51
|
Broxmeyer HE, Kappes F, Mor-Vaknin N, Legendre M, Kinzfogl J, Cooper S, Hangoc G, Markovitz DM. DEK regulates hematopoietic stem engraftment and progenitor cell proliferation. Stem Cells Dev 2012; 21:1449-54. [PMID: 21943234 PMCID: PMC3359622 DOI: 10.1089/scd.2011.0451] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 09/17/2011] [Indexed: 12/16/2022] Open
Abstract
DEK is a biochemically distinct protein that is generally found in the nucleus, where it is vital to global heterochromatin integrity. However, DEK is also secreted by cells (eg, macrophages) and influences other adjacent cells (eg, acts as a chemoattractant for certain mature blood cells). We hypothesized that DEK may modulate functions of hematopoietic stem (HSCs) and progenitor (HPCs) cells. C57Bl/6 mice were used to demonstrate that absolute numbers and cycling status of HPCs (colony forming unit-granulocyte macrophage [CFU-GM], burst forming unit-erythroid [BFU-E], and colony forming unit-granulocyte erythroid macrophage megakaryocyte [CFU-GEMM]) in bone marrow (BM) and spleen were significantly enhanced in DEK -/- as compared with wild-type (WT) control mice. Moreover, purified recombinant DEK protein inhibited colony formation in vitro by CFU-GM, BFU-E, and CFU-GEMM from WT BM cells and human cord blood (CB) cells in a dose-dependent fashion, demonstrating that DEK plays a negative role in HPC proliferation in vitro and in vivo. Suppression was direct acting as determined by inhibition of proliferation of single isolated CD34(+) CB cells in vitro. In contrast, DEK -/- BM cells significantly demonstrated reduced long term competitive and secondary mouse repopulating HSC capacity compared with WT BM cells, demonstrating that DEK positively regulates engrafting capability of self-renewing HSCs. This demonstrates that DEK has potent effects on HSCs, HPCs, and hematopoiesis, information of biological and potential clinical interest.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202-5181, USA.
| | | | | | | | | | | | | | | |
Collapse
|
52
|
Liu S, Wang X, Sun F, Kong J, Li Z, Lin Z. DEK overexpression is correlated with the clinical features of breast cancer. Pathol Int 2012; 62:176-81. [PMID: 22360505 DOI: 10.1111/j.1440-1827.2011.02775.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To investigate the clinicopathological significance of DEK overexpression in breast cancers, a total of 196 cases, including 20 of normal tissues, 12 of intraductal hyperplasia, 31 of ductal carcinoma in situ (DCIS) and 133 of invasive ductal carcinoma of the breast, were selected from the Department of Pathology, Yanbian Tumor Hospital for immunohistochemical staining of DEK, estrogen (ER), progesterone (PR) and Ki-67 proteins. In results, DEK protein had higher positivity in DCIS, compared with the adjacent normal breast tissues. Also, DEK protein was strongly positive in invasive ductal carcinoma of the breast on immunohistochemistry, which was significantly higher than normal breast tissues. However, only two (2/12) cases of intraductal hyperplasia of the breast showed positive staining for DEK protein. Additionally, DEK overexpression was significantly correlated with the increased proliferating index of Ki-67. For the histological grade, DEK positive rate was only 39.6% in G1 breast cancers, but significantly higher in G2 (92.3%) and G3 (97.0%) cases (P<0.05). Also, a strongly positive rate of DEK was lower in Stage-0 (21.4%) and Stage-I (40.9%) compared with Stage-IIa (87.5%), Stage-IIb (89.7%) and Stage-IIIa (92.3%) (P<0.05). And DEK protein showed higher expression level in < 3 years disease free survival breast cancers than it did in ≥ 3 years disease free survival cases (P<0.05). However, no statistically difference was found among DEK expression, lymph node metastasis, and ER and PR expressions. In conclusion, DEK overexpression appears to be associated with breast cancer progression and DEK may potentially be used as a breast cancer biomarker for the early diagnosis, prognostic evaluation and therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Shuangping Liu
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | | | | | | | | | | |
Collapse
|
53
|
Kavanaugh GM, Wise-Draper TM, Morreale RJ, Morrison MA, Gole B, Schwemberger S, Tichy ED, Lu L, Babcock GF, Wells JM, Drissi R, Bissler JJ, Stambrook PJ, Andreassen PR, Wiesmüller L, Wells SI. The human DEK oncogene regulates DNA damage response signaling and repair. Nucleic Acids Res 2011; 39:7465-76. [PMID: 21653549 PMCID: PMC3177200 DOI: 10.1093/nar/gkr454] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 05/16/2011] [Indexed: 12/04/2022] Open
Abstract
The human DEK gene is frequently overexpressed and sometimes amplified in human cancer. Consistent with oncogenic functions, Dek knockout mice are partially resistant to chemically induced papilloma formation. Additionally, DEK knockdown in vitro sensitizes cancer cells to DNA damaging agents and induces cell death via p53-dependent and -independent mechanisms. Here we report that DEK is important for DNA double-strand break repair. DEK depletion in human cancer cell lines and xenografts was sufficient to induce a DNA damage response as assessed by detection of γH2AX and FANCD2. Phosphorylation of H2AX was accompanied by contrasting activation and suppression, respectively, of the ATM and DNA-PK pathways. Similar DNA damage responses were observed in primary Dek knockout mouse embryonic fibroblasts (MEFs), along with increased levels of DNA damage and exaggerated induction of senescence in response to genotoxic stress. Importantly, Dek knockout MEFs exhibited distinct defects in non-homologous end joining (NHEJ) when compared to their wild-type counterparts. Taken together, the data demonstrate new molecular links between DEK and DNA damage response signaling pathways, and suggest that DEK contributes to DNA repair.
Collapse
Affiliation(s)
- Gina M. Kavanaugh
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Trisha M. Wise-Draper
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Richard J. Morreale
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Monique A. Morrison
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Boris Gole
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sandy Schwemberger
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Elisia D. Tichy
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lu Lu
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - George F. Babcock
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - James M. Wells
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rachid Drissi
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - John J. Bissler
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Peter J. Stambrook
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Paul R. Andreassen
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lisa Wiesmüller
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Susanne I. Wells
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Ulm University, D-89075 Ulm, Germany, Research, Shriners Hospitals for Children, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati College of Medicine, Division of Molecular and Developmental Biology, Cincinnati Children's Hospital Medical Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
54
|
Brázda V, Laister RC, Jagelská EB, Arrowsmith C. Cruciform structures are a common DNA feature important for regulating biological processes. BMC Mol Biol 2011; 12:33. [PMID: 21816114 PMCID: PMC3176155 DOI: 10.1186/1471-2199-12-33] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 08/05/2011] [Indexed: 04/10/2023] Open
Abstract
DNA cruciforms play an important role in the regulation of natural processes involving DNA. These structures are formed by inverted repeats, and their stability is enhanced by DNA supercoiling. Cruciform structures are fundamentally important for a wide range of biological processes, including replication, regulation of gene expression, nucleosome structure and recombination. They also have been implicated in the evolution and development of diseases including cancer, Werner's syndrome and others. Cruciform structures are targets for many architectural and regulatory proteins, such as histones H1 and H5, topoisomerase IIβ, HMG proteins, HU, p53, the proto-oncogene protein DEK and others. A number of DNA-binding proteins, such as the HMGB-box family members, Rad54, BRCA1 protein, as well as PARP-1 polymerase, possess weak sequence specific DNA binding yet bind preferentially to cruciform structures. Some of these proteins are, in fact, capable of inducing the formation of cruciform structures upon DNA binding. In this article, we review the protein families that are involved in interacting with and regulating cruciform structures, including (a) the junction-resolving enzymes, (b) DNA repair proteins and transcription factors, (c) proteins involved in replication and (d) chromatin-associated proteins. The prevalence of cruciform structures and their roles in protein interactions, epigenetic regulation and the maintenance of cell homeostasis are also discussed.
Collapse
Affiliation(s)
- Václav Brázda
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v,v,i,, Královopolská 135, Brno, 612 65, Czech Republic.
| | | | | | | |
Collapse
|
55
|
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] [Download PDF] [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
|
56
|
Kappes F, Waldmann T, Mathew V, Yu J, Zhang L, Khodadoust MS, Chinnaiyan AM, Luger K, Erhardt S, Schneider R, Markovitz DM. The DEK oncoprotein is a Su(var) that is essential to heterochromatin integrity. Genes Dev 2011; 25:673-8. [PMID: 21460035 PMCID: PMC3070930 DOI: 10.1101/gad.2036411] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2010] [Accepted: 02/09/2011] [Indexed: 12/18/2022]
Abstract
Heterochromatin integrity is crucial for genome stability and regulation of gene expression, but the factors involved in mammalian heterochromatin biology are only incompletely understood. Here we identify the oncoprotein DEK, an abundant nuclear protein with a previously enigmatic in vivo function, as a Suppressor of Variegation [Su(var)] that is crucial to global heterochromatin integrity. We show that DEK interacts directly with Heterochromatin Protein 1 α (HP1α) and markedly enhances its binding to trimethylated H3K9 (H3K9me3), which is key for maintaining heterochromatic regions. Loss of Dek in Drosophila leads to a Su(var) phenotype and global reduction in heterochromatin. Thus, these findings show that DEK is a key factor in maintaining the balance between heterochromatin and euchromatin in vivo.
Collapse
Affiliation(s)
- Ferdinand Kappes
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | - Tanja Waldmann
- Max-Planck-Institute for Immunobiology, 79108 Freiburg, Germany
| | - Veena Mathew
- CellNetworks-Cluster of Excellence, ZMBH-DKFZ-Alliance, ZMBH, Heidelberg University, Heidelberg 69120, Germany
| | - Jindan Yu
- Department of Pathology, Michigan Center for Translational Pathology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | - Ling Zhang
- Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Michael S. Khodadoust
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
- Program in Immunology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | - Arul M. Chinnaiyan
- Department of Pathology, Michigan Center for Translational Pathology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| | - Karolin Luger
- Department of Biochemistry and Molecular Biology, Howard Hughes Medical Institute, Colorado State University, Fort Collins, Colorado 80523, USA
| | - Sylvia Erhardt
- CellNetworks-Cluster of Excellence, ZMBH-DKFZ-Alliance, ZMBH, Heidelberg University, Heidelberg 69120, Germany
| | | | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
- Program in Immunology, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
- Cellular and Molecular Biology Program, University of Michigan Medical Center, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
57
|
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: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [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
|
58
|
Babaei-Jadidi R, Li N, Saadeddin A, Spencer-Dene B, Jandke A, Muhammad B, Ibrahim EE, Muraleedharan R, Abuzinadah M, Davis H, Lewis A, Watson S, Behrens A, Tomlinson I, Nateri AS. FBXW7 influences murine intestinal homeostasis and cancer, targeting Notch, Jun, and DEK for degradation. ACTA ACUST UNITED AC 2011; 208:295-312. [PMID: 21282377 PMCID: PMC3039859 DOI: 10.1084/jem.20100830] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The E3 ubiquitin ligase component FBXW7 modulates homeostasis and inhibits tumorigenesis in the murine intestine. The Fbxw7 (F-box/WD repeat–containing protein 7; also called CDC4, Sel10, Ago, and Fbw7) component of the SCF (Skp1/Cullin/F-box protein) E3 ubiquitin ligase complex acts as a tumor suppressor in several tissues and targets multiple transcriptional activators and protooncogenes for ubiquitin-mediated degradation. To understand Fbxw7 function in the murine intestine, in this study, we specifically deleted Fbxw7 in the murine gut using Villin-Cre (Fbxw7ΔG). In wild-type mice, loss of Fbxw7 in the gut altered homeostasis of the intestinal epithelium, resulted in elevated Notch and c-Jun expression, and induced development of adenomas at 9–10 mo of age. In the context of APC (adenomatous polyposis coli) deficiency (ApcMin/+ mice), loss of Fbxw7 accelerated intestinal tumorigenesis and death and promoted accumulation of β-catenin in adenomas at late but not early time points. At early time points, Fbxw7 mutant tumors showed accumulation of the DEK protooncogene. DEK expression promoted cell division and altered splicing of tropomyosin (TPM) RNA, which may also influence cell proliferation. DEK accumulation and altered TPM RNA splicing were also detected in FBXW7 mutant human colorectal tumor tissues. Given their reduced lifespan and increased incidence of intestinal tumors, ApcMin/+Fbxw7ΔG mice may be used for testing carcinogenicity and drug screening.
Collapse
Affiliation(s)
- Roya Babaei-Jadidi
- Division of Pre-Clinical Oncology, School of Clinical Sciences, University of Nottingham, Nottingham, England, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
59
|
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] [Download PDF] [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
|
60
|
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: 51] [Impact Index Per Article: 3.4] [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
|
61
|
Abstract
Slight modifications of chromatin dynamics can translate into small- and large-scale changes in DNA replication and DNA repair. Similarly, promoter usage and accessibility are tightly dependent on chromatin architecture. Consequently, it is perhaps not surprising that factors controlling chromatin organization are frequently deregulated (directly or indirectly) in cancer cells. DEK is emerging as a novel class of DNA topology modulators that can be both targets and effectors of protumorigenic events. The locus containing DEK at chromosome 6p22.3 is amplified or reorganized in multiple cancer types. In addition, DEK can be subject to a variety of tumor-associated transcriptional and post-translational modifications. In turn, DEK can favor cell transformation, at least in part by inhibiting cell differentiation and premature senescence. More recently, DEK has also been linked to the resistance of malignant cells to apoptotic inducers. Interestingly, a fraction of DEK can also bind RNA and affect alternative splicing, further illustrating the pleiotropic roles that this protein may exert in cancer cells. Here we will summarize the current literature about the regulation and function(s) of DEK as a proto-oncogene. In addition, the translational relevance of DEK as a putative diagnostic marker and candidate for drug development will be discussed.
Collapse
Affiliation(s)
- Erica Riveiro-Falkenbach
- Centro Nacional de Investigaciones Oncológicas (Spanish National Cancer Research Centre), Madrid, Spain
| | | |
Collapse
|
62
|
Khodadoust MS, Verhaegen M, Kappes F, Riveiro-Falkenbach E, Cigudosa JC, Kim DSL, Chinnaiyan AM, Markovitz DM, Soengas MS. Melanoma proliferation and chemoresistance controlled by the DEK oncogene. Cancer Res 2009; 69:6405-13. [PMID: 19679545 PMCID: PMC2727675 DOI: 10.1158/0008-5472.can-09-1063] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gain of chromosome 6p is a consistent feature of advanced melanomas. However, the identity of putative oncogene(s) associated with this amplification has remained elusive. The chromatin remodeling factor DEK is an attractive candidate as it maps to 6p (within common melanoma-amplified loci). Moreover, DEK expression is increased in metastatic melanomas, although the functional relevance of this induction remains unclear. Importantly, in other tumor types, DEK can display various tumorigenic effects in part through its ability to promote proliferation and inhibit p53-dependent apoptosis. Here, we report a generalized up-regulation of DEK protein in aggressive melanoma cells and tumors. In addition, we provide genetic and mechanistic evidence to support a key role of DEK in the maintenance of malignant phenotypes of melanoma cells. Specifically, we show that long-term DEK down-regulation by independent short hairpin RNAs resulted in premature senescence of a variety of melanoma cell lines. Short-term abrogation of DEK expression was also functionally relevant, as it attenuated the traditional resistance of melanomas to DNA-damaging agents. Unexpectedly, DEK short hairpin RNA had no effect on p53 levels or p53-dependent apoptosis. Instead, we identified a new role for DEK in the transcriptional activation of the antiapoptotic MCL-1. Other MCL-1-related factors such as BCL-2 or BCL-xL were unaffected by changes in the endogenous levels of DEK, indicating a selective effect of this gene on the apoptotic machinery of melanoma cells. These results provide support for DEK as a long sought-after oncogene mapping at chromosome 6, with novel functions in melanoma proliferation and chemoresistance.
Collapse
MESH Headings
- Cell Proliferation
- Cells, Cultured
- Cellular Senescence/drug effects
- Cellular Senescence/genetics
- Chromosomal Proteins, Non-Histone/antagonists & inhibitors
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- Chromosomal Proteins, Non-Histone/physiology
- Chromosomes, Human, Pair 6
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Melanoma/genetics
- Melanoma/metabolism
- Melanoma/pathology
- Myeloid Cell Leukemia Sequence 1 Protein
- Oncogene Proteins/antagonists & inhibitors
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Oncogene Proteins/physiology
- Poly-ADP-Ribose Binding Proteins
- Proto-Oncogene Proteins c-bcl-2/metabolism
- RNA, Small Interfering/pharmacology
- Skin Neoplasms/genetics
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Tumor Suppressor Protein p53/metabolism
- bcl-X Protein/metabolism
Collapse
Affiliation(s)
- Michael S Khodadoust
- Program in Immunology, Department of Dermatology, Michigan Center for Translational Pathology, and Howard Hughes Medical Institute, University of Michigan Medical Center, Ann Arbor, Michigan 48109-5640, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Hua Y, Hu H, Peng X. Progress in studies on the DEK protein and its involvement in cellular apoptosis. SCIENCE IN CHINA. SERIES C, LIFE SCIENCES 2009; 52:637-42. [PMID: 19641868 DOI: 10.1007/s11427-009-0088-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2009] [Accepted: 03/30/2009] [Indexed: 12/12/2022]
Abstract
DEK protein is an ubiquitous phosphorylated nuclear protein. Specific binding of DEK to DNA could change the topology of DNA and then affect the gene activity of the underlying DNA sequences. It is speculated that there might be some potential relationship between the stress reaction of cells and DEK proteins. The phosphorylation status of DEK protein is altered during death-receptor-mediated cell apoptosis. Both phosphorylation and poly(ADP-ribosyl)ation could promote the release of DEK from apoptotic nuclei to extracellular environment, and in this case DEK becomes a potential autoantigen of some autoimmune diseases. The available evidence powerfully suggests that DEK protein is closely relevant to apoptosis. The overexpression of DEK protein has dual function in cell apoptosis, in terms of inhibiting or triggering cell apoptosis.
Collapse
Affiliation(s)
- Ying Hua
- College of Life Sciences and Bioengineering, School of Science, Beijing Jiaotong University, Beijing, 100044, China
| | | | | |
Collapse
|
64
|
Han S, Xuan Y, Liu S, Zhang M, Jin D, Jin R, Lin Z. Clinicopathological significance of DEK overexpression in serous ovarian tumors. Pathol Int 2009; 59:443-7. [PMID: 19563407 DOI: 10.1111/j.1440-1827.2009.02392.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To investigate the significance of DEK protein expression in ovarian lesions, a total of 113 ovarian serous tumors, including 62 serous cystadenocarcinomas and 19 serous borderline tumors, were studied on immunohistochemistry. For comparison, 32 benign serous tumors, including 12 serous papillary cystadenomas, 10 serous cystadenomas, and 10 serous surface papillomas, were also included. DEK was positive in 93.5% of serous cystadenocarcinomas (58/62), 63.2% of serous borderline tumors (12/19), and weakly positive in 15.6% of benign serous tumors (5/32). The strong positive signal was detected only in serous adenocarcinomas (80.6%, 50/62) and borderline tumors (21.1%, 4/19), but no serous benign tumors were strongly positive (0%, 0/32). Meanwhile, the strong positivity rate of DEK protein was significantly higher in grade 2 and grade 3 than in grade 1 ovarian cancers (P < 0.05), but there was no significant association between DEK protein expression level and International Federation of Gynecology and Obstetrics (FIGO) stage of serous ovarian adenocarcinoma (P > 0.05). In summary, DEK plays an important role in the progression of ovarian serous cancers. The detection of DEK protein expression should be useful for the diagnosis and prognosis of ovarian serous cancers, and DEK might be a useful molecular target for ovarian cancer therapy.
Collapse
Affiliation(s)
- Songying Han
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | | | | | | | | | | | | |
Collapse
|
65
|
Xu S, Powers MA. Nuclear pore proteins and cancer. Semin Cell Dev Biol 2009; 20:620-30. [PMID: 19577736 PMCID: PMC2706781 DOI: 10.1016/j.semcdb.2009.03.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 03/05/2009] [Accepted: 03/09/2009] [Indexed: 12/28/2022]
Abstract
Nucleocytoplasmic trafficking of macromolecules, a highly specific and tightly regulated process, occurs exclusively through the nuclear pore complex. This immense structure is assembled from approximately 30 proteins, termed nucleoporins. Here we discuss the four nucleoporins that have been linked to cancers, either through elevated expression in tumors (Nup88) or through involvement in chromosomal translocations that encode chimeric fusion proteins (Tpr, Nup98, Nup214). In each case we consider the normal function of the nucleoporin and its translocation partners, as well as what is known about their mechanistic contributions to carcinogenesis, particularly in leukemias. Studies of nucleoporin-linked cancers have revealed novel mechanisms of oncogenesis and in the future, should continue to expand our understanding of cancer biology.
Collapse
Affiliation(s)
- Songli Xu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| | - Maureen A. Powers
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
66
|
Wise-Draper TM, Morreale RJ, Morris TA, Mintz-Cole RA, Hoskins EE, Balsitis SJ, Husseinzadeh N, Witte DP, Wikenheiser-Brokamp KA, Lambert PF, Wells SI. DEK proto-oncogene expression interferes with the normal epithelial differentiation program. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:71-81. [PMID: 19036808 PMCID: PMC2631320 DOI: 10.2353/ajpath.2009.080330] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Accepted: 09/18/2008] [Indexed: 01/14/2023]
Abstract
Overexpression of the DEK gene is associated with multiple human cancers, but its specific roles as a putative oncogene are not well defined. DEK transcription was previously shown to be induced by the high-risk human papillomavirus (HPV) E7 oncogene via E2F and Rb pathways. Transient DEK overexpression was able to inhibit both senescence and apoptosis in cultured cells. In at least the latter case, this mechanism involved the destabilization of p53 and the decreased expression of p53 target genes. We show here that DEK overexpression disrupts the normal differentiation program in a manner that is independent of either p53 or cell death. DEK expression was distinctly repressed upon the differentiation of cultured primary human keratinocytes, and stable DEK overexpression caused epidermal thickening in an organotypic raft model system. The observed hyperplasia involved a delay in keratinocyte differentiation toward a more undifferentiated state, and expansion of the basal cell compartment was due to increased proliferation, but not apoptosis. These phenotypes were accompanied by elevated p63 expression in the absence of p53 destabilization. In further support of bona fide oncogenic DEK activities, we report here up-regulated DEK protein levels in both human papilloma virus-positive hyperplastic murine skin and a subset of human squamous cell carcinomas. We suggest that DEK up-regulation may contribute to carcinoma development at least in part through increased proliferation and retardation of differentiation.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/virology
- Cell Differentiation/genetics
- Cell Proliferation
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cells, Cultured
- Chromosomal Proteins, Non-Histone/biosynthesis
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- DNA-Binding Proteins/metabolism
- Epithelial Cells/cytology
- Epithelial Cells/pathology
- Epithelial Cells/virology
- Epithelium/metabolism
- Epithelium/pathology
- Fluorescent Antibody Technique
- Foreskin/cytology
- Gene Expression
- Humans
- Hyperplasia/genetics
- Hyperplasia/metabolism
- Hyperplasia/virology
- Keratinocytes/cytology
- Keratinocytes/pathology
- Keratinocytes/virology
- Male
- Membrane Proteins/metabolism
- Mice
- Mice, Transgenic
- Oncogene Proteins/biosynthesis
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Papillomavirus E7 Proteins/genetics
- Poly-ADP-Ribose Binding Proteins
- Proto-Oncogene Mas
- Reverse Transcriptase Polymerase Chain Reaction
- Up-Regulation
Collapse
Affiliation(s)
- Trisha M Wise-Draper
- Division of Pediatric Hematology/Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Abstract
The purpose of the present paper was to investigate the significance of DEK protein expression in uterine cervical lesions and its relationship with HPV infection status. DEK protein expression was studied in 253 cervical lesions, including 30 non-neoplastic cervix with or without squamous metaplasia, 64 cervical intra-epithelial neoplasias (CIN; CIN-1, n = 28; CIN-2, n = 17; CIN-3, n = 19), 102 squamous cell carcinomas (SCC), 51 adenocarcinomas, and six adenosquamous cell carcinomas (adenoSCC) on immunohistochemistry. For comparison, HPV-positive and -negative cervical cancer cell lines were also included. The HPV screening was performed using TaKaRa polymerase chain reaction. On immunohistochemistry DEK was found to be negative in all 30 non-neoplastic cervical epithelia, but it was positive in 96.1% of SCC (98/102), 92.2% of adenocarcinomas (47/51), 100% of adenoSCC (6/6), 85.7% of CIN-1 (24/28), 94.1% of CIN-2 (16/17), and 89.5% of CIN-3 (17/19). There was no significant difference between HPV-positive and -negative cervical lesions. Also, strongly positive staining was observed in all aforementioned cervical cancer cell lines regardless of HPV infection, according to immunocytochemistry. In summary, DEK plays an important role in the carcinogenesis of cervical cancers, and can be helpful for early diagnosis, and is a potential therapeutic target.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adenocarcinoma/virology
- Biomarkers, Tumor/metabolism
- Carcinoma, Adenosquamous/metabolism
- Carcinoma, Adenosquamous/pathology
- Carcinoma, Adenosquamous/virology
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/virology
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- DNA, Viral/analysis
- Female
- HeLa Cells
- Humans
- Oncogene Proteins/genetics
- Oncogene Proteins/metabolism
- Papillomaviridae/genetics
- Papillomaviridae/isolation & purification
- Papillomavirus Infections/genetics
- Papillomavirus Infections/metabolism
- Papillomavirus Infections/virology
- Poly-ADP-Ribose Binding Proteins
- Polymerase Chain Reaction
- Uterine Cervical Neoplasms/metabolism
- Uterine Cervical Neoplasms/pathology
- Uterine Cervical Neoplasms/virology
- Uterine Cervical Dysplasia/metabolism
- Uterine Cervical Dysplasia/pathology
- Uterine Cervical Dysplasia/virology
Collapse
Affiliation(s)
- Qunying Wu
- Department of Pathology, Yanbian University College of Medicine, Yanji, China
| | | | | | | | | | | |
Collapse
|
68
|
Kappes F, Fahrer J, Khodadoust MS, Tabbert A, Strasser C, Mor-Vaknin N, Moreno-Villanueva M, Bürkle A, Markovitz DM, Ferrando-May E. DEK is a poly(ADP-ribose) acceptor in apoptosis and mediates resistance to genotoxic stress. Mol Cell Biol 2008; 28:3245-57. [PMID: 18332104 PMCID: PMC2423161 DOI: 10.1128/mcb.01921-07] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 01/08/2008] [Accepted: 03/04/2008] [Indexed: 12/28/2022] Open
Abstract
DEK is a nuclear phosphoprotein implicated in oncogenesis and autoimmunity and a major component of metazoan chromatin. The intracellular cues that control the binding of DEK to DNA and its pleiotropic functions in DNA- and RNA-dependent processes have remained mainly elusive so far. Our recent finding that the phosphorylation status of DEK is altered during death receptor-mediated apoptosis suggested a potential involvement of DEK in stress signaling. In this study, we show that in cells committed to die, a portion of the cellular DEK pool is extensively posttranslationally modified by phosphorylation and poly(ADP-ribosyl)ation. Through interference with DEK expression, we further show that DEK promotes the repair of DNA lesions and protects cells from genotoxic agents that typically trigger poly(ADP-ribose) polymerase activation. The posttranslational modification of DEK during apoptosis is accompanied by the removal of the protein from chromatin and its release into the extracellular space. Released modified DEK is recognized by autoantibodies present in the synovial fluids of patients affected by juvenile rheumatoid arthritis/juvenile idiopathic arthritis. These findings point to a crucial role of poly(ADP-ribosyl)ation in shaping DEK's autoantigenic properties and in its function as a promoter of cell survival.
Collapse
Affiliation(s)
- F Kappes
- University of Konstanz, Department of Biology, Box X911, D-78457 Konstanz, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Paderova J, Orlic-Milacic M, Yoshimoto M, da Cunha Santos G, Gallie B, Squire JA. Novel 6p rearrangements and recurrent translocation breakpoints in retinoblastoma cell lines identified by spectral karyotyping and mBAND analyses. ACTA ACUST UNITED AC 2008; 179:102-11. [PMID: 18036396 DOI: 10.1016/j.cancergencyto.2007.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Accepted: 08/28/2007] [Indexed: 01/09/2023]
Abstract
Gain of the short arm of chromosome 6, usually through isochromosome 6p formation, is present in approximately 50% of retinoblastoma tumors. The minimal region of gain maps to chromosome band 6p22. Two genes, DEK and E2F3, are implicated as candidate oncogenes. However, chromosomal translocations have been overlooked as a potential mechanism of activation of oncogenes at 6p22 in retinoblastoma. Here, we report combined spectral karyotyping), 4',6-diamidino-2-phenylindole banding, mBAND, and locus-specific fluorescence in situ hybridization analyses of four retinoblastoma cell lines, RB1021, RB247c, RB383, and Y79. In RB1021 and RB247c, 6p undergoes structural rearrangements involving a common translocation breakpoint at 6p22. These data imply that 6p translocations may represent another mechanism of activation of 6p oncogene(s) in a subset of retinoblastomas, besides the copy number increase. In addition to 6p22, other recurrent translocation breakpoints identified in this study are 4p16, 11p15, 17q21.3, and 20q13. Common regions of gain map to chromosomal arms 1q, 2p, 6p, 17q, and 21q.
Collapse
Affiliation(s)
- Jana Paderova
- Department of Applied Molecular Oncology, Ontario Cancer Institute, Princess Maragaret Hospital, 610 University Avenue, Toronto, Ontario, Canada M5G 2M9
| | | | | | | | | | | |
Collapse
|
70
|
Hu HG, Scholten I, Gruss C, Knippers R. The distribution of the DEK protein in mammalian chromatin. Biochem Biophys Res Commun 2007; 358:1008-14. [PMID: 17524367 DOI: 10.1016/j.bbrc.2007.05.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Accepted: 05/02/2007] [Indexed: 11/19/2022]
Abstract
DEK is an abundant and ubiquitous chromatin protein. Here we investigate whether DEK is regularly distributed in the chromatin of human HeLa cells. We show that DEK appears to be excluded from the heterochromatic compartment. However, DEK seems to colocalize with a subfraction of chromatin bearing acetylated histone H4. We examined certain DNA sequences in specifically immunoprecipitated chromatin for four selected human genes. We found that most of the investigated gene sequences were moderately enriched in immunoprecipitated chromatin. In contrast, a promoter-proximal element of the human TOP1 gene was highly enriched in the chromatin immunoprecipitates. This enrichment was lost when cells were treated with alpha-amanitin showing that DEK binds to this particular site only when the TOP1 gene is actively expressed. Our conclusion is that DEK could serve as an architectural protein at the promoter or enhancer sites of a subfraction of human genes.
Collapse
Affiliation(s)
- Hong-gang Hu
- Department of Biology, University of Konstanz, D-78457 Konstanz, Germany.
| | | | | | | |
Collapse
|
71
|
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
|
72
|
Vera J, Estanyol JM, Canela N, Llorens F, Agell N, Itarte E, Bachs O, Jaumot M. Proteomic analysis of SET-binding proteins. Proteomics 2007; 7:578-587. [PMID: 17309103 DOI: 10.1002/pmic.200600458] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The protein SET is involved in essential cell processes such as chromatin remodeling, apoptosis and cell cycle progression. It also plays a critical role in cell transformation and tumorogenesis. With the aim to study new SET functions we have developed a system to identify SET-binding proteins by combining affinity chromatography, MS, and functional studies. We prepared SET affinity chromatography columns by coupling the protein to activated Sepharose 4B. The proteins from mouse liver lysates that bind to the SET affinity columns were resolved with 2-DE and identified by MS using a MALDI-TOF. This experimental approach allowed the recognition of a number of SET-binding proteins which have been classified in functional clusters. The identification of four of these proteins (CK2, eIF2alpha, glycogen phosphorylase (GP), and TCP1-beta) was confirmed by Western blotting and their in vivo interactions with SET were demonstrated by immunoprecipitation. Functional experiments revealed that SET is a substrate of CK2 in vitro and that SET interacts with the active form of GP but not with its inactive form. These data confirm this proteomic approach as a useful tool for identifying new protein-protein interactions.
Collapse
Affiliation(s)
- Jorge Vera
- Departament de Biologia Cel·lular i Anatomia Patològica, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Josep M Estanyol
- Unitat de Proteòmica, Serveis Científico-tècnics, Universitat de Barcelona, Spain
| | - Nuria Canela
- Unitat de Proteòmica, Serveis Científico-tècnics, Universitat de Barcelona, Spain
| | - Franc Llorens
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Neus Agell
- Departament de Biologia Cel·lular i Anatomia Patològica, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Emilio Itarte
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Oriol Bachs
- Departament de Biologia Cel·lular i Anatomia Patològica, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Montserrat Jaumot
- Departament de Biologia Cel·lular i Anatomia Patològica, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
73
|
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: 76] [Impact Index Per Article: 4.0] [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
|
74
|
Tabbert A, Kappes F, Knippers R, Kellermann J, Lottspeich F, Ferrando-May E. Hypophosphorylation of the architectural chromatin protein DEK in death-receptor-induced apoptosis revealed by the isotope coded protein label proteomic platform. Proteomics 2006; 6:5758-72. [PMID: 17001602 DOI: 10.1002/pmic.200600197] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
During apoptosis nuclear morphology changes dramatically due to alterations of chromatin architecture and cleavage of structural nuclear proteins. To characterize early events in apoptotic nuclear dismantling we have performed a proteomic study of apoptotic nuclei. To this end we have combined a cell-free apoptosis system with a proteomic platform based on the differential isotopic labeling of primary amines with N-nicotinoyloxy-succinimide. We exploited the ability of this system to produce nuclei arrested at different stages of apoptosis to analyze proteome alterations which occur prior to or at a low level of caspase activation. We show that the majority of proteins affected at the onset of apoptosis are involved in chromatin architecture and RNA metabolism. Among them is DEK, an architectural chromatin protein which is linked to autoimmune disorders. The proteomic analysis points to the occurrence of multiple PTMs in early apoptotic nuclei. This is confirmed by showing that the level of phosphorylation of DEK is decreased following apoptosis induction. These results suggest the unexpected existence of an early crosstalk between cytoplasm and nucleus during apoptosis. They further establish a previously unrecognized link between DEK and cell death, which will prove useful in the elucidation of the physiological function of this protein.
Collapse
Affiliation(s)
- Anja Tabbert
- University of Konstanz, Molecular Toxicology Group, Konstanz, Germany
| | | | | | | | | | | |
Collapse
|
75
|
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: 105] [Impact Index Per Article: 5.5] [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
|
76
|
Kress TL, Guthrie C. Molecular biology. Accurate RNA siting and splicing gets help from a DEK-hand. Science 2006; 312:1886-7. [PMID: 16809518 DOI: 10.1126/science.1130324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Tracy L Kress
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143-0448, USA
| | | |
Collapse
|
77
|
Soares LMM, Zanier K, Mackereth C, Sattler M, Valcárcel J. Intron removal requires proofreading of U2AF/3' splice site recognition by DEK. Science 2006; 312:1961-5. [PMID: 16809543 DOI: 10.1126/science.1128659] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Discrimination between splice sites and similar, nonsplice sequences is essential for correct intron removal and messenger RNA formation in eukaryotes. The 65- and 35-kD subunits of the splicing factor U2AF, U2AF65 and U2AF35, recognize, respectively, the pyrimidine-rich tract and the conserved terminal AG present at metazoan 3' splice sites. We report that DEK, a chromatin- and RNA-associated protein mutated or overexpressed in certain cancers, enforces 3' splice site discrimination by U2AF. DEK phosphorylated at serines 19 and 32 associates with U2AF35, facilitates the U2AF35-AG interaction and prevents binding of U2AF65 to pyrimidine tracts not followed by AG. DEK and its phosphorylation are required for intron removal, but not for splicing complex assembly, which indicates that proofreading of early 3' splice site recognition influences catalytic activation of the spliceosome.
Collapse
|
78
|
Sarno S, Ruzzene M, Frascella P, Pagano MA, Meggio F, Zambon A, Mazzorana M, Di Maira G, Lucchini V, Pinna LA. Development and exploitation of CK2 inhibitors. Mol Cell Biochem 2006; 274:69-76. [PMID: 16335530 DOI: 10.1007/s11010-005-3079-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A number of quite specific and fairly potent inhibitors of protein kinase CK2, belonging to the classes of condensed polyphenolic compounds, tetrabromobenzimidazole/triazole derivatives and indoloquinazolines are available to date. The structural basis for their selectivity is provided by a hydrophobic pocket adjacent to the ATP/GTP binding site, which in CK2 is smaller than in the majority of other protein kinases due to the presence of a number of residues whose bulky side chains are generally replaced by smaller ones. Consequently a doubly substituted CK2 mutant V66A,I174A is much less sensitive than CK2 wild type to these classes of inhibitors. The most efficient inhibitors both in terms of potency and selectivity are 4,5,6,7-tetrabromo-1H-benzotriazole, TBB (Ki = 0.4 microM), the TBB derivative 2-dimethylamino-4,5,6,7-tetrabromo-1H-benzimidazole, DMAT (Ki = 0.040 microM), the emodin related coumarinic compound 8-hydroxy-4-methyl-9-nitrobenzo[g]chromen-2-one, NBC (Ki = 0.22 microM) and the indoloquinazoline derivative ([5-oxo-5,6-dihydroindolo-(1,2a)quinazolin-7-yl]acetic acid), IQA (Ki = 0.17 microM). These inhibitors are cell permeable as judged from ability to block CK2 in living cells and they have been successfully employed, either alone or in combination with CK2 mutants refractory to inhibition, to dissect signaling pathways affected by CK2 and to identify the endogenous substrates of this pleitropic kinase. By blocking CK2 these inhibitors display a remarkable pro-apoptotic efficacy on a number of tumor derived cell lines, a property which can be exploited in perspective to develop antineoplastic drugs.
Collapse
Affiliation(s)
- Stefania Sarno
- Dipartimento di Chimica Biologica, Universita' di Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Jonker HRA, Wechselberger RW, Pinkse M, Kaptein R, Folkers GE. Gradual phosphorylation regulates PC4 coactivator function. FEBS J 2006; 273:1430-44. [PMID: 16689930 DOI: 10.1111/j.1742-4658.2006.05165.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The unstructured N-terminal domain of the transcriptional cofactor PC4 contains multiple phosphorylation sites that regulate activity. The phosphorylation status differentially influences the various biochemical functions performed by the structured core of PC4. Binding to ssDNA is slightly enhanced by phosphorylation of one serine residue, which is not augmented by further phosphorylation. The presence of at least two phosphoserines decreases DNA-unwinding activity and abrogates binding to the transcriptional activator VP16. Phosphorylation gradually decreases the binding affinity for dsDNA. These phosphorylation-dependent changes in PC4 activities correlate with the sequential functions PC4 fulfils throughout the transcription cycle. MS and NMR revealed that up to eight serines are progressively phosphorylated towards the N-terminus, resulting in gradual environmental changes in the C-terminal direction of the following lysine-rich region. Also within the structured core, primarily around the interaction surfaces, environmental changes are observed. We propose a model for co-ordinated changes in PC4 cofactor functions, mediated by phosphorylation status-dependent gradual masking of the lysine-rich region causing shielding or exposure of interaction surfaces.
Collapse
Affiliation(s)
- Hendrik R A Jonker
- Department of NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, the Netherlands
| | | | | | | | | |
Collapse
|
80
|
Gordon S, Akopyan G, Garban H, Bonavida B. Transcription factor YY1: structure, function, and therapeutic implications in cancer biology. Oncogene 2006; 25:1125-42. [PMID: 16314846 DOI: 10.1038/sj.onc.1209080] [Citation(s) in RCA: 580] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ubiquitous transcription factor Yin Yang 1 (YY1) is known to have a fundamental role in normal biologic processes such as embryogenesis, differentiation, replication, and cellular proliferation. YY1 exerts its effects on genes involved in these processes via its ability to initiate, activate, or repress transcription depending upon the context in which it binds. Mechanisms of action include direct activation or repression, indirect activation or repression via cofactor recruitment, or activation or repression by disruption of binding sites or conformational DNA changes. YY1 activity is regulated by transcription factors and cytoplasmic proteins that have been shown to abrogate or completely inhibit YY1-mediated activation or repression; however, these mechanisms have not yet been fully elucidated. Since expression and function of YY1 are known to be intimately associated with progression through phases of the cell cycle, the physiologic significance of YY1 activity has recently been applied to models of tumor biology. The majority of the data are consistent with the hypothesis that YY1 overexpression and/or activation is associated with unchecked cellular proliferation, resistance to apoptotic stimuli, tumorigenesis and metastatic potential. Studies involving hematopoetic tumors, epithelial-based tumors, endocrine organ malignancies, hepatocellular carcinoma, and retinoblastoma support this hypothesis. Molecular mechanisms that have been investigated include YY1-mediated downregulation of p53 activity, interference with poly-ADP-ribose polymerase, alteration in c-myc and nuclear factor-kappa B (NF-kappaB) expression, regulation of death genes and gene products, and differential YY1 binding in the presence of inflammatory mediators. Further, recent findings implicate YY1 in the regulation of tumor cell resistance to chemotherapeutics and immune-mediated apoptotic stimuli. Taken together, these findings provide strong support of the hypothesis that YY1, in addition to its regulatory roles in normal biologic processes, may possess the potential to act as an initiator of tumorigenesis and may thus serve as both a diagnostic and prognostic tumor marker; furthermore, it may provide an effective target for antitumor chemotherapy and/or immunotherapy.
Collapse
Affiliation(s)
- S Gordon
- Department of Surgery Division of Transplantation, Dumont-UCLA Transplant Center, University of California, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
81
|
Samaniego R, Jeong SY, de la Torre C, Meier I, Moreno Díaz de la Espina S. CK2 phosphorylation weakens 90 kDa MFP1 association to the nuclear matrix in Allium cepa. JOURNAL OF EXPERIMENTAL BOTANY 2006; 57:113-24. [PMID: 16291799 DOI: 10.1093/jxb/erj010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
MFP1 is a conserved plant coiled-coil protein located on the stroma side of the chloroplast thylakoids, as well as in the nuclear matrix. It displays species-specific variability in the number of genes, proteins, and expression. Allium cepa has two nuclear proteins antigenically related to MFP1 with different M(r), pI, distribution, and expression, but only the 90 kDa MFP1 protein is a nuclear matrix component that associates with both the nucleoskeletal filaments and a new category of nuclear bodies. The 90 kDa AcMFP1 migrates in two-dimensional blots as two sets of spots. The hypo-phosphorylated forms (pI approximately 9.5) are tightly bound to the nuclear matrix, while high ionic strength buffers release the more acidic hyper-phosphorylated ones (pI approximately 8.5), suggesting that the protein is post-translationally modified, and that these modifications control its attachment to the nuclear matrix. Dephosphorylation by exogenous alkaline phosphatase and phosphorylation by exogenous CK2, as well as specific inhibition and stimulation of endogenous CK2 with heparin and spermine and spermidine, respectively, revealed that the protein is an in vitro and in vivo substrate of this enzyme, and that CK2 phosphorylation weakens the strength of its binding to the nuclear matrix. In synchronized cells, the nuclear 90 kDa AcMFP1 phosphorylation levels vary during the cell cycle with a moderate peak in G2. These results provide the first evidence for AcMFP1 in vivo phosphorylation, and open up further research on its nuclear functions.
Collapse
Affiliation(s)
- Rafael Samaniego
- Nuclear Matrix Laboratory, Centro de Investigaciones Biológicas, CSIC, 28040-Madrid, Spain
| | | | | | | | | |
Collapse
|
82
|
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: 99] [Impact Index Per Article: 5.0] [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
|
83
|
Grasemann C, Gratias S, Stephan H, Schüler A, Schramm A, Klein-Hitpass L, Rieder H, Schneider S, Kappes F, Eggert A, Lohmann DR. Gains and overexpression identify DEK and E2F3 as targets of chromosome 6p gains in retinoblastoma. Oncogene 2005; 24:6441-9. [PMID: 16007192 DOI: 10.1038/sj.onc.1208792] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2005] [Revised: 04/18/2005] [Accepted: 04/22/2005] [Indexed: 11/09/2022]
Abstract
The paediatric eye tumour retinoblastoma is initiated by inactivation of RB1, a tumour suppressor on chromosome 13q. In addition to RB1 loss, many retinoblastomas show other genetic alterations including gains on chromosomes 6p21-pter and 1q31-q32. Recently, the minimal region of gains on chromosome 6 was narrowed to band p22. We examined genomic gains and expression changes in primary retinoblastomas to identify potential target genes in 6p22. Quantitative multiplex PCR detected copy numbers > or = 3 in 25 (33%) tumours and no gains in 31 of 76 (40%) tumours. The remaining 20 (26%) samples showed gains only at some loci, most often including E2F3 and DEK in 6p22.3. Analysis of RNA from 21 primary retinoblastomas showed that expression levels of these and some other genes in 6p22 correspond to DNA gains. However, KIF 13A, a reported candidate oncogene on 6p, was expressed at low levels or absent. Clinical manifestation of tumours with gains at all 6p22 loci was distinct in that distribution of age at diagnosis was markedly shifted to older age compared to tumours with no or partial gains. In summary, our results suggest that DEK and E2F3 are potential targets of 6p gains in retinoblastoma.
Collapse
Affiliation(s)
- Corinna Grasemann
- University Children's Hospital of Essen, Universitätsklinikum Essen, D-45122 Essen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Sarno S, Salvi M, Battistutta R, Zanotti G, Pinna LA. Features and potentials of ATP-site directed CK2 inhibitors. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2005; 1754:263-70. [PMID: 16198160 DOI: 10.1016/j.bbapap.2005.07.043] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2005] [Revised: 07/18/2005] [Accepted: 07/20/2005] [Indexed: 11/22/2022]
Abstract
A panel of quite specific, fairly potent and cell-permeable inhibitors of protein kinase CK2 belonging to the classes of condensed polyphenolic compounds, tetrabromobenzimidazole/triazole derivatives and indoloquinazolines have been developed, with K(i) values in the submicromolar range. Nine structures have been solved to date of complexes between the catalytic alpha subunit of CK2 and a number of these compounds, many of which display a remarkable specificity toward CK2 as compared to a panel of >30 kinases tested. The structural basis for such selectivity appears to reside in the shape and size of a hydrophobic pocket adjacent to the ATP binding site where these ATP competitive ligands are entrapped mainly by van der Waals interactions and by an energy contribution derived from the hydrophobic effect. In CK2, this cavity is smaller than in the majority of other protein kinases due to a number of unique bulky apolar residues. Consequently, the replacement of two of these residues (V66 and I174) in human CK2 alpha with alanines gives rise to mutants, which are markedly less susceptible than wild type to these classes of inhibitors. Cell-permeable CK2 inhibitors have been successfully employed, either alone or in combination with CK2 mutants refractory to inhibition, to dissect signalling pathways affected by CK2 and/or to validate the identification of in vivo targets of this pleiotropic kinase. Moreover, the remarkable pro-apoptotic efficacy of these compounds toward cell lines derived from a wide spectrum of tumors, disclose the possibility that in perspective CK2 inhibitors might become leads for the development of anti-cancer drugs.
Collapse
Affiliation(s)
- Stefania Sarno
- Department of Biological Chemistry, University of Padova, Viale G. Colombo 3, 35129 Padova, Italy
| | | | | | | | | |
Collapse
|
85
|
Abstract
Protein kinase CK2 has diverse links to gene control and cell cycle. Comparative genome-wide expression profiling of CK2 mutants of the budding yeast Saccharomyces cerevisiae at cell cycle entry has revealed that a significant proportion of cell-cycle genes are affected by CK2. Here, we examine how CK2 realizes this effect. We show that the CK2 action may be directed to gene promoters causing genes with promoter homologies to respond comparably to CK2 perturbation. Examples are metabolic pathway and nutrition supply genes such as the PHO and MET regulon genes, responsible for phosphate maintenance and methionine biosynthesis, respectively. CK2 perturbation affects both regulons permanently and both via repression of a central transcription factor, but with different mechanisms: In the PHO regulon, the gene encoding the central transcription factor Pho4 is repressed and, in addition, Pho4 and/or the cyclin-dependent kinase of the regulon's control complex may be affected by CK2 phosphorylation. In the MET regulon, the repression of the central transcription factor Met4 occurs not by expression inhibition, but rather by availability tuning via a CK2-mediated phosphorylation of a degradation complex. On the other hand, the CK2 action may be directed to the chromatin regulon, thus affecting globally the expression of genes, i.e., the CK2 perturbation results either in comparable responses of genes which have no promoter homologies or in deviating responses despite promoter homologies. The effect is rather transient and concerns aside various cell cycle control genes a notable number of genes encoding chromatin remodeling and modification proteins with functions in chromatin assembly and (anti-)silencing as well as in histone (de-)acetylation, and frequently are also substrates of CK2, suggesting additional tuning at protein level. In line with these findings, we observe in human cells sequence-independent but cell-cycle-dependent CK2 associations with promoters of cell-cycle-regulated genes at periods of extensive gene expression alterations, including cell cycle entry. Our observations are compatible with the idea that the gene control by CK2 is achieved via different mechanisms and at different levels of organization and includes a global role in transcription-related chromatin remodelling and modification.
Collapse
Affiliation(s)
- Walter Pyerin
- Biochemische Zellphysiologie (A135), Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | | | | |
Collapse
|
86
|
Böhm F, Kappes F, Scholten I, Richter N, Matsuo H, Knippers R, Waldmann T. The SAF-box domain of chromatin protein DEK. Nucleic Acids Res 2005; 33:1101-10. [PMID: 15722484 PMCID: PMC549417 DOI: 10.1093/nar/gki258] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2004] [Revised: 01/31/2005] [Accepted: 01/31/2005] [Indexed: 01/16/2023] Open
Abstract
DEK is an abundant chromatin protein in metazoans reaching copy numbers of several millions/nucleus. Previous work has shown that human DEK, a protein of 375 amino acids, has two functional DNA-binding domains, of which one resides in a central part of the molecule and contains sequences corresponding to the scaffold attachment factor-box (SAF-box) domain as found in a growing number of nuclear proteins. Isolated SAF-box peptides (amino acids 137-187) bind weakly to DNA in solution, but when many SAF-box peptides are brought into close proximity on the surface of Sephadex beads, cooperative effects lead to a high affinity to DNA. Furthermore, a peptide (amino acids 87-187) that includes a sequence on the N-terminal side of the SAF-box binds efficiently to DNA. This peptide prefers four-way junction DNA over straight DNA and induces supercoils in relaxed circular DNA just like the full-length DEK. Interestingly, however, the 87-187 amino acid peptide introduces negative supercoils in contrast to the full-length DEK, which is known to introduce positive supercoils. We found that two adjacent regions (amino acids 68-87 and 187-250) are necessary for the formation of positive supercoils. Our data contribute to the ongoing characterization of the abundant and ubiquitous DEK chromatin protein.
Collapse
Affiliation(s)
- Friederike Böhm
- Department of Biology, University of Konstanz78457 Konstanz, Germany
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolis, MN 55455, USA
| | - Ferdinand Kappes
- Department of Biology, University of Konstanz78457 Konstanz, Germany
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolis, MN 55455, USA
| | - Ingo Scholten
- Department of Biology, University of Konstanz78457 Konstanz, Germany
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolis, MN 55455, USA
| | - Nicole Richter
- Department of Biology, University of Konstanz78457 Konstanz, Germany
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolis, MN 55455, USA
| | - Hiroshi Matsuo
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolis, MN 55455, USA
| | - Rolf Knippers
- Department of Biology, University of Konstanz78457 Konstanz, Germany
- Department of Biochemistry, Molecular Biology and Biophysics, University of MinnesotaMinneapolis, MN 55455, USA
| | - Tanja Waldmann
- To whom correspondence should be addressed at Max Planck Institute for Immunology Stübeweg 51, D-79108 Freiburg, Germany. Tel: +49 761 5108377; Fax: +49 761 5108220;
| |
Collapse
|
87
|
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: 98] [Impact Index Per Article: 4.7] [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
|
88
|
Malik P, Clements JB. Protein kinase CK2 phosphorylation regulates the interaction of Kaposi's sarcoma-associated herpesvirus regulatory protein ORF57 with its multifunctional partner hnRNP K. Nucleic Acids Res 2004; 32:5553-69. [PMID: 15486205 PMCID: PMC524287 DOI: 10.1093/nar/gkh876] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
ORF57 protein of Kaposi's sarcoma-associated herpesvirus has a counterpart in all herpesvirus of mammals and birds and regulates gene expression at transcriptional and post-transcriptional levels. ORF57 was capable of self-interaction and bound a rapidly migrating form of heterogeneous nuclear ribonucleoprotein K (hnRNP K), a multifunctional cellular protein involved in gene expression. In virus infected cell extracts, ORF57 was present in a complex with hnRNP K that had protein kinase CK2 activity, and was phosphorylated by CK2. Different regions of ORF57 bound both catalytic alpha/alpha' and regulatory beta subunits of CK2. CK2 modification enhanced the ORF57-hnRNP K interaction, and may regulate the presence and activities of components in the complex. We suggest that ORF57 and hnRNP K interaction may modulate ORF57-mediated regulation of viral gene expression. Herpesviral ORF57 (Rhadinovirus) and ICP27 (Simplexvirus) proteins both interact with hnRNP K and CK2 implying that adaptation of the ancestral hnRNP K and CK2 to associate with viral regulatory ancestor protein likely pre-dates divergence of these Herpesviridae genera that occurred 200 million years ago.
Collapse
Affiliation(s)
- Poonam Malik
- Division of Virology, Institute of Biomedical and Life Sciences, University of Glasgow, Church Street, Glasgow, G11 5JR, Scotland, UK
| | | |
Collapse
|
89
|
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: 72] [Impact Index Per Article: 3.4] [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
|