Physical and Functional Interaction Between WT1 and P53 Proteins SHYAMALA MAHESWARAN*, SEON PARK*, AMY BERNARD*, JENNIFER F

Total Page:16

File Type:pdf, Size:1020Kb

Physical and Functional Interaction Between WT1 and P53 Proteins SHYAMALA MAHESWARAN*, SEON PARK*, AMY BERNARD*, JENNIFER F Proc. Natl. Acad. Sci. USA Vol. 90, pp. 5100-5104, June 1993 Genetics Physical and functional interaction between WT1 and p53 proteins SHYAMALA MAHESWARAN*, SEON PARK*, AMY BERNARD*, JENNIFER F. MoRRISt, FRANK J. RAUSCHER IIlt, DAVID E. HILLY, AND DANIEL A. HABER*§ *Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA 02129; tThe Wistar Institute, Philadelphia, PA 19104; and tOncogene Science, Inc., Cambridge, MA 02139 Communicated by Kurt J. Isselbacher, February 22, 1993 ABSTRACT WTI is a tumor-suppressor gene expressed in rabbit polyclonal antibodies directed against overlapping the developing kidney, whose inactivation leads to the devel- N-terminal peptides, aa 1-173 and 85-173, respectively (9); opment of Wilms tumor, a pediatric kidney cancer. WTI DG10 is a monoclonal antibody generated against human encodes a transcription factor which binds to the EGRI con- WT1 synthesized in Escherichia coli. For immunoprecipita- sensus sequence, mediating transcriptional repression. We now tions, cells were labeled with [35S]methionine and extracted demonstrate that p53, the product of a tumor-suppressor gene with either ELB buffer (50 mM Hepes, pH 7.0/250 mM with ubiquitous expression, physically associates with WT1 in NaCl/0.5 mM EDTA/0.1% Nonidet P-40) or RIPA buffer (10 transfected cells. The interaction between WT1 and p53 mod- mM Tris, pH 7.4/150 mM NaCl, 1% Triton X-100/1% sodium deoxycholate/0.1% SDS). For sequential immunoprecipita- ulates their ability to transactivate their respective targets. In tions, cellular lysates were extracted with ELB buffer and the absence of p53, WT1 acts as a potent transcriptional immunoprecipitated with the first antibody. The immune activator of the early growth response gene 1 (EGRI) site, complex was then dissociated in RIPA buffer and the re- rather than a transcriptional repressor. In contrast, WT1 leased proteins were immunoprecipitated with the second exerts a cooperative effect on p53, enhancing its ability to antibody. Peptide maps ofimmunoprecipitated WT1 proteins transactivate the muscle creatine kinase promoter. were generated using various concentrations ofStaphylococ- cus aureus V8 protease (10). For immunoprecipitation/ Wilms tumor has been linked to the inactivation of the WTI Western analysis, anti-p53 antibodies were covalently tumor-suppressor gene at the 11p13 chromosomal locus crosslinked to protein A-Sepharose (10), and after immuno- (reviewed in ref. 1). WTI encodes a developmentally regu- precipitation, proteins were released from the antibody by lated transcription factor of 52-54 kDa. The C terminus incubation with 2% SDS/50 mM Tris, pH 6.8, at room contains four zinc fingers which confer binding specificity to temperature for 5 min. Western blot analysis was performed the EGRI DNA consensus (2), while the N terminus mediates according to the ECL protocol (Amersham), using antibody transcriptional repression in transient transfection assays (3, WT-6F1 (9) at a dilution of 1:2000. To crosslink proteins in 4). A complex pattern of alternative splicing leads to distinct cells, cultures were incubated with 3 mM dithiobis(succin- WTI gene products (5). Alternative splice II, which encodes imidyl propionate) (DSP, Pierce; 100-mg/ml stock in dimeth- three amino acids [Lys-Thr-Ser (KTS)], interrupts the spac- yl sulfoxide) for 30 min, DSP was inactivated with 30 mM ing between the third and fourth zinc fingers, altering the ammonium acetate (pH 7.0), and protein was extracted. For DNA-binding specificity of WT1 protein (2, 6). WTI muta- two-dimensional gel electrophoresis, proteins were cross- tions detected in Wilms tumor specimens have also provided linked with DSP, immunoprecipitated, and resolved by SDS/ reagents to dissect the functional properties of WT1 protein. 3-10% PAGE. The lanes were excised, the crosslinking was Of particular interest is a dominant negative mutation, reversed by incubation in 5% 2-mercaptoethanol/10%o glyc- WTAR (7), which encodes an in-frame deletion of the third erol/2.3% SDS, 62.5 mM Tris, pH 6.8, and the proteins were zinc finger and demonstrates oncogenic potential in baby rat electrophoresed in the second dimension. For gel filtration, kidney (BRK) cell transformation assays (8). Disruption of ELB extracts from unlabeled ceils were resolved on a Su- the DNA-binding domain by the WTAR mutation suggests perose 12 FPLC column (Pharmacia); fractions were con- that its dominant effect may result from interactions with centrated by ethanol precipitation and analyzed by Western other cellular proteins. blotting. Using stable BRK cell lines immortalized by transfection Chloramphenicol Acetyltransferase (CAT) Assays. Cultures with the adenovirus EIA gene along with WTI, we demon- were transfected by calcium phosphate/DNA precipitation strate the presence of a complex containing WT1 and p53 with expression constructs under the control ofthe cytomeg- proteins. This complex is also observed in BRK cells ex- alovirus promoter, and the total amount of promoter se- pressing mutant WTAR and a mutated p53 gene (codon 248), quence transfected into each dish was equalized by the as well as in specimens of sporadic Wilms tumor. A potential addition ofvector DNA. Transfection efficiencies were stan- functional interaction between WT1 and p53 is suggested by dardized by cotransfection of a human growth hormone transactivation assays using their respective target se- reporter construct, and all experiments were repeated at least quences. While WT1 enhances transcriptional activation by three times. CAT activity was quantitated by excising the p53, wild-type p53 appears to convert WT1 from a transcrip- appropriate sections ofthe TLC plates for scintillation count- tional activator to a transcriptional repressor. ing. MATERIALS AND METHODS RESULTS Coimmunoprecipitation of WT1 and p53 in Transfected Immunoprecipitations and Western Blot Analyses. Three BRK Cells. To study WT1-associated proteins in an appro- anti-WT1 antibodies were used: WT-6F1 and WT-91 are Abbreviations: BRK, baby rat kidney; DSP, dithiobis(succinimidyl The publication costs ofthis article were defrayed in part by page charge propionate); MCK, muscle creatine kinase; CAT, chloramphenicol payment. This article must therefore be hereby marked "advertisement" acetyltransferase. in accordance with 18 U.S.C. §1734 solely to indicate this fact. §To whom reprint requests should be addressed. 5100 Downloaded by guest on September 26, 2021 Genetics: Maheswaran et al. Proc. Natl. Acad. Sci. USA 90 (1993) 5101 a 1st Ab Nor.specifir aWf-91 priate kidney-derived cell type, we made use of EIA- immortalized BRK cell lines, stably transfected with con- N c\ C\j structs encoding either wild-type WT1 (clone A6) or mutant WTAR (clone 6.3) (8). Immunoprecipitation of WT1 from 2nd Ab C L BRK cells under nonionic detergent conditions (ELB buffer) ~ Q resulted in the coprecipitation of a protein comigrating with M p53, while immunoprecipitation of p53 coprecipitated a band kDa that comigrated with WT1. Coprecipitation of these proteins was reduced by extraction under more stringent conditions 66- (RIPA buffer), consistent with an unstable protein associa- .- 5 tion. To determine whether WT1 and p53 were in fact coimmunoprecipitated, we performed sequential immuno- precipitations with antibodies directed against these two proteins. We first used 6.3 cells, expressing transfected 45 - WTAR and a mutated endogenous p53 (with an Arg His substitution in codon 246 of the rat p53 gene, analogous to human codon 248). Radiolabeled, ELB-extracted lysates were immunoprecipitated with anti-WT1 antibody WT-91, a p53 a p53 and the immune complex was dissociated in RIPA buffer 1st Ab Nonspecific PAB 122 PAS 122 before immunoprecipitation with anti-p53 antibody PAb421 or PAb122. Fig. la shows the precipitation of a protein, to to .-Q released from the WT1 immune complex, that comigrated 9D- 2nd Ab a an C-1 with authentic p53. Substitution of nonspecific antibodies of a: 33 4 + the same isotype for either the anti-WT1 or the anti-p53 kDa antibody abolished precipitation of p53. In the reciprocal experiment, immunoprecipitation of extracts with anti-p53 antibody PAbl22, dissociation of the immune complex, and 66 - immunoprecipitation with either of two anti-WT1 antibodies (WT-91 and DG10) precipitated a protein that comigrated - WTAR --- with authentic WTAR (Fig. lb). Use of nonspecific antibod- 45 - ies or preincubation of DG10 with unlabeled vaccinia- produced WT1 protein abolished the immunoprecipitation of WTAR from the p53 immune complex (Fig. lb). Sequential- immunoprecipitation experiments also demonstrated the co- precipitation of p53 and wild-type WT1 from extracts of A6 cells. Partial V8 protease digestion of WT1 either directly C V8 (,u g) 1 5 50 1 5 immunoprecipitated from A6 cells or sequentially immuno- kDa precipitated with anti-p53 followed by anti-WT1 antibodies 45 - confirmed the presence of authentic WT1 within the p53 immune complex (Fig. lc). Immunoprecipitation/Western Blot Analysis of BRK Cells and Wilms Tumor Specimens. We used a second approach to 29 - demonstrate the association between WT1 and p53, consist- ing of immunoprecipitation with the first antibody, followed 21 - by Western blot analysis of the immunoprecipitate with the second antibody. Unlabeled ELB-extracted proteins were 14.3 -. - .: immunoprecipitated with anti-p53 antibody and analyzed by translated p53 or WT1. RIPA buffer was sufficiently stringent to dissociate the WT1/p53 complex without affecting the primary antigen-antibody interaction. (b) Immunoprecipitation with anti-p53 antibody followed by anti-WT1 antibodies. Radiolabeled proteins extracted with ELB buffer from 6.3 cells were first immunoprecip- itated with either PAb122 (lst Ab, lanes 3 and 4) or a nonspecific antibody of the same isotype (lst Ab, lanes 1 and 2). Immune WTI direct ip WTI Seq ip complexes were dissociated and reprecipitated with anti-WT-91 (2nd aWT DGIO ap53 PAbl22 Ab, lanes 2 and 4) or a 10-fold excess of nonspecific rabbit antibody Il (2nd Ab, lanes 1 and 3). WTAR directly immunoprecipitated from 6.3 aWTI DGI10 cells served as a marker (lane M). In a separate experiment, proteins FIG.
Recommended publications
  • Wt1) – an Effector in Leukemogenesis?
    WILMS’ TUMOUR GENE 1 PROTEIN (WT1) – AN EFFECTOR IN LEUKEMOGENESIS? Vidovic, Karina 2010 Link to publication Citation for published version (APA): Vidovic, K. (2010). WILMS’ TUMOUR GENE 1 PROTEIN (WT1) – AN EFFECTOR IN LEUKEMOGENESIS?. Lund University: Faculty of Medicine. Total number of authors: 1 General rights Unless other specific re-use rights are stated the following general rights apply: Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal Read more about Creative commons licenses: https://creativecommons.org/licenses/ Take down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim. LUND UNIVERSITY PO Box 117 221 00 Lund +46 46-222 00 00 Download date: 07. Oct. 2021 Division of Hematology and Transfusion Medicine Lund University, Lund, Sweden WILMS’ TUMOUR GENE 1 PROTEIN (WT1) – AN EFFECTOR IN LEUKEMOGENESIS? Karina Vidovic Thesis 2010 Contact adress Karina Vidovic Division of Hematology and Transfusion Medicine BMC, C14 Klinikgatan 28 SE- 221 84 Lund Sweden Phone +46 46 222 07 30 e-mail: [email protected] ISBN 978-91-86443-99-3 ! Karina Vidovic Printed by Media-Tryck, Lund, Sweden 2 ”The journey of a thousand miles begins with one step” Lao Tzu (Chinese taoist), 600-531 BC 3 4 LIST OF PAPERS This thesis is based on the following papers, referred to in the text by their Roman numerals I.
    [Show full text]
  • Wild-Type P53 Can Down-Modulate the Activity of Various Promoters
    Proc. Natl. Acad. Sci. USA Vol. 88, pp. 9979-9983, November 1991 Biochemistry Wild-type p53 can down-modulate the activity of various promoters DORON GINSBERG*, FATIMA MECHTAt, MOSHE YANIVt, AND MOSHE OREN*t *Department of Chemical Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel; and tDepartment of Biotechnology, Pasteur Institute, 75724 Paris Cedex 15, France Communicated by Robert A. Weinberg, August 16, 1991 ABSTRACT The wild-type (wt) p53 protein is the product out whether it could also modulate c-fos expression. We of a tumor suppressor gene that is a frequent target for report here that wt p53 can inhibit c-fos gene expression. This inactivation in many types of tumors. The nuclear localization effect of wt p53 is rapid, suggesting that it may precede ofthe protein, as well as additional features, suggest that it may growth arrest. In addition, cotransfection experiments indi- be involved in the regulation ofgene expression. To explore this cate that wt p53 can down-modulate the activity of a number possibility, the effects of overproduced wt p53 were investi- of promoters. While the effect of wt p53 appeared to be gated in a number of systems. Induction of growth arrest via relatively nonspecific, it probably did not reflect a general- the antiproliferative effect ofwt p53 greatly impaired the ability ized transcriptional shut-off. Our findings suggest that wt p53 of cells to exhibit an increase in c-fos mRNA upon serum may contribute to growth inhibition by down-modulating, stimulation. Experiments in which cells were cotransfected directly or indirectly, the expression of genes that are re- with p53 expression plasmids together with a reporter gene quired for ongoing cell proliferation.
    [Show full text]
  • An Integrated Genome Screen Identifies the Wnt Signaling Pathway As a Major Target of WT1
    An integrated genome screen identifies the Wnt signaling pathway as a major target of WT1 Marianne K.-H. Kima,b, Thomas J. McGarryc, Pilib O´ Broind, Jared M. Flatowb, Aaron A.-J. Goldend, and Jonathan D. Lichta,b,1 aDivision of Hematology/Oncology and cFeinberg Cardiovascular Research Institute, Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; bRobert H. Lurie Cancer Center, Northwestern University, Chicago, IL 60611; and dDepartment of Information Technology, National University of Ireland, Galway, Republic of Ireland Edited by Peter K. Vogt, The Scripps Research Institute, La Jolla, CA, and approved May 18, 2009 (received for review February 12, 2009) WT1, a critical regulator of kidney development, is a tumor suppressor control. A first generation of in vitro cotransfection and DNA for nephroblastoma but in some contexts functions as an oncogene. binding assays have given way to more biologically relevant exper- A limited number of direct transcriptional targets of WT1 have been iments where manipulation of WT1 levels has been accompanied identified to explain its complex roles in tumorigenesis and organo- by examination of candidate or global gene expression. Classes of genesis. In this study we performed genome-wide screening for direct WT1 targets thus identified consist of inducers of differentiation, WT1 targets, using a combination of ChIP–ChIP and expression arrays. regulators of cell growth and modulators of cell death. WT1 target Promoter regions bound by WT1 were highly G-rich and resembled genes include CDKN1A (22), a negative regulator of the cell cycle; the sites for a number of other widely expressed transcription factors AREG (23), a facilitator of kidney differentiation; WNT4 (24), a such as SP1, MAZ, and ZNF219.
    [Show full text]
  • Original Article Regulation of Exogenous P53 Combined with Dickkopf-1 on Human Osteosarcoma MG-63 Cells
    Int J Clin Exp Med 2017;10(6):9252-9258 www.ijcem.com /ISSN:1940-5901/IJCEM0052476 Original Article Regulation of exogenous P53 combined with Dickkopf-1 on human osteosarcoma MG-63 cells Fuyu Gao, Benjun Bi, Guangjun Liao Department of Orthopedic Surgery, Yantaishan Hospital, Yantai, Shandong, China Received March 8, 2017; Accepted April 22, 2017; Epub June 15, 2017; Published June 30, 2017 Abstract: Objective: To regulate the growth of human osteosarcoma cell strain MG-63 in vitro through the combined drugs of exogenous P53 gene and Dickkopf-1 and to explore its possible mechanisms in order to provide the refer- ence for clinical treatment of osteosarcoma. Methods: Human osteosarcoma MG-63 cell strain was subcultured in vitro, and exogenous P53 and Dickkopf-1 were used separately or jointly to act on MG-63 cells. The proliferation and apoptosis of MG-63 cells were respectively detected by trypan blue exclusion and flow cytometry; the expres- sion levels of tumor suppressor gene P53 and oncogene c-fos and c-myc were detected by real-time fluorescent quantitative and western blot in mRNA and protein levels. Results: The proliferation test results showed that the combination of exogenous P53 and Dickkopf-1 could significantly inhibit the growth of MG-63 cells, compared with the single drug using (P<0.05), and cell cycle assay found that the combined drugs caused the cell cycle arrest at G0/G1 phase (P<0.05). Flow cytometry assay indicated the apoptosis of MG-63 cells caused by the combined drugs was significantly higher than that of using exogenous P53 or Dickkopf-1 alone, and the results of western blot manifested the combined drugs could down-regulate the expression levels of c-fos and c-myc whichwere related to the proliferation and differentiation of MG-63 cells, and increase the expression of tumor suppressor gene P53.
    [Show full text]
  • Carcinogeninduced Hepatic Tumors in KLF6+/ Mice Recapitulate
    HEPATOBILIARY MALIGNANCIES Carcinogen-Induced Hepatic Tumors in KLF61/2 Mice Recapitulate Aggressive Human Hepatocellular Carcinoma Associated with p53 Pathway Deregulation Mirko Tarocchi,1,2 Rebekka Hannivoort,1,3 Yujin Hoshida,4 Ursula E. Lee,1 Diana Vetter,1 Goutham Narla,5,6 Augusto Villanueva,7,8 Moshe Oren,8 Josep M. Llovet,1,7,9 and Scott L. Friedman1 Inactivation of KLF6 is common in hepatocellular carcinoma (HCC) associated with hepa- titis C virus (HCV) infection, thereby abrogating its normal antiproliferative activity in liver cells. The aim of the study was to evaluate the impact of KLF6 depletion on human HCC and experimental hepatocarcinogenesis in vivo. In patients with surgically resected HCC, reduced tumor expression of KLF6 was associated with decreased survival. Consistent with its role as a tumor suppressor, KLF61/2 mice developed significantly more tumors in response to the chemical carcinogen diethyl nitrosamine (DEN) than wild-type animals. Gene expression signatures in both surrounding tissue and tumors of KLF61/2 mice closely recapitulated those associated with aggressive human HCCs. Expression microarray profiling also revealed an increase in Mdm2 mRNA in tumors from KLF61/2 compared with KLF61/1 mice, which was validated by way of quantitative real-time polymerase chain reaction and western blot analysis in both human HCC and DEN-induced murine tumors. Moreover, chromatin immunoprecipitation and cotransfection assays established the P2 intronic promoter of Mdm2 as a bona fide transcriptional target repressed by KLF6. Whereas KLF6 overexpression in HCC cell lines and primary hepatocytes led to reduced MDM2 levels and increased p53 protein and transcriptional activity, reduction in KLF6 by small interfering RNA led to increased MDM2 and reduced p53.
    [Show full text]
  • Teacher Background on P53 Tumor Suppressor Protein
    Cancer Lab p53 – Teacher Background on p53 Tumor Suppressor Protein Note: The Teacher Background Section is meant to provide information for the teacher about the topic and is tied very closely to the PowerPoint slide show. For greater understanding, the teacher may want to play the slide show as he/she reads the background section. For the students, the slide show can be used in its entirety or can be edited as necessary for a given class. What Is p53 and Where Is the Gene Located? While commonly known as p53, the official name of this gene is Tumor Protein p53 and its official symbol is TP53. TheTP53 gene codes for the TP53 (p53) protein which acts as a tumor suppressor and works in response to DNA damage to orchestrate the repair of damaged DNA. If the DNA cannot be repaired, the p53 protein prevents the cell from dividing and signals it to undergo apoptosis (programmed cell death). The name p53 is due to protein’s 53 kilo-Dalton molecular mass. The gene which codes for this protein is located on the short (p) arm of chromosome 17 at position 13.1 (17p13.1). The gene begins at base pair 7,571,719 and ends at base pair 7, 590,862 making it 19,143 base pairs long. (1, 2) What Does the p53 Gene Look Like When Translated Into Protein? The TP53 gene has 11 exons and a very large 10 kb intron between exons 1 and 2. In humans, exon 1 is non-coding and it has been shown that this region could form a stable stem-loop structure which binds tightly to normal p53 but not to mutant p53 proteins.
    [Show full text]
  • Nuclear Localization of DP and E2F Transcription Factors by Heterodimeric Partners and Retinoblastoma Protein Family Members
    Journal of Cell Science 109, 1717-1726 (1996) 1717 Printed in Great Britain © The Company of Biologists Limited 1996 JCS7086 Nuclear localization of DP and E2F transcription factors by heterodimeric partners and retinoblastoma protein family members Junji Magae1, Chin-Lee Wu2, Sharon Illenye1, Ed Harlow2 and Nicholas H. Heintz1,* 1Department of Pathology, University of Vermont, Burlington VT 05405, USA 2Massachusetts General Hospital Cancer Center, Charlestown MA 02129, USA *Author for correspondence SUMMARY E2F is a family of transcription factors implicated in the showed that regions of E2F-1 and DP-1 that are required regulation of genes required for progression through G1 for stable association of the two proteins were also required and entry into the S phase. The transcriptionally active for nuclear localization of DP-1. Unlike E2F-1, -2, and -3, forms of E2F are heterodimers composed of one polypep- E2F-4 did not accumulate in the nucleus unless it was coex- tide encoded by the E2F gene family and one polypeptide pressed with DP-2. p107 and p130, but not pRb, stimulated encoded by the DP gene family. The transcriptional activity nuclear localization of E2F-4, either alone or in combina- of E2F/DP heterodimers is influenced by association with tion with DP-2. These results indicate that DP proteins the members of the retinoblastoma tumor suppressor preferentially associate with specific E2F partners, and protein family (pRb, p107, and p130). Here the intracellu- suggest that the ability of specific E2F/DP heterodimers to lar distribution of E2F and DP proteins was investigated in localize in the nucleus contributes to the regulation of E2F transiently transfected Chinese hamster and human cells.
    [Show full text]
  • TP63 and TP73 in Cancer, an Unresolved В€Œfamily∕ Puzzle Of
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector FEBS Letters 588 (2014) 2590–2599 journal homepage: www.FEBSLetters.org Review TP63 and TP73 in cancer, an unresolved ‘‘family’’ puzzle of complexity, redundancy and hierarchy Antonio Costanzo a, Natalia Pediconi b,c, Alessandra Narcisi a, Francesca Guerrieri c,d, Laura Belloni c,d, ⇑ Francesca Fausti a, Elisabetta Botti a, Massimo Levrero c,d, a Dermatology Unit, Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Italy b Laboratory of Molecular Oncology, Department of Molecular Medicine, Sapienza University of Rome, Italy c Center for Life Nanosciences (CNLS) – IIT/Sapienza, Rome, Italy d Laboratory of Gene Expression, Department of Internal Medicine (DMISM), Sapienza University of Rome, Italy article info abstract Article history: TP53 belongs to a small gene family that includes, in mammals, two additional paralogs, TP63 and Received 19 May 2014 TP73. The p63 and p73 proteins are structurally and functionally similar to p53 and their activity as Revised 16 June 2014 transcription factors is regulated by a wide repertoire of shared and unique post-translational mod- Accepted 16 June 2014 ifications and interactions with regulatory cofactors. p63 and p73 have important functions in Available online 28 June 2014 embryonic development and differentiation but are also involved in tumor suppression. The biology Edited by Shairaz Baksh, Giovanni Blandino of p63 and p73 is complex since both TP63 and TP73 genes are transcribed into a variety of different and Wilhelm Just isoforms that give rise to proteins with antagonistic properties, the TA-isoforms that act as tumor- suppressors and DN-isoforms that behave as proto-oncogenes.
    [Show full text]
  • Jdp2 Downregulates Trp53 Transcription to Promote Leukaemogenesis in the Context of Trp53 Heterozygosity
    Oncogene (2013) 32, 397 --402 & 2013 Macmillan Publishers Limited All rights reserved 0950-9232/13 www.nature.com/onc SHORT COMMUNICATION Jdp2 downregulates Trp53 transcription to promote leukaemogenesis in the context of Trp53 heterozygosity L van der Weyden1, AG Rust1,4, RE McIntyre1,4, CD Robles-Espinoza1, M del Castillo Velasco-Herrera1, R Strogantsev2, AC Ferguson-Smith2, S McCarthy1, TM Keane1, MJ Arends3 and DJ Adams1 We performed a genetic screen in mice to identify candidate genes that are associated with leukaemogenesis in the context of Trp53 heterozygosity. To do this we generated Trp53 heterozygous mice carrying the T2/Onc transposon and SB11 transposase alleles to allow transposon-mediated insertional mutagenesis to occur. From the resulting leukaemias/lymphomas that developed in these mice, we identified nine loci that are potentially associated with tumour formation in the context of Trp53 heterozygosity, including AB041803 and the Jun dimerization protein 2 (Jdp2). We show that Jdp2 transcriptionally regulates the Trp53 promoter, via an atypical AP-1 site, and that Jdp2 expression negatively regulates Trp53 expression levels. This study is the first to identify a genetic mechanism for tumour formation in the context of Trp53 heterozygosity. Oncogene (2013) 32, 397--402; doi:10.1038/onc.2012.56; published online 27 February 2012 Keywords: p53; Jdp2; transposon; heterozygosity; lymphoma; mice INTRODUCTION targeted by transposon insertion events leading to upregulated Genetic alterations of TP53 are frequent events in tumourigenesis Jdp2 expression and a decrease in Trp53 expression levels. Further and promote genomic instability, impair apoptosis, and contribute we illustrate that Jdp2 regulates the Trp53 promoter via an atypical to aberrant self-renewal.1--4 The spectrum of mutations that occur AP-1 binding site.
    [Show full text]
  • RB1 and P53 at the Crossroad of EMT and Triple Negative Breast Cancer
    PERSPECTIVE PERSPECTIVE Cell Cycle 10:10, 1-8; May 15, 2011; © 2011 Landes Bioscience RB1 and p53 at the crossroad of EMT and triple negative breast cancer Zhe Jiang,1 Robert Jones,1 Jeff C. Liu,1 Tao Deng,1 Tyler Robinson,1 Philip E.D. Chung,1 Sharon Wang,1 Jason I. Herschkowitz,2 Sean E. Egan,3 Charles M. Perou4 and Eldad Zacksenhaus1,* 1Division of Cell and Molecular Biology; Toronto General Research Institute; University Health Network; Toronto, Ontario, Canada; 2Department of Molecular and Cellular Biology; Baylor College of Medicine; Houston, TX USA; 3Program in Developmental and Stem Cell Biology; The Hospital for Sick Children; Department of Molecular Genetics; University of Toronto; Toronto, Ontario, Canada; 4Lineberger Comprehensive Cancer Center; Department of Genetics and Pathology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA riple negative breast cancer (TNBC) NEU-positive and Triple Negative (TN) Tis a heterogeneous disease that tumors, the latter of which do not express includes Basal-like and Claudin-low hormone receptors or HER2.1-7 TNBC tumors. The Claudin-low tumors are affects 15–30% of patients. By IHC it can enriched for features associated with be further divided into Basal-like breast epithelial-to-mesenchymal transition cancer and non-basal tumors, some of (EMT) and possibly for tumor initiating which exhibit features of EMT.8 Basal-like cells. Primary TNBCs respond relatively BCs express the basal cytokeratins (CK) well to conventional chemotherapy; CK5/6, CK14, CK17, and/or epidermal © 2011 Landes Bioscience. Landes ©2011 however, metastatic disease is virtually growth factor receptor (EGFR), whereas incurable.
    [Show full text]
  • Crucial Role of C-Jun Phosphorylation at Ser63/73 Mediated by PHLPP Protein Degradation in the Cheliensisin a Inhibition of Cell Transformation
    Published OnlineFirst October 3, 2014; DOI: 10.1158/1940-6207.CAPR-14-0233 Cancer Prevention Research Article Research Crucial Role of c-Jun Phosphorylation at Ser63/73 Mediated by PHLPP Protein Degradation in the Cheliensisin A Inhibition of Cell Transformation Junlan Zhu1,2, Jingjie Zhang1, Haishan Huang1,2, Jingxia Li1, Yonghui Yu1, Honglei Jin1,2, Yang Li1,2, Xu Deng3, Jimin Gao2, Qinshi Zhao3, and Chuanshu Huang1 Abstract Cheliensisin A (Chel A), as a novel styryl-lactone isolated from Goniothalamus cheliensis Hu, has been demonstrated to have an inhibition of EGF-induced Cl41 cell transformation via stabilizing p53 protein in a Chk1-dependent manner, suggesting its chemopreventive activity in our previous studies. However, its underlying molecular mechanisms have not been fully characterized yet. In the current study, we found that Chel A treatment could increase c-Jun protein phosphorylation and activation, whereas the inhibition of c-Jun phosphorylation, by ectopic expression of a dominant-negative mutant of c-Jun, TAM67, reversed the Chel A inhibition of EGF-induced cell transformation and impaired Chel A induction of p53 protein and apoptosis. Moreover, our results indicated that Chel A treatment led to a PHLPP downregulation by promoting PHLPP protein degradation. We also found that PHLPP could interact with and bind to c-Jun protein, whereas ectopic PHLPP expression blocked c-Jun activation, p53 protein and apoptotic induction by Chel A, and further reversed the Chel A inhibition of EGF-induced cell transformation. With the findings, we have demonstrated that Chel A treatment promotes a PHLPP protein degradation, which can bind to c-Jun and mediates c-Jun phosphorylation, and further leading to p53 protein induction, apoptotic responses, subsequently resulting in cell transformation inhibition and chemopreventive activity of Chel A.
    [Show full text]
  • AP-1 in Cell Proliferation and Survival
    Oncogene (2001) 20, 2390 ± 2400 ã 2001 Nature Publishing Group All rights reserved 0950 ± 9232/01 $15.00 www.nature.com/onc AP-1 in cell proliferation and survival Eitan Shaulian1 and Michael Karin*,1 1Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, California, CA 92093-0636, USA A plethora of physiological and pathological stimuli extensively discussed previously (Angel and Karin, induce and activate a group of DNA binding proteins 1991; Karin, 1995). that form AP-1 dimers. These proteins include the Jun, The mammalian AP-1 proteins are homodimers and Fos and ATF subgroups of transcription factors. Recent heterodimers composed of basic region-leucine zipper studies using cells and mice de®cient in individual AP-1 (bZIP) proteins that belong to the Jun (c-Jun, JunB proteins have begun to shed light on their physiological and JunD), Fos (c-Fos, FosB, Fra-1 and Fra-2), Jun functions in the control of cell proliferation, neoplastic dimerization partners (JDP1 and JDP2) and the closely transformation and apoptosis. Above all such studies related activating transcription factors (ATF2, LRF1/ have identi®ed some of the target genes that mediate the ATF3 and B-ATF) subfamilies (reviewed by (Angel eects of AP-1 proteins on cell proliferation and death. and Karin, 1991; Aronheim et al., 1997; Karin et al., There is evidence that AP-1 proteins, mostly those that 1997; Liebermann et al., 1998; Wisdom, 1999). In belong to the Jun group, control cell life and death addition, some of the Maf proteins (v-Maf, c-Maf and through their ability to regulate the expression and Nrl) can heterodimerize with c-Jun or c-Fos (Nishiza- function of cell cycle regulators such as Cyclin D1, p53, wa et al., 1989; Swaroop et al., 1992), whereas other p21cip1/waf1, p19ARF and p16.
    [Show full text]