosphorylation by protein kinase CK2 changes the DNA binding properties of the human chromatin protein DEK. Mol Cell Biol 2004; 24:6011-20. [PMID: 15199154 PMCID: PMC480878 DOI: 10.1128/mcb.24.13.6011-6020.2004] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2004] [Accepted: 04/03/2004] [Indexed: 01/15/2023] Open
Abstract
We have examined the posttranslational modification of the human chromatin protein DEK and found that DEK is phosphorylated by the protein kinase CK2 in vitro and in vivo. Phosphorylation sites were mapped by quadrupole ion trap mass spectrometry and found to be clustered in the C-terminal region of the DEK protein. Phosphorylation fluctuates during the cell cycle with a moderate peak during G(1) phase. Filter binding assays, as well as Southwestern analysis, demonstrate that phosphorylation weakens the binding of DEK to DNA. In vivo, however, phosphorylated DEK remains on chromatin. We present evidence that phosphorylated DEK is tethered to chromatin throughout the cell cycle by the un- or underphosphorylated form of DEK.
Collapse
|
42
|
Kappes F, Scholten I, Richter N, Gruss C, Waldmann T. Functional domains of the ubiquitous chromatin protein DEK. Mol Cell Biol 2004; 24:6000-10. [PMID: 15199153 PMCID: PMC480879 DOI: 10.1128/mcb.24.13.6000-6010.2004] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2004] [Revised: 04/03/2004] [Accepted: 04/09/2004] [Indexed: 02/07/2023] Open
Abstract
DEK was originally described as a proto-oncogene protein and is now known to be a major component of metazoan chromatin. DEK is able to modify the structure of DNA by introducing supercoils. In order to find interaction partners and functional domains of DEK, we performed yeast two-hybrid screens and mutational analyses. Two-hybrid screening yielded C-terminal fragments of DEK, suggesting that DEK is able to multimerize. We could localize the domain to amino acids 270 to 350 and show that multimerization is dependent on phosphorylation by CK2 kinase in vitro. We also found two DNA binding domains of DEK, one on a fragment including amino acids 87 to 187 and containing the SAF-box DNA binding motif, which is located between amino acids 149 and 187. This region is sufficient to introduce supercoils into DNA. The second DNA binding domain is located between amino acids 270 and 350 and thus overlaps the multimerization domain. We show that the two DNA-interacting domains differ in their binding properties and in their abilities to respond to CK2 phosphorylation.
Collapse
|
43
|
Waldmann T, Baack M, Richter N, Gruss C. Structure-specific binding of the proto-oncogene protein DEK to DNA. Nucleic Acids Res 2004; 31:7003-10. [PMID: 14627833 PMCID: PMC290247 DOI: 10.1093/nar/gkg864] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The ubiquitous proto-oncogene protein DEK has been found to be associated with chromatin during the entire cell cycle. It changes the topology of DNA in chromatin and protein-free DNA through the introduction of positive supercoils. The sequence and structure specificities of DEK-DNA interactions are not completely understood. The binding of DEK to DNA is not sequence specific, but we describe here that DEK has a clear preference for supercoiled and four-way junction DNA. In the presence of topoisomerase II, DEK stimulates intermolecular catenation of circular DNA molecules. DEK also increases the probability of intermolecular ligation of linear DNA molecules by DNA ligase. These binding properties qualify DEK as an architectural protein.
Collapse
Affiliation(s)
- Tanja Waldmann
- University of Konstanz, Department of Biology, D-78457 Konstanz, Germany
| | | | | | | |
Collapse
|
44
|
Ouellet M, Barbeau B, Tremblay MJ. Protein tyrosyl phosphatases in T cell activation: implication for human immunodeficiency virus transcriptional activity. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2004; 73:69-105. [PMID: 12882515 DOI: 10.1016/s0079-6603(03)01003-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The protein tyrosine phosphatases (PTPs) superfamily is a large group of enzymes showing a wide diversity of structure and biological functions. Their implication in the regulation of signal transduction processes is critical for homeostasis and efficient cellular activation. Disturbance of the delicate balance between protein tyrosine kinase and protein tyrosine phosphatase activities is at the heart of a large number of diseases. Control of cellular activation is especially important for human immunodeficiency virus type 1 (HIV-1) since this retrovirus requires activated T cells in order to replicate efficiently. Identification of PTPs implicated in signaling pathways leading to upregulation of HIV-1 gene transcription therefore contributes to the general understanding of cellular factors needed for strong HIV-1 replication and progression to AIDS. The use of bisperoxovanadium compounds as potent, specific, and highly purified PTP inhibitors releases HIV-1 from PTP control and strongly increases HIV-1 gene expression. These inhibitors can thus be used to study signal transduction mechanisms regulated by PTP activity that are important for HIV-1 replication and provide new and interesting therapeutic avenues for the efficient control of this debilitating retroviral infection.
Collapse
Affiliation(s)
- Michel Ouellet
- Centre de Recherche en Infectiologie, Hôpital CHUL, Centre Hospitalier Universitaire de Québec, Canada, G1V 4G2
| | | | | |
Collapse
|
45
|
McCracken S, Longman D, Johnstone IL, Cáceres JF, Blencowe BJ. An evolutionarily conserved role for SRm160 in 3'-end processing that functions independently of exon junction complex formation. J Biol Chem 2003; 278:44153-60. [PMID: 12944400 DOI: 10.1074/jbc.m306856200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SRm160 (the SR-related nuclear matrix protein of 160 kDa) functions as a splicing coactivator and 3'-end cleavage-stimulatory factor. It is also a component of the splicing-dependent exon-junction complex (EJC), which has been implicated in coupling of pre-mRNA splicing with mRNA turnover and mRNA export. We have investigated whether the association of SRm160 with the EJC is important for efficient 3'-end cleavage. The EJC components RNPS1, REF, UAP56, and Y14 interact with SRm160. However, when these factors were tethered to transcripts, only SRm160 and RNPS1 stimulated 3'-end cleavage. Whereas SRm160 stimulated cleavage to a similar extent in the presence or absence of an active intron, stimulation of 3'-end cleavage by tethered RNPS1 is dependent on an active intron. Assembly of an EJC adjacent to the cleavage and polyadenylation signal in vitro did not significantly affect cleavage efficiency. These results suggest that SRm160 stimulates cleavage independently of its association with EJC components and that the cleavage-stimulatory activity of RNPS1 may be an indirect consequence of its ability to stimulate splicing. Using RNA interference (RNAi) in Caenorhabditis elegans, we determined whether interactions between SRm160 and the cleavage machinery are important in a whole organism context. Simultaneous RNAi of SRm160 and the cleavage factor CstF-50 (Cleavage stimulation factor 50-kDa subunit) resulted in late embryonic developmental arrest. In contrast, RNAi of CstF-50 in combination with RNPS1 or REFs did not result in an apparent phenotype. Our combined results provide evidence for an evolutionarily conserved interaction between SRm160 and the 3'-end cleavage machinery that functions independently of EJC formation.
Collapse
Affiliation(s)
- Susan McCracken
- Banting and Best Department of Medical Research, C. H. Best Institute, University of Toronto, Toronto, Ontario M5G 1L6, Canada
| | | | | | | | | |
Collapse
|
46
|
Chan EM, Bulman DE, Paterson AD, Turnbull J, Andermann E, Andermann F, Rouleau GA, Delgado-Escueta AV, Scherer SW, Minassian BA. Genetic mapping of a new Lafora progressive myoclonus epilepsy locus (EPM2B) on 6p22. J Med Genet 2003; 40:671-5. [PMID: 12960212 PMCID: PMC1735578 DOI: 10.1136/jmg.40.9.671] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Lafora disease is a progressive myoclonus epilepsy with polyglucosan accumulations and a peculiar neurodegeneration with generalised organellar disintegration. It causes severe seizures, leading to dementia and eventually death in early adulthood. METHODS One Lafora disease gene, EPM2A, has been identified on chromosome 6q24. Locus heterogeneity led us to search for a second gene using a genome wide linkage scan in French-Canadian families. RESULTS We mapped a second Lafora disease locus, EPM2B, to a 2.2 Mb region at 6p22, a region known to code for several proteins, including kinesins. Kinesins are microtubule dependent motor proteins that are involved in transporting cellular components. In neurones, they play a major role in axonal and dendritic transport. CONCLUSION Analysis of the present locus in other non-EPM2A families will reveal whether there is further locus heterogeneity. Identification of the disease gene will be of major importance towards our understanding of the pathogenesis of Lafora disease.
Collapse
Affiliation(s)
- E M Chan
- Program in Genetics and Genomic Biology, The Hospital for Sick Children and Department of Molecular and Medical Genetics, The University of Toronto, 555 University Ave, Toronto, Ontario M5G 1X8, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Adams BS, Cha HC, Cleary J, Haiying T, Wang H, Sitwala K, Markovitz DM. DEK binding to class II MHC Y-box sequences is gene- and allele-specific. Arthritis Res Ther 2003; 5:R226-33. [PMID: 12823858 PMCID: PMC165066 DOI: 10.1186/ar774] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2002] [Revised: 04/08/2003] [Accepted: 04/29/2003] [Indexed: 02/07/2023] Open
Abstract
Using electrophoretic mobility shift assays, we examined sequence-specific binding of DEK, a potential autoantigen in juvenile rheumatoid arthritis, to conserved Y-box regulatory sequences in class II MHC gene promoters. Nuclear extracts from several cell lines of different phenotypes contained sequence-specific binding activity recognizing DRA, DQA1*0101, and DQA1*0501 Y-box sequences. Participation of both DEK and NF-Y in the DQA1 Y-box binding complex was confirmed by 'supershifting' with anti-DEK and anti-NF-Y antibodies. Recombinant DEK also bound specifically to the DQA1*0101 Y box and to the polymorphic DQA1*0501 Y box, but not to the consensus DRA Y box. Measurement of the apparent dissociation constants demonstrated a two- to fivefold difference in DEK binding to the DQA1 Y-box sequence in comparison with other class II MHC Y-box sequences. Residues that are crucial for DEK binding to the DQA1*0101 Y box were identified by DNase I footprinting. The specific characteristics of DEK binding to these related sequences suggests a potential role for DEK in differential regulation of class II MHC expression, and thus in the pathogenesis of juvenile rheumatoid arthritis and other autoimmune diseases.
Collapse
Affiliation(s)
- Barbara S Adams
- Department of Pediatrics, Division of Pediatric Rheumatology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Hyuk C Cha
- Department of Pediatrics, Division of Pediatric Rheumatology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Joanne Cleary
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Tan Haiying
- Department of Pediatrics, Division of Pediatric Rheumatology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Hongling Wang
- Department of Pediatrics, Division of Pediatric Rheumatology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Kajal Sitwala
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - David M Markovitz
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| |
Collapse
|
48
|
Campillos M, García MA, Valdivieso F, Vázquez J. Transcriptional activation by AP-2alpha is modulated by the oncogene DEK. Nucleic Acids Res 2003; 31:1571-5. [PMID: 12595566 PMCID: PMC149840 DOI: 10.1093/nar/gkg247] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2002] [Revised: 11/15/2002] [Accepted: 01/12/2003] [Indexed: 12/30/2022] Open
Abstract
Cell differentiation and development are highly regulated processes at the transcriptional level. One of the main transcription factors that regulate these processes is AP-2alpha, a cell-type specific protein required for vertebrate development and embryogenesis. AP-2alpha also regulates apoptosis and cell-cycle specific events by interacting with the oncogene c-Myc. In searching for novel AP-2alpha- interacting factors, using an affinity chromatography approach, we have observed that oncoprotein DEK interacts with AP-2alpha in vitro. The existence of an interaction between AP-2alpha and DEK in cellular cultures was demonstrated by expression of a tagged AP-2alpha form followed by immunodetection. By transient co-expression experiments using a reporter for APOE promoter activity we have found that DEK stimulates the transactivation activity of AP-2alpha over APOE promoter. Finally, electrophoretic mobility shift assays suggested that DEK enhances the DNA-binding activity of AP-2alpha. Our data suggest a novel cellular function of DEK as a transcriptional co-activator.
Collapse
Affiliation(s)
- Mónica Campillos
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, 28049 Cantoblanco, Madrid, Spain
| | | | | | | |
Collapse
|
49
|
Sitwala KV, Adams K, Markovitz DM. YY1 and NF-Y binding sites regulate the transcriptional activity of the dek and dek-can promoter. Oncogene 2002; 21:8862-70. [PMID: 12483538 DOI: 10.1038/sj.onc.1206041] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2002] [Revised: 08/27/2002] [Accepted: 09/11/2002] [Indexed: 11/09/2022]
Abstract
The mammalian protein DEK has been implicated in multiple cellular processes, including transcriptional regulation, mRNA processing, and chromatin remodeling, and is associated with a number of clinical autoimmune and neoplastic conditions. The connection between DEK and cancer exists at multiple levels: (a) the t(6;9) chromosomal translocation that characterizes a subtype of acute myelogenous leukemia cases results in the formation of a DEK-CAN fusion oncoprotein; (b) a fragment of dek cDNA is capable of partially reversing the radiation-sensitive phenotype of fibroblasts cultured from ataxia-telangiectasia patients; and (c) increased levels of dek mRNA have been found to be associated with hepatocellular carcinoma, glioblastoma, and melanoma. Despite the growing list of cancer subtypes with a connection to DEK, the factors that mediate its expression have yet to be characterized. Here we undertake the analysis of DEK regulation by mapping the discrete elements within the proximal promoter that are responsible for constitutive transcription of dek in transformed cells. We find that functional elements include an inverted CCAAT box and a YY1 consensus binding site, and the introduction of point mutations into these sites markedly diminishes transcriptional activity. In addition, we identify the transcriptional activator NF-Y as a member of the CCAAT-binding complex, and verify binding of the transcription factor YY1 at its consensus site in the dek promoter. The discovery of NF-Y and YY1 as regulatory determinants of DEK expression is consistent with the well-documented roles of these two factors in cellular proliferation and transformation.
Collapse
Affiliation(s)
- Kajal V Sitwala
- Department of Internal Medicine and Program in Cellular and Molecular Biology, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, Michigan, MI 48109-0640, USA
| | | | | |
Collapse
|
50
|
Krithivas A, Fujimuro M, Weidner M, Young DB, Hayward SD. Protein interactions targeting the latency-associated nuclear antigen of Kaposi's sarcoma-associated herpesvirus to cell chromosomes. J Virol 2002; 76:11596-604. [PMID: 12388720 PMCID: PMC136775 DOI: 10.1128/jvi.76.22.11596-11604.2002] [Citation(s) in RCA: 167] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2002] [Accepted: 08/13/2002] [Indexed: 12/25/2022] Open
Abstract
Maintenance of Kaposi's sarcoma-associated herpesvirus (KSHV) latent infection depends on the viral episomes in the nucleus being distributed to daughter cells following cell division. The latency-associated nuclear antigen (LANA) is constitutively expressed in all KSHV-infected cells. LANA binds sequences in the terminal repeat regions of the KSHV genome and tethers the viral episomes to chromosomes. To better understand the mechanism of chromosomal tethering, we performed glutathione S-transferase (GST) affinity and yeast two-hybrid assays to identify LANA-interacting proteins with known chromosomal association. Two of the interactors were the methyl CpG binding protein MeCP2 and the 43-kDa protein DEK. The interactions of MeCP2 and DEK with LANA were confirmed by coimmunoprecipitation. The MeCP2-interacting domain was mapped to the previously described chromatin binding site in the N terminus of LANA, while the DEK-interacting domain mapped to LANA amino acids 986 to 1043 in the C terminus. LANA was unable to associate with mouse chromosomes in chromosome spreads of transfected NIH 3T3 cells. However, LANA was capable of targeting to mouse chromosomes in the presence of human MeCP2 or DEK. The data indicate that LANA is tethered to chromosomes through two independent chromatin binding domains that interact with different protein partners.
Collapse
Affiliation(s)
- Anita Krithivas
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21231, USA
| | | | | | | | | |
Collapse
|