101
|
Balmer MT, Katz RD, Liao S, Goodwine JS, Gal S. Doxorubicin and 5-fluorouracil induced accumulation and transcriptional activity of p53 are independent of the phosphorylation at serine 15 in MCF-7 breast cancer cells. Cancer Biol Ther 2014; 15:1000-12. [PMID: 24801380 DOI: 10.4161/cbt.29112] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The chemotherapeutic agents doxorubicin (dox) or 5-fluorouracil (5FU) are used to treat cancer cells as they cause irreparable DNA damage, inducing these aberrant cells to undergo cell death. The mediator of this process is presumed to be in part the tumor suppressor p53 which regulates genes involved in DNA repair and cell death. When MCF-7 breast cancer cells are treated with these drugs, we observed that the level of p53 and the p53 negative regulator, Mdm2, increased, as seen by others. But contrary to some reports, we observed minimal phosphorylation of p53 at serine 15 in MCF-7 cells after drug treatment. Interestingly, we determined that there was differential regulation of the kinases ATM and Chk2 with the drug treatments, likely the cause for the lack of phosphorylation of p53. We found a dramatic drop in p53 DNA binding affinity for p21 and other gene response elements (RE) after drug treatment. To determine if the p53 that accumulated in the drug treated cells was functionally active, we monitored changes in the protein products of two p53-regulated genes following drug treatment with and without the addition of a p53-specific siRNA. In response to 5FU, both p21 and Mdm2 proteins increased and that increase was alleviated if a p53-specific siRNA was added. This effect was not seen with the addition of dox. Thus, the phosphorylation at serine 15 is not necessary for the functional activation of this transcription factor. We propose a new model for the regulation of p53, Mdm2, and MdmX after drug treatment.
Collapse
Affiliation(s)
- Matthew T Balmer
- Department of Biological Sciences; Binghamton University; Binghamton, NY USA
| | - Ryan D Katz
- Department of Biological Sciences; Binghamton University; Binghamton, NY USA
| | - Si Liao
- Department of Biological Sciences; Binghamton University; Binghamton, NY USA
| | - James S Goodwine
- Department of Biological Sciences; Binghamton University; Binghamton, NY USA
| | - Susannah Gal
- Department of Biological Sciences; Binghamton University; Binghamton, NY USA
| |
Collapse
|
102
|
Mairinger FD, Walter RFH, Ting S, Vollbrecht C, Kollmeier J, Griff S, Hager T, Mairinger T, Christoph DC, Theegarten D, Schmid KW, Wohlschlaeger J. Mdm2 protein expression is strongly associated with survival in malignant pleural mesothelioma. Future Oncol 2014; 10:995-1005. [DOI: 10.2217/fon.13.261] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
ABSTRACT: Aims: TP53 mutations are extremely rare in malignant pleural mesothelioma (MPM). In TP53 wild-type tumors, the functional p53 protein can be inactivated by MDM2. Materials & methods: A total of 61 patient samples were tested for their Mdm2 and p53 protein expression levels via immunohistochemistry. Results: This study demonstrates nuclear Mdm2 expression in three out of four mesothelioma cell lines and 21.3% of the MPM specimens investigated. After silencing of the MDM2 gene by siRNA in MPM cell lines, Mdm2 immunoexpression is lost and cells show changes indicative of severe damage. Mdm2 protein expression in MPM is detected in epithelioid and biphasic subtypes only and is significantly associated with poor survival compared with Mdm2-negative tumors. This may be explained by increased Mdm2 levels possibly leading to an increased ubiquitilation and proteasomal degradation of functional p53 protein. Conclusion: Expression of Mdm2 is a strong prognostic factor associated with shortened overall survival in MPM.
Collapse
Affiliation(s)
- Fabian D Mairinger
- Institute of Pathology & Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Robert FH Walter
- Institute of Pathology & Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Saskia Ting
- Institute of Pathology & Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Jens Kollmeier
- Department of Pneumology, Helios Klinikum Emil von Behring, Berlin, Germany
| | - Sergei Griff
- Department of Pathology, Helios Klinikum Emil von Behring, Berlin, Germany
| | - Thomas Hager
- Institute of Pathology & Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Thomas Mairinger
- Department of Pathology, Helios Klinikum Emil von Behring, Berlin, Germany
| | - Daniel C Christoph
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Medicine, Division of Medical Oncology, University of Colorado Denver, Aurora, CO, USA
| | - Dirk Theegarten
- Institute of Pathology & Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kurt Werner Schmid
- Institute of Pathology & Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jeremias Wohlschlaeger
- Institute of Pathology & Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
103
|
Lang V, Pallara C, Zabala A, Lobato-Gil S, Lopitz-Otsoa F, Farrás R, Hjerpe R, Torres-Ramos M, Zabaleta L, Blattner C, Hay RT, Barrio R, Carracedo A, Fernandez-Recio J, Rodríguez MS, Aillet F. Tetramerization-defects of p53 result in aberrant ubiquitylation and transcriptional activity. Mol Oncol 2014; 8:1026-42. [PMID: 24816189 DOI: 10.1016/j.molonc.2014.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 03/19/2014] [Accepted: 04/02/2014] [Indexed: 11/26/2022] Open
Abstract
The tumor suppressor p53 regulates the expression of genes involved in cell cycle progression, senescence and apoptosis. Here, we investigated the effect of single point mutations in the oligomerization domain (OD) on tetramerization, transcription, ubiquitylation and stability of p53. As predicted by docking and molecular dynamics simulations, p53 OD mutants show functional defects on transcription, Mdm2-dependent ubiquitylation and 26S proteasome-mediated degradation. However, mutants unable to form tetramers are well degraded by the 20S proteasome. Unexpectedly, despite the lower structural stability compared to WT p53, p53 OD mutants form heterotetramers with WT p53 when expressed transiently or stably in cells wild type or null for p53. In consequence, p53 OD mutants interfere with the capacity of WT p53 tetramers to be properly ubiquitylated and result in changes of p53-dependent protein expression patterns, including the pro-apoptotic proteins Bax and PUMA under basal and adriamycin-induced conditions. Importantly, the patient derived p53 OD mutant L330R (OD1) showed the more severe changes in p53-dependent gene expression. Thus, in addition to the well-known effects on p53 stability, ubiquitylation defects promote changes in p53-dependent gene expression with implications on some of its functions.
Collapse
Affiliation(s)
- Valérie Lang
- Ubiquitylation and Cancer Molecular Biology Laboratory, Inbiomed, Mikeletegi 81, San Sebastián-Donostia 20009, Gipuzkoa, Spain.
| | - Chiara Pallara
- Joint BSC-IRB Research Program in Computational Biology, Life Sciences Department, Barcelona Supercomputing Center, Carrer Jordi Girona 29, 08034 Barcelona, Spain.
| | - Amaia Zabala
- CIC bioGUNE, Ed 801A Parque Tecnológico de Bizkaia, 48160 Derio, Bizkaia, Spain.
| | - Sofia Lobato-Gil
- Ubiquitylation and Cancer Molecular Biology Laboratory, Inbiomed, Mikeletegi 81, San Sebastián-Donostia 20009, Gipuzkoa, Spain.
| | | | - Rosa Farrás
- Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, 46012 Valencia, Spain.
| | - Roland Hjerpe
- CIC bioGUNE, Ed 801A Parque Tecnológico de Bizkaia, 48160 Derio, Bizkaia, Spain.
| | - Monica Torres-Ramos
- CIC bioGUNE, Ed 801A Parque Tecnológico de Bizkaia, 48160 Derio, Bizkaia, Spain.
| | - Lorea Zabaleta
- Ubiquitylation and Cancer Molecular Biology Laboratory, Inbiomed, Mikeletegi 81, San Sebastián-Donostia 20009, Gipuzkoa, Spain.
| | - Christine Blattner
- Karlsruher Institute of Technology, Institute of Toxicology and Genetics, Fritz-Erler-Straße 23, 76133 Karlsruhe, Germany.
| | - Ronald T Hay
- Center for Interdisciplinary Research, School of Life Sciences, University of Dundee, Dow Street, DD15EH Scotland, United Kingdom.
| | - Rosa Barrio
- CIC bioGUNE, Ed 801A Parque Tecnológico de Bizkaia, 48160 Derio, Bizkaia, Spain.
| | - Arkaitz Carracedo
- CIC bioGUNE, Ed 801A Parque Tecnológico de Bizkaia, 48160 Derio, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, 48011 Bilbao, Spain; Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), P.O. Box 644, E-48080 Bilbao, Spain.
| | - Juan Fernandez-Recio
- Joint BSC-IRB Research Program in Computational Biology, Life Sciences Department, Barcelona Supercomputing Center, Carrer Jordi Girona 29, 08034 Barcelona, Spain.
| | - Manuel S Rodríguez
- Ubiquitylation and Cancer Molecular Biology Laboratory, Inbiomed, Mikeletegi 81, San Sebastián-Donostia 20009, Gipuzkoa, Spain.
| | - Fabienne Aillet
- Ubiquitylation and Cancer Molecular Biology Laboratory, Inbiomed, Mikeletegi 81, San Sebastián-Donostia 20009, Gipuzkoa, Spain.
| |
Collapse
|
104
|
Read ML, Seed RI, Fong JCW, Modasia B, Ryan GA, Watkins RJ, Gagliano T, Smith VE, Stratford AL, Kwan PK, Sharma N, Dixon OM, Watkinson JC, Boelaert K, Franklyn JA, Turnell AS, McCabe CJ. The PTTG1-binding factor (PBF/PTTG1IP) regulates p53 activity in thyroid cells. Endocrinology 2014; 155:1222-34. [PMID: 24506068 PMCID: PMC4759943 DOI: 10.1210/en.2013-1646] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The PTTG1-binding factor (PBF/PTTG1IP) has an emerging repertoire of roles, especially in thyroid biology, and functions as a protooncogene. High PBF expression is independently associated with poor prognosis and lower disease-specific survival in human thyroid cancer. However, the precise role of PBF in thyroid tumorigenesis is unclear. Here, we present extensive evidence demonstrating that PBF is a novel regulator of p53, a tumor suppressor protein with a key role in maintaining genetic stability, which is infrequently mutated in differentiated thyroid cancer. By coimmunoprecipitation and proximity-ligation assays, we show that PBF binds specifically to p53 in thyroid cells and significantly represses transactivation of responsive promoters. Further, we identify that PBF decreases p53 stability by enhancing ubiquitination, which appears dependent on the E3 ligase activity of Mdm2. Impaired p53 function was evident in a transgenic mouse model with thyroid-specific PBF overexpression (transgenic PBF mice), which had significantly increased genetic instability as indicated by fluorescent inter simple sequence repeat-PCR analysis. Consistent with this, approximately 40% of all DNA repair genes examined were repressed in transgenic PBF primary cultures, including genes with critical roles in maintaining genomic integrity such as Mgmt, Rad51, and Xrcc3. Our data also revealed that PBF induction resulted in up-regulation of the E2 enzyme Rad6 in murine thyrocytes and was associated with Rad6 expression in human thyroid tumors. Overall, this work provides novel insights into the role of the protooncogene PBF as a negative regulator of p53 function in thyroid tumorigenesis, in which PBF is generally overexpressed and p53 mutations are rare compared with other tumor types.
Collapse
Affiliation(s)
- Martin L Read
- School of Clinical and Experimental Medicine (M.L.R., R.I.S., J.C.W.F., B.M., G.A.R., R.J.W., V.E.S., P.K.K., N.S., O.M.D., K.B., J.A.F., C.J.M.) and School of Cancer Sciences (A.S.T.), University of Birmingham, Birmingham, United Kingdom; Department of Medical Sciences (T.G.), University of Ferrara, Ferrara, Italy; Department of Pediatrics (A.L.S.), University of British Columbia, Vancouver, British Columbia, Canada; and University Hospitals Birmingham National Health Service Foundation Trust (J.C.W.), Birmingham, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Brd4 and HEXIM1: multiple roles in P-TEFb regulation and cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:232870. [PMID: 24592384 PMCID: PMC3925632 DOI: 10.1155/2014/232870] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/19/2013] [Indexed: 12/31/2022]
Abstract
Bromodomain-containing protein 4 (Brd4) and hexamethylene bisacetamide (HMBA) inducible protein 1 (HEXIM1) are two opposing regulators of the positive transcription elongation factor b (P-TEFb), which is the master modulator of RNA polymerase II during transcriptional elongation. While Brd4 recruits P-TEFb to promoter-proximal chromatins to activate transcription, HEXIM1 sequesters P-TEFb into an inactive complex containing the 7SK small nuclear RNA. Besides regulating P-TEFb's transcriptional activity, recent evidence demonstrates that both Brd4 and HEXIM1 also play novel roles in cell cycle progression and tumorigenesis. Here we will discuss the current knowledge on Brd4 and HEXIM1 and their implication as novel therapeutic options against cancer.
Collapse
|
106
|
Hock AK, Vousden KH. The role of ubiquitin modification in the regulation of p53. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:137-49. [DOI: 10.1016/j.bbamcr.2013.05.022] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 04/16/2013] [Accepted: 05/23/2013] [Indexed: 01/09/2023]
|
107
|
Nag S, Zhang X, Srivenugopal K, Wang MH, Wang W, Zhang R. Targeting MDM2-p53 interaction for cancer therapy: are we there yet? Curr Med Chem 2014; 21:553-74. [PMID: 24180275 PMCID: PMC6690199 DOI: 10.2174/09298673113206660325] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 10/02/2013] [Accepted: 10/22/2013] [Indexed: 11/22/2022]
Abstract
Inactivation of the tumor suppressor p53 and/or overexpression of the oncogene MDM2 frequently occur in human cancers, and are associated with poor prognosis, advanced forms of the disease, and chemoresistance. MDM2, the major negative regulator of p53, induces p53 degradation and inactivates its tumor suppressing activity. In turn, p53 regulates MDM2 expression. This MDM2-p53 negative feedback loop has been widely studied and presents an attractive target for cancer therapy, with a few of the inhibitors of this interaction already having advanced into clinical trials. Additionally, there is an increasing interest in understanding MDM2's p53-independent activities in carcinogenesis and cancer progression, which may also have implications for cancer therapy. This review aims to highlight the various roles that the MDM2-p53 interaction plays in cancer, the p53 independent oncogenic activities of MDM2 and the various strategies that may be used to target MDM2 and the MDM2-p53 interaction. We will summarize the major preclinical and clinical evidences of MDM2 inhibitors for human cancer treatment and make suggestions to further improve efficacy and safety of this interesting class of cancer therapeutics.
Collapse
Affiliation(s)
- S. Nag
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - X. Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - K.S. Srivenugopal
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - M.-H. Wang
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - W. Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - R. Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
108
|
DeHart CJ, Chahal JS, Flint SJ, Perlman DH. Extensive post-translational modification of active and inactivated forms of endogenous p53. Mol Cell Proteomics 2013; 13:1-17. [PMID: 24056736 DOI: 10.1074/mcp.m113.030254] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The p53 tumor suppressor protein accumulates to very high concentrations in normal human fibroblasts infected by adenovirus type 5 mutants that cannot direct assembly of the viral E1B 55-kDa protein-containing E3 ubiquitin ligase that targets p53 for degradation. Despite high concentrations of nuclear p53, the p53 transcriptional program is not induced in these infected cells. We exploited this system to examine select post-translational modifications (PTMs) present on a transcriptionally inert population of endogenous human p53, as well as on p53 activated in response to etoposide treatment of normal human fibroblasts. These forms of p53 were purified from whole cell lysates by means of immunoaffinity chromatography and SDS-PAGE, and peptides derived from them were subjected to nano-ultra-high-performance LC-MS and MS/MS analyses on a high-resolution accurate-mass MS platform (data available via ProteomeXchange, PXD000464). We identified an unexpectedly large number of PTMs, comprising phosphorylation of Ser and Thr residues, methylation of Arg residues, and acetylation, ubiquitinylation, and methylation of Lys residues-for example, some 150 previously undescribed modifications of p53 isolated from infected cells. These modifications were distributed across all functional domains of both forms of the endogenous human p53 protein, as well as those of an orthologous population of p53 isolated from COS-1 cells. Despite the differences in activity, including greater in vitro sequence-specific DNA binding activity exhibited by p53 isolated from etoposide-treated cells, few differences were observed in the location, nature, or relative frequencies of PTMs on the two populations of human p53. Indeed, the wealth of PTMs that we have identified is consistent with a far greater degree of complex, combinatorial regulation of p53 by PTM than previously anticipated.
Collapse
Affiliation(s)
- Caroline J DeHart
- Department of Molecular Biology, Lewis Thomas Laboratory, Princeton University, Princeton, New Jersey 08544
| | | | | | | |
Collapse
|
109
|
Abstract
SUMO (small ubiquitin-like modifier) emerged from the shadow of the well-established ubiquitin some 15 years ago when it was shown that a distinct conjugation pathway was responsible for SUMO modification. Since then it has been established that SUMO modifies over a thousand substrates and plays diverse roles in many important biological processes. Recognition of SUMO is mediated by short peptide sequences known as SIMs (SUMO-interaction motifs) that allow effector proteins to engage SUMO-modified substrates. Like ubiquitin, SUMO can form polymeric chains, and these chains can be recognized by proteins containing multiple SIMs. One protein that contains such a sequence of SIMs also contains a RING (really interesting new gene) domain that is the hallmark of a ubiquitin E3 ligase. This ubiquitin ligase known as RNF4 (RING finger protein 4) has the unique property that it can recognize SUMO-modified proteins and target them for ubiquitin-mediated proteolysis. Structural and biochemical analyses of RNF4 has shed light on the long sought after mechanism of ubiquitin transfer and illustrates how its RING domain primes the ubiquitin-loaded E2 for catalysis.
Collapse
|
110
|
Ciechanover A, Stanhill A. The complexity of recognition of ubiquitinated substrates by the 26S proteasome. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:86-96. [PMID: 23872423 DOI: 10.1016/j.bbamcr.2013.07.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/02/2013] [Accepted: 07/08/2013] [Indexed: 02/03/2023]
Abstract
The Ubiquitin Proteasome System (UPS) was discovered in two steps. Initially, APF-1 (ATP-dependent proteolytic Factor 1) later identified as ubiquitin (Ub), a hitherto known protein of unknown function, was found to covalently modify proteins. This modification led to degradation of the tagged protein by - at that time - an unknown protease. This was followed later by the identification of the 26S proteasome complex which is composed of a previously identified Multi Catalytic Protease (MCP) and an additional regulatory complex, as the protease that degrades Ub-tagged proteins. While Ub conjugation and proteasomal degradation are viewed as a continued process responsible for most of the regulated proteolysis in the cell, the two processes have also independent roles. In parallel and in the years that followed, the hallmark signal that links the substrate to the proteasome was identified as an internal Lys48-based polyUb chain. However, since these initial findings were described, our understanding of both ends of the process (i.e. Ub-conjugation to proteins, and their recognition and degradation), have advanced significantly. This enabled us to start bridging the ends of this continuous process which suffered until lately from limited structural data regarding the 26S proteasomal architecture and the structure and diversity of the Ub chains. These missing pieces are of great importance because the link between ubiquitination and proteasomal processing is subject to numerous regulatory steps and are found to function improperly in several pathologies. Recently, the molecular architecture of the 26S proteasome was resolved in great detail, enabling us to address mechanistic questions regarding the various molecular events that polyubiquitinated (polyUb) substrates undergo during binding and processing by the 26S proteasome. In addition, advancement in analytical and synthetic methods enables us to better understand the structure and diversity of the degradation signal. The review summarizes these recent findings and addresses the extrapolated meanings in light of previous reports. Finally, it addresses some of the still remaining questions to be solved in order to obtain a continuous mechanistic view of the events that a substrate undergoes from its initial ubiquitination to proteasomal degradation. This article is part of a Special Issue entitled: Ubiquitin-Proteasome System. Guest Editors: Thomas Sommer and Dieter H. Wolf.
Collapse
Affiliation(s)
- Aaron Ciechanover
- The David and Janet Polak Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | | |
Collapse
|
111
|
Lew QJ, Chu KL, Chia YL, Cheong N, Chao SH. HEXIM1, a New Player in the p53 Pathway. Cancers (Basel) 2013; 5:838-56. [PMID: 24202322 PMCID: PMC3795367 DOI: 10.3390/cancers5030838] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/24/2013] [Accepted: 06/24/2013] [Indexed: 01/10/2023] Open
Abstract
Hexamethylene bisacetamide-inducible protein 1 (HEXIM1) is best known as the inhibitor of positive transcription elongation factor b (P-TEFb), which controls transcription elongation of RNA polymerase II and Tat transactivation of human immunodeficiency virus. Besides P-TEFb, several proteins have been identified as HEXIM1 binding proteins. It is noteworthy that more than half of the HEXIM1 binding partners are involved in cancers. P53 and two key regulators of the p53 pathway, nucleophosmin (NPM) and human double minute-2 protein (HDM2), are among the factors identified. This review will focus on the functional importance of the interactions between HEXIM1 and p53/NPM/HDM2. NPM and the cytoplasmic mutant of NPM, NPMc+, were found to regulate P-TEFb activity and RNA polymerase II transcription through the interaction with HEXIM1. Importantly, more than one-third of acute myeloid leukemia (AML) patients carry NPMc+, suggesting the involvement of HEXIM1 in tumorigenesis of AML. HDM2 was found to ubiquitinate HEXIM1. The HDM2-mediated ubiquitination of HEXIM1 did not lead to protein degradation of HEXIM1 but enhanced its inhibitory activity on P-TEFb. Recently, HEXIM1 was identified as a novel positive regulator of p53. HEXIM1 prevented p53 ubiquitination by competing with HDM2 in binding to p53. Taken together, the new evidence suggests a role of HEXIM1 in regulating the p53 pathway and tumorigenesis.
Collapse
Affiliation(s)
- Qiao Jing Lew
- Expression Engineering Group, Bioprocessing Technology Institute, A*STAR (Agency for Science, Technology and Research), 20 Biopolis Way, #06-01, Singapore 138668, Singapore.
| | | | | | | | | |
Collapse
|
112
|
Nag S, Qin J, Srivenugopal KS, Wang M, Zhang R. The MDM2-p53 pathway revisited. J Biomed Res 2013; 27:254-71. [PMID: 23885265 PMCID: PMC3721034 DOI: 10.7555/jbr.27.20130030] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 04/12/2013] [Indexed: 12/15/2022] Open
Abstract
The p53 tumor suppressor is a key transcription factor regulating cellular pathways such as DNA repair, cell cycle, apoptosis, angiogenesis, and senescence. It acts as an important defense mechanism against cancer onset and progression, and is negatively regulated by interaction with the oncoprotein MDM2. In human cancers, the TP53 gene is frequently mutated or deleted, or the wild-type p53 function is inhibited by high levels of MDM2, leading to downregulation of tumor suppressive p53 pathways. Thus, the inhibition of MDM2-p53 interaction presents an appealing therapeutic strategy for the treatment of cancer. However, recent studies have revealed the MDM2-p53 interaction to be more complex involving multiple levels of regulation by numerous cellular proteins and epigenetic mechanisms, making it imperative to reexamine this intricate interplay from a holistic viewpoint. This review aims to highlight the multifaceted network of molecules regulating the MDM2-p53 axis to better understand the pathway and exploit it for anticancer therapy.
Collapse
Affiliation(s)
- Subhasree Nag
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | | | | | | | | |
Collapse
|
113
|
Qin JJ, Nag S, Voruganti S, Wang W, Zhang R. Natural product MDM2 inhibitors: anticancer activity and mechanisms of action. Curr Med Chem 2013; 19:5705-25. [PMID: 22830335 DOI: 10.2174/092986712803988910] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Revised: 06/04/2012] [Accepted: 06/04/2012] [Indexed: 12/12/2022]
Abstract
The mdm2 oncogene has recently been suggested to be a valuable target for cancer therapy and prevention. Overexpression of mdm2 is often seen in various human cancers and correlates with high-grade, late-stage, and more treatment-resistant tumors. The MDM2-p53 auto-regulatory loop has been extensively investigated and is an attractive cancer target, which indeed has been the main focus of anti-MDM2 drug discovery. Much effort has been expended in the development of small molecule MDM2 antagonists targeting the MDM2-p53 interaction, and a few of these have advanced into clinical trials. However, MDM2 exerts its oncogenic activity through both p53-dependent and -independent mechanisms. Recently, there is an increasing interest in identifying natural MDM2 inhibitors; some of them have been shown to decrease MDM2 expression and activity in vitro and in vivo. These identified natural MDM2 inhibitors include a plethora of diverse chemical frameworks, ranging from flavonoids, steroids, and sesquiterpenes to alkaloids. In addition to a brief review of synthetic MDM2 inhibitors, this review focuses on natural product MDM2 inhibitors, summarizing their biological activities in vitro and in vivo and the underlying molecular mechanisms of action, targeting MDM2 itself, regulators of MDM2, and/or the MDM2-p53 interaction. These MDM2 inhibitors can be used alone or in combination with conventional treatments, improving the prospects for cancer therapy and prevention. Their complex and unique molecular architectures may provide a stimulus for developing synthetic analogs in the future.
Collapse
Affiliation(s)
- J-J Qin
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX 79106, USA
| | | | | | | | | |
Collapse
|
114
|
Xu J, Zhou X, Wang J, Li Z, Kong X, Qian J, Hu Y, Fang JY. RhoGAPs attenuate cell proliferation by direct interaction with p53 tetramerization domain. Cell Rep 2013; 3:1526-38. [PMID: 23684608 DOI: 10.1016/j.celrep.2013.04.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 03/13/2013] [Accepted: 04/21/2013] [Indexed: 12/15/2022] Open
Abstract
Many Rho GTPase activation proteins (RhoGAPs) are deleted or downregulated in cancers, but the functional consequences are still unclear. Here, we show that the RhoGAP ArhGAP11A induces cell-cycle arrest and apoptosis by binding to the tumor suppressor p53. The RhoGAP domain of ArhGAP11A binds to the tetramerization domain of p53, but not to its family members p63 or p73. The interaction stabilizes the tetrameric conformation of p53 and enhances its DNA-binding activity, thereby inducing cell-cycle arrest and apoptosis. Upon DNA damage stress, ArhGAP11A accumulates in the nucleus and interacts with p53, whereas knockdown of ArhGAP11A partially blocks p53 transcriptional activity. These findings explain why RhoGAPs are frequently deleted in cancers and suggest that the RhoGAP family sits at the crossroads between the cell-migration and proliferation pathways.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Institute for Digestive Diseases, Shanghai Jiao-Tong University School of Medicine, Key Laboratory of Gastroenterology & Hepatology, Ministry of Health, 145 Middle Shandong Road, Shanghai 200001, China.
| | | | | | | | | | | | | | | |
Collapse
|
115
|
Arora A, Gera S, Maheshwari T, Raghav D, Alam MJ, Singh RKB, Agarwal SM. The dynamics of stress p53-Mdm2 network regulated by p300 and HDAC1. PLoS One 2013; 8:e52736. [PMID: 23437037 PMCID: PMC3577848 DOI: 10.1371/journal.pone.0052736] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 11/21/2012] [Indexed: 11/18/2022] Open
Abstract
We construct a stress p53-Mdm2-p300-HDAC1 regulatory network that is activated and stabilised by two regulatory proteins, p300 and HDAC1. Different activation levels of [Formula: see text] observed due to these regulators during stress condition have been investigated using a deterministic as well as a stochastic approach to understand how the cell responds during stress conditions. We found that these regulators help in adjusting p53 to different conditions as identified by various oscillatory states, namely fixed point oscillations, damped oscillations and sustain oscillations. On assessing the impact of p300 on p53-Mdm2 network we identified three states: first stabilised or normal condition where the impact of p300 is negligible, second an interim region where p53 is activated due to interaction between p53 and p300, and finally the third regime where excess of p300 leads to cell stress condition. Similarly evaluation of HDAC1 on our model led to identification of the above three distinct states. Also we observe that noise in stochastic cellular system helps to reach each oscillatory state quicker than those in deterministic case. The constructed model validated different experimental findings qualitatively.
Collapse
Affiliation(s)
- Akshit Arora
- Bioinformatics Division, Institute of Cytology and Preventive Oncology, Noida, India
| | - Saurav Gera
- Bioinformatics Division, Institute of Cytology and Preventive Oncology, Noida, India
| | - Tanuj Maheshwari
- Bioinformatics Division, Institute of Cytology and Preventive Oncology, Noida, India
| | - Dhwani Raghav
- Bioinformatics Division, Institute of Cytology and Preventive Oncology, Noida, India
| | - Md. Jahoor Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - R. K. Brojen Singh
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Subhash M. Agarwal
- Bioinformatics Division, Institute of Cytology and Preventive Oncology, Noida, India
| |
Collapse
|
116
|
Silden E, Hjelle SM, Wergeland L, Sulen A, Andresen V, Bourdon JC, Micklem DR, McCormack E, Gjertsen BT. Expression of TP53 isoforms p53β or p53γ enhances chemosensitivity in TP53(null) cell lines. PLoS One 2013; 8:e56276. [PMID: 23409163 PMCID: PMC3569410 DOI: 10.1371/journal.pone.0056276] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 01/12/2013] [Indexed: 11/18/2022] Open
Abstract
The carboxy-terminal truncated p53 alternative spliced isoforms, p53β and p53γ, are expressed at disparate levels in cancer and are suggested to influence treatment response and therapy outcome. However, their functional role in cancer remains to be elucidated. We investigated their individual functionality in the p53null background of cell lines H1299 and SAOS-2 by stable retroviral transduction or transient transfection. Expression status of p53β and p53γ protein was found to correlate with increased response to camptothecin and doxorubicin chemotherapy. Decreased DNA synthesis and clonogenicity in p53β and p53γ congenic H1299 was accompanied by increased p21(CIP1/WAF1), Bax and Mdm2 proteins. Chemotherapy induced p53 isoform degradation, most prominent for p53γ. The proteasome inhibitor bortezomib substantially increased basal p53γ protein level, while the level of p53β protein was unaffected. Treatment with dicoumarol, a putative blocker of the proteasome-related NAD(P)H quinone oxidoreductase NQO1, effectively attenuated basal p53γ protein level in spite of bortezomib treatment. Although in vitro proliferation and clonogenicity assays indicated a weak suppressive effect by p53β and p53γ expression, studies of in vivo subcutaneous H1299 tumor growth demonstrated a significantly increased growth by expression of either p53 isoforms. This study suggests that p53β and p53γ share functionality in chemosensitizing and tumor growth enhancement but comprise distinct regulation at the protein level.
Collapse
Affiliation(s)
- Elisabeth Silden
- Hematology Section, Institute of Medicine, University of Bergen, Bergen, Norway
| | - Sigrun M. Hjelle
- Hematology Section, Institute of Medicine, University of Bergen, Bergen, Norway
| | - Line Wergeland
- Hematology Section, Institute of Medicine, University of Bergen, Bergen, Norway
| | - André Sulen
- Hematology Section, Institute of Medicine, University of Bergen, Bergen, Norway
| | - Vibeke Andresen
- Hematology Section, Institute of Medicine, University of Bergen, Bergen, Norway
| | - Jean-Christophe Bourdon
- Inserm-European Associated Laboratory Inserm U858, Department of Surgery and Molecular Oncology, University of Dundee Medical School, Dundee, Scotland, United Kingdom
| | | | - Emmet McCormack
- Hematology Section, Institute of Medicine, University of Bergen, Bergen, Norway
| | - Bjørn Tore Gjertsen
- Hematology Section, Institute of Medicine, University of Bergen, Bergen, Norway
- Hematology Section, Department of Internal Medicine, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| |
Collapse
|
117
|
Micel LN, Tentler JJ, Smith PG, Eckhardt GS. Role of ubiquitin ligases and the proteasome in oncogenesis: novel targets for anticancer therapies. J Clin Oncol 2013; 31:1231-8. [PMID: 23358974 DOI: 10.1200/jco.2012.44.0958] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The ubiquitin proteasome system (UPS) regulates the ubiquitination, and thus degradation and turnover, of many proteins vital to cellular regulation and function. The UPS comprises a sequential series of enzymatic processes using four key enzyme families: E1 (ubiquitin-activating enzymes), E2 (ubiquitin-carrier proteins), E3 (ubiquitin-protein ligases), and E4 (ubiquitin chain assembly factors). Because the UPS is a crucial regulator of the cell cycle, and abnormal cell-cycle control can lead to oncogenesis, aberrancies within the UPS pathway can result in a malignant cellular phenotype and thus has become an attractive target for novel anticancer agents. This article will provide an overall review of the mechanics of the UPS, describe aberrancies leading to cancer, and give an overview of current drug therapies selectively targeting the UPS.
Collapse
|
118
|
Abstract
The tumor suppressor p53 is essential for several cellular processes that are involved in the response to diverse genotoxic stress, including cell cycle arrest, DNA repair, apoptosis and senescence. Studies of the regulation of p53 have mostly focused on its stability and transactivation; however, new regulatory molecules for p53 have also been frequently identified. Here, we report that human ssDNA binding protein SSB1 (hSSB1), a novel DNA damage-associated protein, can interact with p53 and protect p53 from ubiquitin-mediated degradation. Furthermore, hSSB1 also associates with the acetyltransferase p300 and is required for efficient transcriptional activation of the p53 target gene p21 by affecting the acetylation of p53 at lysine382. Functionally, the hSSB1 knockdown-induced abrogation of the G2/M checkpoint is partially dependent on p53 or p300. Collectively, our results indicate that hSSB1 may regulate DNA damage checkpoints by positively modulating p53 and its downstream target p21.
Collapse
|
119
|
Zorić A, Horvat A, Slade N. Differential effects of diverse p53 isoforms on TAp73 transcriptional activity and apoptosis. Carcinogenesis 2012. [PMID: 23188674 DOI: 10.1093/carcin/bgs370] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The p53 activities are due, at least in part, to its ability to form oligomers that bind to specific DNA sequences and activate transcription. Since some mutant p53 proteins and ΔNp73 isoforms form heterocomplexes with TAp73, we asked whether p53 isoforms can do the same and potentially act as dominant-negative inhibitors of TAp73. Moreover, it has already been found that some isoforms form complex with wtp53 and some of them inhibit p53 tumor-suppressor functions. Therefore, we studied the complex formation and co-immunoprecipitation assays show that all six p53 isoforms examined can form complexes with TAp73β, whereas only Δ133p53α/β/γ isoforms form complex with TAp73α. All p53 isoforms counteract TAp73β transactivation function but with different efficiency and in a promoter-dependent manner. Furthermore, apoptotic activity of TAp73β was augmented by coexpression of p53β, whereas Δ133p53α and β inhibit its apoptotic activity most efficiently. We have determined the half-life of different p53 isoforms: p53γ isoform has the shortest half-life, whereas Δ133p53γ has the longest half-life. Inhibitory interactions of two proteins in complex often lead to their stabilization. However, only three isoforms (Δ133p53α, Δ133p53β and Δ40p53α) stabilize TAp73β. We are convinced that defining the interactions between p53/p73 would give a new insight into how the p53 isoforms modulate the p73 functions in tumorigenesis.
Collapse
Affiliation(s)
- Arijana Zorić
- Division of Molecular Medicine, Rudjer Bošković Institute, Bijenička 54, 10000 Zagreb, Croatia
| | | | | |
Collapse
|
120
|
Ponnuswamy A, Hupp T, Fåhraeus R. Concepts in MDM2 Signaling: Allosteric Regulation and Feedback Loops. Genes Cancer 2012; 3:291-7. [PMID: 23150762 DOI: 10.1177/1947601912454140] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The function and regulation of MDM2 as a component of a p53-dependent negative feedback loop has formed a core paradigm in the p53 field. This concept, now 20 years old, has been solidified by fields of protein science, transgenic technology, and drug discovery in human cancer. However, it has been noted that a simple negative feedback loop between p53 and MDM2 lacks an intrinsic "activating" step that counteracts this inhibition and permits oscillation of the feedback to occur as p53 is switched on and off. More recent work has identified a solution to the missing piece of the picture that counters the negative feedback loop, which is MDM2 itself. Under conditions of genotoxic stress, MDM2 helps to activate p53 by increasing its rate of protein synthesis. This simple observation makes certain aspects of the p53 response more comprehensible such as why MDM2 is upregulated by p53 early on following DNA damage and how phosphorylation of MDM2 at the C-terminal Ser395 by ATM translates into p53 activation. The latter acts by inducing allosteric changes in the RING domain of MDM2 that expose its RNA binding pocket, support p53 synthesis, and suppress its degradation. This allosteric nature of MDM2 in the C-terminus mirrors the allosteric effects of the binding of small molecules to the p53 interacting pocket at the N-terminus of MDM2, which opens the core domain of MDM2 to central domains of p53, which controls p53 ubiquitination. Thus, the highly allosteric nature of MDM2 provides the basis for dynamic protein-protein interactions and protein-RNA interactions through which MDM2's activity is regulated in p53 protein destruction or in p53 protein synthesis. We discuss these mechanisms and how this information can be exploited for drug development programs aimed at activating p53 via targeting MDM2.
Collapse
Affiliation(s)
- Anand Ponnuswamy
- Cibles Therapeutiques, INSERM Unité 940, Institut de Génétique Moléculaire, IUH Hôpital St. Louis, Paris, France ; RECAMO, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | | | | |
Collapse
|
121
|
Kravtsova-Ivantsiv Y, Sommer T, Ciechanover A. The lysine48-based polyubiquitin chain proteasomal signal: not a single child anymore. Angew Chem Int Ed Engl 2012; 52:192-8. [PMID: 23124625 DOI: 10.1002/anie.201205656] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Indexed: 11/07/2022]
Abstract
The conjugation of ubiquitin (Ub) to proteins is involved in the regulation of many processes. The modification serves as a recognition element in trans, in which downstream effectors bind to the modified protein and determine its fate and/or function. A polyUb chain that is linked through internal lysine (Lys)-48 of Ub and anchored to an internal Lys residue of the substrate has become the accepted "canonical" signal for proteasomal targeting and degradation. However, recent studies show that the signal is far more diverse and that chains based on other internal linkages, as well as linear or heterologous chains made of Ub and Ub-like proteins and even monoUb, are recognized by the proteasome. In addition, chains linked to residues other than internal Lys were described, all challenging the current paradigm.
Collapse
Affiliation(s)
- Yelena Kravtsova-Ivantsiv
- Cancer and Vascular Biology Research Center, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | | | | |
Collapse
|
122
|
Retzlaff M, Rohrberg J, Küpper NJ, Lagleder S, Bepperling A, Manzenrieder F, Peschek J, Kessler H, Buchner J. The regulatory domain stabilizes the p53 tetramer by intersubunit contacts with the DNA binding domain. J Mol Biol 2012; 425:144-55. [PMID: 23103206 DOI: 10.1016/j.jmb.2012.10.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/16/2012] [Accepted: 10/18/2012] [Indexed: 12/24/2022]
Abstract
The tumor suppressor protein p53 is often referred to as the guardian of the genome. In the past, controversial findings have been presented for the role of the C-terminal regulatory domain (RD) of p53 as both a negative regulator and a positive regulator of p53 activity. However, the underlying mechanism remained enigmatic. To understand the function of the RD and of a dominant phosphorylation site within the RD, we analyzed p53 variants in vivo and in vitro. Our experiments revealed, surprisingly, that the p53 RD of one subunit interacts with the DNA binding domain of an adjacent subunit in the tetramer. This leads to the formation of intersubunit contacts that stabilize the tetrameric state of p53 and enhance its transcriptional activity in a cooperative manner. These effects are further modulated by phosphorylation of a conserved serine within the RD.
Collapse
Affiliation(s)
- Marco Retzlaff
- Center for Integrated Protein Science Munich at the Department Chemie, Technische Universität München, Lichtenbergstrasse 4, 85747 Garching, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Lew QJ, Chia YL, Chu KL, Lam YT, Gurumurthy M, Xu S, Lam KP, Cheong N, Chao SH. Identification of HEXIM1 as a positive regulator of p53. J Biol Chem 2012; 287:36443-54. [PMID: 22948151 DOI: 10.1074/jbc.m112.374157] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hexamethylene bisacetamide-inducible protein 1 (HEXIM1) is best known as the inhibitor of positive transcription elongation factor b (P-TEFb), which regulates the transcription elongation of RNA polymerase II and controls 60-70% of mRNA synthesis. Our previous studies show that HEXIM1 interacts with two key p53 regulators, nucleophosmin and human double minute-2 protein (HDM2), implying a possible connection between HEXIM1 and the p53 signaling pathway. Here we report the interaction between p53 and HEXIM1 in breast cancer, acute myeloid leukemia, and colorectal carcinoma cells. The C-terminal regions of p53 and HEXIM1 are required for the protein-protein interaction. Overexpression of HEXIM1 prevents the ubiquitination of p53 by HDM2 and enhances the protein stability of p53, resulting in up-regulation of p53 target genes, such as Puma and p21. Induction of p53 can be achieved by several means, such as UV radiation and treatment with anti-cancer agents (including doxorubicin, etoposide, roscovitine, flavopiridol, and nutlin-3). Under all the conditions examined, elevated protein levels of p53 are found to associate with the increased p53-HEXIM1 interaction. In addition, knockdown of HEXIM1 significantly inhibits the induction of p53 and releases the cell cycle arrest caused by p53. Finally, the transcription of the p53 target genes is regulated by HEXIM1 in a p53-dependent fashion. Our results not only identify HEXIM1 as a positive regulator of p53, but also propose a novel molecular mechanism of p53 activation caused by the anti-cancer drugs and compounds.
Collapse
Affiliation(s)
- Qiao Jing Lew
- Expression Engineering Group, Bioprocessing Technology Institute, Agency for Science, Technology, and Research (A*STAR), Singapore 138668, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
124
|
|
125
|
Spasser L, Brik A. Chemistry and Biology of the Ubiquitin Signal. Angew Chem Int Ed Engl 2012; 51:6840-62. [DOI: 10.1002/anie.201200020] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Indexed: 01/07/2023]
|
126
|
Ching LY, Yeung BHY, Wong CKC. Synergistic effect of p53 on TSA-induced stanniocalcin 1 expression in human nasopharyngeal carcinoma cells, CNE2. J Mol Endocrinol 2012; 48:241-50. [PMID: 22493143 DOI: 10.1530/jme-11-0159] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Human stanniocalcin 1 (STC1) has recently been identified as a putative protein factor involved in cellular apoptosis. The use of histone deacetylase inhibitor (i.e. trichostatin A (TSA)) and doxorubicin (Dox) is one of the common treatment methods to induce apoptosis in human cancer cells. A study on TSA and Dox-mediated apoptosis may shed light on the regulation and function of STC1 in cancer treatment. In this study, TSA and Dox cotreatment in human nasopharyngeal carcinoma cells (CNE2) elicited synergistic effects on STC1 gene expression and cellular apoptosis. An activation of p53 (TP53) transcriptional activity in Dox- or Dox+TSA-treated cells was revealed by the increased expression levels of p53 mRNA/protein as well as p53-driven luciferase activities. To elucidate the possible involvement of p53 in STC1 gene transcription, a vector expressing wild-type or dominant negative (DN) p53 was transiently transfected into the cells. Both STC1 promoter luciferase constructs and chromatin immunoprecipitation assays did not support the direct role of p53 in STC1 gene transactivation. However, the synergistic effects of p53 on the induction of NF-κB phosphorylation and the recruitment of acetylated histone H3 in STC1 promoter were observed in TSA-cotreated cells. The overexpression of exogenous STC1 sensitized apoptosis in Dox-treated cells. Taken together, this study provides data to show the cross talk of NF-κB, p53, and histone protein in the regulation of STC1 expression and function.
Collapse
Affiliation(s)
- L Y Ching
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | | | | |
Collapse
|
127
|
A protein transduction method using oligo-arginine (3R) for the delivery of transcription factors into cell nuclei. Biomaterials 2012; 33:4665-72. [DOI: 10.1016/j.biomaterials.2012.02.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 02/27/2012] [Indexed: 11/23/2022]
|
128
|
Yamada S, Kanno H, Kawahara N. Trans-membrane peptide therapy for malignant glioma by use of a peptide derived from the MDM2 binding site of p53. J Neurooncol 2012; 109:7-14. [PMID: 22528789 DOI: 10.1007/s11060-012-0860-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 03/28/2012] [Indexed: 02/03/2023]
Abstract
A new strategy is required against glioblastoma, a highly aggressive and fatal disease. In recent studies the protein transduction domains (PTDs) of some proteins, which are able to cross biological membranes, have been identified as critical domains for protein transduction. Here, we show that this protein-delivery system is a powerful tool for transduction of p53, a biologically active tumor-suppressor protein, into cancer cells, to suppress their proliferation. A 15-amino-acid sequence corresponding to the mouse double minutes clone2 (MDM2) binding site of p53 was shown by cell proliferation assay and MTT assay to have a proliferation-inhibiting effect on glioma cells. The polyarginine11R as a PTD, nuclear localization sequence (NLS), and laminin (Ln) fused to the p53 peptide corresponding to the MDM2 binding site (p53-NLS-Ln-11R) effectively penetrated the plasma membrane of the glioma cells and was translocated into the nucleus. At a 10 μM: concentration, this peptide inhibited the proliferation of human glioma cells, whether the p53 gene had mutated or not. These results suggest that this protein-transduction method using the p53-NSL-Ln-11R peptide may become a promising glioma therapy as an alternative gene therapy.
Collapse
Affiliation(s)
- Sachiko Yamada
- Department of Neurosurgery, Yokohama City University School of Medicine, Yokohama, Japan.
| | | | | |
Collapse
|
129
|
Camus S, Ménendez S, Fernandes K, Kua N, Liu G, Xirodimas DP, Lane DP, Bourdon JC. The p53 isoforms are differentially modified by Mdm2. Cell Cycle 2012; 11:1646-55. [PMID: 22487680 DOI: 10.4161/cc.20119] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The discovery that the single p53 gene encodes several different p53 protein isoforms has initiated a flurry of research into the function and regulation of these novel p53 proteins. Full-length p53 protein level is primarily regulated by the E3-ligase Mdm2, which promotes p53 ubiquitination and degradation. Here, we report that all of the novel p53 isoforms are ubiquitinated and degraded to varying degrees in an Mdm2-dependent and -independent manner, and that high-risk human papillomavirus can degrade some but not all of the novel isoforms, demonstrating that full-length p53 and the p53 isoforms are differentially regulated. In addition, we provide the first evidence that Mdm2 promotes the NEDDylation of p53β. Altogether, our data indicates that Mdm2 can distinguish between the p53 isoforms and modify them differently.
Collapse
|
130
|
Jenkins LMM, Durell SR, Mazur SJ, Appella E. p53 N-terminal phosphorylation: a defining layer of complex regulation. Carcinogenesis 2012; 33:1441-9. [PMID: 22505655 DOI: 10.1093/carcin/bgs145] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The p53 tumor suppressor is a critical component of the cellular response to stress. As it can inhibit cell growth, p53 is mutated or functionally inactivated in most tumors. A multitude of protein-protein interactions with transcriptional cofactors are central to p53-dependent responses. In its activated state, p53 is extensively modified in both the N- and C-terminal regions of the protein. These modifications, especially phosphorylation of serine and threonine residues in the N-terminal transactivation domain, affect p53 stability and activity by modulating the affinity of protein-protein interactions. Here, we review recent findings from in vitro and in vivo studies on the role of p53 N-terminal phosphorylation. These modifications can either positively or negatively affect p53 and add a second layer of complex regulation to the divergent interactions of the p53 transactivation domain.
Collapse
Affiliation(s)
- Lisa M Miller Jenkins
- Laboratory of Cell Biology, National Cancer Institute, NIH, 37 Convent Drive, Room 2140, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
131
|
Dancy BCR, Ming SA, Papazyan R, Jelinek CA, Majumdar A, Sun Y, Dancy BM, Drury WJ, Cotter RJ, Taverna SD, Cole PA. Azalysine analogues as probes for protein lysine deacetylation and demethylation. J Am Chem Soc 2012; 134:5138-48. [PMID: 22352831 PMCID: PMC3313494 DOI: 10.1021/ja209574z] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Reversible lysine acetylation and methylation regulate the function of a wide variety of proteins, including histones. Here, we have synthesized azalysine-containing peptides in acetylated and unacetylated forms as chemical probes of the histone deacetylases (HDAC8, Sir2Tm, and SIRT1) and the histone demethylase, LSD1. We have shown that the acetyl-azalysine modification is a fairly efficient substrate for the sirtuins, but a weaker substrate for HDAC8, a classical HDAC. In addition to deacetylation by sirtuins, the acetyl-azalysine analogue generates a novel ADP-ribose adduct that was characterized by mass spectrometry, Western blot analysis, and nuclear magnetic resonance spectroscopy. This peptide-ADP-ribose adduct is proposed to correspond to a derailed reaction intermediate, providing unique evidence for the direct 2'-hydroxyl attack on the O-alkylimidate intermediate that is formed in the course of sirtuin catalyzed deacetylation. An unacetylated azalysine-containing H3 peptide proved to be a potent inhibitor of the LSD1 demethylase, forming an FAD adduct characteristic of previously reported related structures, providing a new chemical probe for mechanistic analysis.
Collapse
Affiliation(s)
| | | | | | | | - Ananya Majumdar
- The Johns Hopkins Biomolecular NMR Center, Johns Hopkins University, Baltimore, MD 21218
| | | | | | | | | | | | - Philip A. Cole
- Corresponding Author: Philip A. Cole, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 725 N. Wolfe St, 316 Hunterian Building, Baltimore, MD 21205, Tel: 410-614-8849/614-0540, Fax: 410-955-3023,
| |
Collapse
|
132
|
Abstract
Regulation of the elongation phase of transcription by RNA polymerase II (Pol II) is utilized extensively to generate the pattern of mRNAs needed to specify cell types and to respond to environmental changes. After Pol II initiates, negative elongation factors cause it to pause in a promoter proximal position. These polymerases are poised to respond to the positive transcription elongation factor P-TEFb, and then enter productive elongation only under the appropriate set of signals to generate full-length properly processed mRNAs. Recent global analyses of Pol II and elongation factors, mechanisms that regulate P-TEFb involving the 7SK small nuclear ribonucleoprotein (snRNP), factors that control both the negative and positive elongation properties of Pol II, and the mRNA processing events that are coupled with elongation are discussed.
Collapse
Affiliation(s)
- Qiang Zhou
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, USA.
| | | | | |
Collapse
|
133
|
Portbury AL, Ronnebaum SM, Zungu M, Patterson C, Willis MS. Back to your heart: ubiquitin proteasome system-regulated signal transduction. J Mol Cell Cardiol 2012; 52:526-37. [PMID: 22085703 PMCID: PMC3294005 DOI: 10.1016/j.yjmcc.2011.10.023] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 10/28/2011] [Accepted: 10/30/2011] [Indexed: 12/19/2022]
Abstract
Awareness of the regulation of cell signaling by post-translational ubiquitination has emerged over the past 2 decades. Like phosphorylation, post-translational modification of proteins with ubiquitin can result in the regulation of numerous cellular functions, for example, the DNA damage response, apoptosis, cell growth, and the innate immune response. In this review, we discuss recently published mechanisms by which the ubiquitin proteasome system regulates key signal transduction pathways in the heart, including MAPK JNK, calcineurin, FOXO, p53, and estrogen receptors α and β. We then explore how ubiquitin proteasome system-specific regulation of these signal transduction pathways plays a role in the pathophysiology of common cardiac diseases, such as cardiac hypertrophy, heart failure, ischemia reperfusion injury, and diabetes. This article is part of a Special Section entitled "Post-translational Modification."
Collapse
Affiliation(s)
- Andrea L. Portbury
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
| | - Sarah M. Ronnebaum
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
| | - Makhosazane Zungu
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Cam Patterson
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
- Departments of Cell and Developmental Biology, Medicine, and Pharmacology, University of North Carolina, Chapel Hill, NC
| | - Monte S. Willis
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC USA
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC USA
| |
Collapse
|
134
|
Abstract
Mdm2 is an essential regulator of the p53 tumor suppressor. Mdm2 is modified at transcriptional, post-transcriptional, and post-translational levels to control p53 activity in normal versus stressed cells. Importantly, errors in these regulatory mechanisms can result in aberrant Mdm2 expression and failure to initiate programmed cell death in response to DNA damage. Such errors can have severe consequences as evidenced by tumor phenotypes resulting from amplification at the Mdm2 locus and changes in post-transcriptional and post-translational regulation of Mdm2. Although Mdm2 mediated inhibition of p53 is well characterized, Mdm2 interacts with many additional proteins and also targets many of these for proteosomal degradation. Mdm2 also has E3-ligase independent functions and p53-independent functions that have important implications for genome stability and cancer.
Collapse
Affiliation(s)
- Maurisa F Riley
- Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
135
|
Nelson LE, Valentine RJ, Cacicedo JM, Gauthier MS, Ido Y, Ruderman NB. A novel inverse relationship between metformin-triggered AMPK-SIRT1 signaling and p53 protein abundance in high glucose-exposed HepG2 cells. Am J Physiol Cell Physiol 2012; 303:C4-C13. [PMID: 22378745 DOI: 10.1152/ajpcell.00296.2011] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AMP-activated protein kinase (AMPK) and the NAD(+)-dependent histone/protein deacetylase sirtuin 1 (SIRT1) are metabolic sensors that can increase each other's activity. They are also both activated by the antidiabetic drug metformin and downregulated in the liver under conditions of nutrient excess (e.g., hyperglycemia, high-fat diet, obesity). In these situations, the abundance of the tumor suppressor p53 is increased; however, the relevance of this to the changes in AMPK and SIRT1 is not known. In the present study we investigated this question in HepG2 cells under high glucose conditions. Metformin induced activation of AMPK and SIRT1 and decreased p53 protein abundance. It also decreased triglyceride accumulation and cytosolic oxidative stress (a trigger for p53 accumulation) and increased the deacetylation of p53 at a SIRT1-targeted site. The decrease in p53 abundance caused by metformin was abolished by inhibition of murine double minute 2 (MDM2), a ubiquitin ligase that mediates p53 degradation, as well as by overexpression of a dominant-negative AMPK or a shRNA-mediated knockdown of SIRT1. In addition, overexpression of p53 decreased SIRT1 gene expression and protein abundance, as well as AMPK activity in metformin-treated cells. It also diminished the triglyceride-lowering action of metformin, an effect that was rescued by incubation with the SIRT1 activator SRT2183. Collectively, these findings suggest the existence of a novel reciprocal interaction between AMPK/SIRT1 and p53 that may have implications for the pathogenesis and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Lauren E Nelson
- Endocrinology, Diabetes, and Nutrition Section, Department of Medicine, Boston University School of Medicine, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
136
|
Li Y, Li X, Fan G, Fukushi JI, Matsumoto Y, Iwamoto Y, Zhu Y. Impairment of p53 acetylation by EWS-Fli1 chimeric protein in Ewing family tumors. Cancer Lett 2012; 320:14-22. [PMID: 22266186 DOI: 10.1016/j.canlet.2012.01.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 12/21/2011] [Accepted: 01/12/2012] [Indexed: 01/18/2023]
Abstract
The chromosomal translocation t(11;22)(q24;q12) yields the EWS-Fli1 fusion gene, which contributes to the development of Ewing Family Tumors (EFTs). Previous studies have shown the ability of EWS-Fli1 chimeric protein to silence p53 activity. Here we demonstrate that the introduction of EWS-Fli1 significantly inhibited p300-mediated acetylation of p53 at Lys-382 and depletion of EWS-Fli1 protein by small interfering RNAs (siRNA) in EFTs cells facilitated it in response to DNA damage. Furthermore, the deacetylation of p53 by EWS-Fli1 suppressed its transcriptional activity and enhanced mdm2-mediated p53 degradation. On the other hand, immunoprecipitation study shows that N-terminal region of EWS-Fli1 associated with histone deacetylase 1 (HDAC1) to forms a complex with p53. Knockdown of HDAC1, but not HDAC2 or HDAC3 protein restored the expression of p53 Lys-382 in EFTs cells. Overexpression of HDAC1 also significantly inhibited p53 transcriptional activity. Pharmacologic inhibitor of HDAC, trichostatin A (TSA) promoted p53-p300 interaction and recruitment of p53 Lys-382 to promoter regions of its target genes p21 and Puma, consequently inducing apoptosis and stabilizing the acetylation of p53 at Lys-382 together with the upregulation of p21 and Puma, which were impaired in EFTs cells after the knockdown of p53 expression. Our data indicate EWS-Fli1 might deacetylate p53 to inhibit its transcriptional function and protein stability via the recruitment of HDAC1. These results might elucidate a novel molecular mechanism about the abrogation of p53 pathway by EWS-Fli1 in EFTs pathogenesis.
Collapse
Affiliation(s)
- Yan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, China Medical University, Shenyang, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
137
|
Wang YE, Pernet O, Lee B. Regulation of the nucleocytoplasmic trafficking of viral and cellular proteins by ubiquitin and small ubiquitin-related modifiers. Biol Cell 2011; 104:121-38. [PMID: 22188262 PMCID: PMC3625690 DOI: 10.1111/boc.201100105] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 11/22/2011] [Indexed: 12/29/2022]
Abstract
Nucleocytoplasmic trafficking of many cellular proteins is regulated by nuclear import/export signals as well as post-translational modifications such as covalent conjugation of ubiquitin and small ubiquitin-related modifiers (SUMOs). Ubiquitination and SUMOylation are rapid and reversible ways to modulate the intracellular localisation and function of substrate proteins. These pathways have been co-opted by some viruses, which depend on the host cell machinery to transport their proteins in and out of the nucleus. In this review, we will summarise our current knowledge on the ubiquitin/SUMO-regulated nuclear/subnuclear trafficking of cellular proteins and describe examples of viral exploitation of these pathways.
Collapse
Affiliation(s)
- Yao E Wang
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|
138
|
Histone deacetylase inhibitors sensitize human non-small cell lung cancer cells to ionizing radiation through acetyl p53-mediated c-myc down-regulation. J Thorac Oncol 2011; 6:1313-9. [PMID: 21642861 DOI: 10.1097/jto.0b013e318220caff] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Histone deacetylase inhibitors (HDACIs) induce growth arrest and apoptosis in cancer cells. In addition to their intrinsic anticancer properties, HDACIs modulate cellular responses to ionizing radiation (IR). We examined the molecular mechanism(s) associated with the radiosensitizing effects of HDACIs in human lung cancer cells. METHODS Lung cancer cells were pretreated with the appropriate concentrations of suberoylanilide hydroxamic acid or trichostatin A. After 2 hours, cells were irradiated with various doses of γ-IR, and then we performed 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, fluorescence-activated cell sorting analysis, clonogenic assay, and Western blotting to detect cell viability or apoptosis and changes of specific proteins expression levels. RESULTS In this study, we showed that HDACIs (including suberoylanilide hydroxamic acid and trichostatin A) and IR synergistically trigger cell death in human non-small cell lung cancer cells. Cell viability and clonogenic survival were markedly decreased in cultures cotreated with HDACIs and IR. Interestingly, p53 acetylation at lysine 382 was significantly increased, and c-myc expression simultaneously down-regulated in cotreated cells. Radiosensitization by HDACIs was inhibited on transfection with small interfering RNA against p53 and c-myc overexpression, supporting the involvement of p53 and c-myc in this process. Furthermore, c-myc down-regulation and apoptotic cell death coinduced by IR and HDACI were suppressed in cells transfected with mutant K382R p53 and C135Y p53 displaying loss of acetylation at lysine 382 and DNA-binding activity, respectively. CONCLUSIONS Our results collectively demonstrate that the degree of radiosensitization by HDACIs is influenced by acetyl p53-mediated c-myc down-regulation.
Collapse
|
139
|
Positive regulation of p53 stability and activity by the deubiquitinating enzyme Otubain 1. EMBO J 2011; 31:576-92. [PMID: 22124327 DOI: 10.1038/emboj.2011.434] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2011] [Accepted: 11/08/2011] [Indexed: 12/19/2022] Open
Abstract
The ubiquitin (Ub)-proteasome system plays a pivotal role in the regulation of p53 protein stability and activity. p53 is ubiquitinated and destabilized by MDM2 and several other Ub E3s, whereas it is deubiquitinated and stabilized by Ub-specific protease (USP)7 and USP10. Here we show that the ovarian tumour domain-containing Ub aldehyde-binding protein 1 (Otub1) is a novel p53 regulator. Otub1 directly suppresses MDM2-mediated p53 ubiquitination in cells and in vitro. Overexpression of Otub1 drastically stabilizes and activates p53, leading to apoptosis and marked inhibition of cell proliferation in a p53-dependent manner. These effects are independent of its catalytic activity but require residue Asp88. Mutation of Asp88 to Ala (Otub1(D88A)) abolishes activity of Otub1 to suppress p53 ubiquitination. Further, wild-type Otub1 and its catalytic mutant (Otub1(C91S)), but not Otub1(D88A), bind to the MDM2 cognate E2, UbcH5, and suppress its Ub-conjugating activity in vitro. Overexpression of Otub1(D88A) or ablation of endogenous Otub1 by siRNA markedly impaired p53 stabilization and activation in response to DNA damage. Together, these results reveal a novel function for Otub1 in regulating p53 stability and activity.
Collapse
|
140
|
David Y, Ternette N, Edelmann MJ, Ziv T, Gayer B, Sertchook R, Dadon Y, Kessler BM, Navon A. E3 ligases determine ubiquitination site and conjugate type by enforcing specificity on E2 enzymes. J Biol Chem 2011; 286:44104-44115. [PMID: 21965653 DOI: 10.1074/jbc.m111.234559] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ubiquitin-conjugating enzymes (E2s) have a dominant role in determining which of the seven lysine residues of ubiquitin is used for polyubiquitination. Here we show that tethering of a substrate to an E2 enzyme in the absence of an E3 ubiquitin ligase is sufficient to promote its ubiquitination, whereas the type of the ubiquitin conjugates and the identity of the target lysine on the substrate are promiscuous. In contrast, when an E3 enzyme is introduced, a clear decision between mono- and polyubiquitination is made, and the conjugation type as well as the identity of the target lysine residue on the substrate becomes highly specific. These features of the E3 can be further regulated by auxiliary factors as exemplified by MDMX (Murine Double Minute X). In fact, we show that this interactor reconfigures MDM2-dependent ubiquitination of p53. Based on several model systems, we propose that although interaction with an E2 is sufficient to promote substrate ubiquitination the E3 molds the reaction into a specific, physiologically relevant protein modification.
Collapse
Affiliation(s)
- Yael David
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Nicola Ternette
- The Henry Wellcome Building for Molecular Physiology, Nuffield Department of Clinical Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Mariola J Edelmann
- The Henry Wellcome Building for Molecular Physiology, Nuffield Department of Clinical Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Tamar Ziv
- Smoler Proteomics Center, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Batya Gayer
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Rotem Sertchook
- Faculty of Biochemistry, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yakir Dadon
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Benedikt M Kessler
- The Henry Wellcome Building for Molecular Physiology, Nuffield Department of Clinical Medicine, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Ami Navon
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
141
|
Abstract
The ubiquitination pathway is a highly dynamic and coordinated process that regulates degradation as well as numerous processes of proteins within a cell. The p53 tumor suppressor and several factors in the pathway are regulated by ubiquitin as well as ubiquitin-like proteins. These modifications are critical for the function of p53 and control both the degradation of the protein as well as localization and activity. Importantly, more recent studies have identified deubiquitination enzymes that can specifically remove ubiquitin moieties from p53 or other factors in the pathway, and the reversible nature of this process adds yet another layer of regulatory control of p53. This review highlights the recent advances in our knowledge of ubiquitin and the p53 pathway.
Collapse
Affiliation(s)
- Christopher L Brooks
- Stemline Therapeutics, Inc., 1675 York Avenue, Suite 34-L, New York, NY 10128, USA.
| | | |
Collapse
|
142
|
Ma W, Lin Y, Xuan W, Iversen PL, Smith LJ, Benchimol S. Inhibition of p53 expression by peptide-conjugated phosphorodiamidate morpholino oligomers sensitizes human cancer cells to chemotherapeutic drugs. Oncogene 2011; 31:1024-33. [PMID: 21765469 DOI: 10.1038/onc.2011.300] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The p53 tumor suppressor gene encodes a transcription factor that is commonly mutated in cancer. Tumors arise when premalignant cells are unable to undergo p53-dependent apoptosis, cell cycle arrest or DNA repair. The p53-signaling pathway affects not only tumor development, but also the response of tumors to chemotherapeutic drugs. In this study, we use cell penetrating peptide conjugates of phosphorodiamidate morpholino oligomers (PPMOs) to inhibit p53 expression. We examine the functional properties of endogenous p53 isoforms that are produced upon PPMO-mediated inhibition of p53 translation and splicing, and report that loss of N-terminal or C-terminal sequences interferes with the transcriptional activity of p53. Importantly, we report that PPMO-mediated inhibition of p53 expression sensitizes human cancer cells with wild-type p53 to chemotherapeutic drugs.
Collapse
Affiliation(s)
- W Ma
- Department of Biology, York University, 4700 Keele St., Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
143
|
The impact of acetylation and deacetylation on the p53 pathway. Protein Cell 2011; 2:456-62. [PMID: 21748595 DOI: 10.1007/s13238-011-1063-9] [Citation(s) in RCA: 225] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 05/09/2011] [Indexed: 01/24/2023] Open
Abstract
The p53 tumor suppressor is a sequence-specific transcription factor that undergoes an abundance of post-translational modifications for its regulation and activation. Acetylation of p53 is an important reversible enzymatic process that occurs in response to DNA damage and genotoxic stress and is indispensible for p53 transcriptional activity. p53 was the first non-histone protein shown to be acetylated by histone acetyl transferases, and a number of more recent in vivo models have underscored the importance of this type of modification for p53 activity. Here, we review the current knowledge and recent findings of p53 acetylation and deacetylation and discuss the implications of these processes for the p53 pathway.
Collapse
|
144
|
Wang YV, Leblanc M, Fox N, Mao JH, Tinkum KL, Krummel K, Engle D, Piwnica-Worms D, Piwnica-Worms H, Balmain A, Kaushansky K, Wahl GM. Fine-tuning p53 activity through C-terminal modification significantly contributes to HSC homeostasis and mouse radiosensitivity. Genes Dev 2011; 25:1426-1438. [PMID: 21724834 PMCID: PMC3134085 DOI: 10.1101/gad.2024411] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 05/26/2011] [Indexed: 11/25/2022]
Abstract
Cell cycle regulation in hematopoietic stem cells (HSCs) is tightly controlled during homeostasis and in response to extrinsic stress. p53, a well-known tumor suppressor and transducer of diverse stress signals, has been implicated in maintaining HSC quiescence and self-renewal. However, the mechanisms that control its activity in HSCs, and how p53 activity contributes to HSC cell cycle control, are poorly understood. Here, we use a genetically engineered mouse to show that p53 C-terminal modification is critical for controlling HSC abundance during homeostasis and HSC and progenitor proliferation after irradiation. Preventing p53 C-terminal modification renders mice exquisitely radiosensitive due to defects in HSC/progenitor proliferation, a critical determinant for restoring hematopoiesis after irradiation. We show that fine-tuning the expression levels of the cyclin-dependent kinase inhibitor p21, a p53 target gene, contributes significantly to p53-mediated effects on the hematopoietic system. These results have implications for understanding cell competition in response to stresses involved in stem cell transplantation, recovery from adverse hematologic effects of DNA-damaging cancer therapies, and development of radioprotection strategies.
Collapse
Affiliation(s)
- Yunyuan V. Wang
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Mathias Leblanc
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Norma Fox
- Department of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Jian-Hua Mao
- The Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | - Kelsey L. Tinkum
- Department of Cell Biology and Physiology, Washington University Medical School, St. Louis, Missouri 63110, USA
- Mallinckrodt Institute of Radiology, Washington University Medical School, St. Louis, Missouri 63110, USA
- The BRIGHT Institute, Washington University Medical School, St. Louis, Missouri 63110, USA
| | - Kurt Krummel
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Dannielle Engle
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - David Piwnica-Worms
- Mallinckrodt Institute of Radiology, Washington University Medical School, St. Louis, Missouri 63110, USA
- The BRIGHT Institute, Washington University Medical School, St. Louis, Missouri 63110, USA
- Department of Developmental Biology, Washington University Medical School, St. Louis, Missouri 63110, USA
| | - Helen Piwnica-Worms
- Department of Cell Biology and Physiology, Washington University Medical School, St. Louis, Missouri 63110, USA
- The BRIGHT Institute, Washington University Medical School, St. Louis, Missouri 63110, USA
- Department of Medicine, Washington University Medical School, St. Louis, Missouri 63110, USA
- the Howard Hughes Medical Institute, St. Louis, Missouri 63130, USA
| | - Allan Balmain
- The Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94158, USA
| | - Kenneth Kaushansky
- Department of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Geoffrey M. Wahl
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| |
Collapse
|
145
|
E3 ubiquitin ligase Hades negatively regulates the exonuclear function of p53. Cell Death Differ 2011; 18:1865-75. [PMID: 21597459 DOI: 10.1038/cdd.2011.57] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Following DNA damage, p53 translocates to the cytoplasm and mitochondria, where it triggers transcription-independent apoptosis by binding to Bcl-2 family proteins. However, little is known about how this exonuclear function of p53 is regulated. Here, we identify and characterize a p53-interacting protein called Hades, an E3 ligase that interacts with p53 in the mitochondria. Hades reduces p53 stability via a mechanism that requires its RING-finger domain with ubiquitin ligase activity. Hades polyubiquitinates p53 in vitro independent of Mdm2 and targets a critical lysine residue in p53 (lysine 24) distinct from those targeted by Mdm2. Hades inhibits a p53-dependent mitochondrial cell death pathway by inhibiting p53 and Bcl-2 interactions. These findings show that Hades-mediated p53 ubiquitination is a novel mechanism for negatively regulating the exonuclear function of p53.
Collapse
|
146
|
Ozaki T, Nakagawara A. Role of p53 in Cell Death and Human Cancers. Cancers (Basel) 2011; 3:994-1013. [PMID: 24212651 PMCID: PMC3756401 DOI: 10.3390/cancers3010994] [Citation(s) in RCA: 473] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Revised: 02/22/2011] [Accepted: 02/22/2011] [Indexed: 02/08/2023] Open
Abstract
p53 is a nuclear transcription factor with a pro-apoptotic function. Since over 50% of human cancers carry loss of function mutations in p53 gene, p53 has been considered to be one of the classical type tumor suppressors. Mutant p53 acts as the dominant-negative inhibitor toward wild-type p53. Indeed, mutant p53 has an oncogenic potential. In some cases, malignant cancer cells bearing p53 mutations display a chemo-resistant phenotype. In response to a variety of cellular stresses such as DNA damage, p53 is induced to accumulate in cell nucleus to exert its pro-apoptotic function. Activated p53 promotes cell cycle arrest to allow DNA repair and/or apoptosis to prevent the propagation of cells with serious DNA damage through the transactivation of its target genes implicated in the induction of cell cycle arrest and/or apoptosis. Thus, the DNA-binding activity of p53 is tightly linked to its tumor suppressive function. In the present review article, we describe the regulatory mechanisms of p53 and also p53-mediated therapeutic strategies to cure malignant cancers.
Collapse
Affiliation(s)
- Toshinori Ozaki
- Laboratory of Anti-tumor Research, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuoh-ku, Chiba 260-8717, Japan; E-Mail:
| | - Akira Nakagawara
- Division of Biochemistry and Laboratory of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuoh-ku, Chiba 260-8717, Japan
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-43-264-5431; Fax: +81-43-265-4459
| |
Collapse
|
147
|
Wu H, Pomeroy SL, Ferreira M, Teider N, Mariani J, Nakayama KI, Hatakeyama S, Tron VA, Saltibus LF, Spyracopoulos L, Leng RP. UBE4B promotes Hdm2-mediated degradation of the tumor suppressor p53. Nat Med 2011; 17:347-55. [PMID: 21317885 DOI: 10.1038/nm.2283] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 11/30/2010] [Indexed: 11/09/2022]
Abstract
The TP53 gene (encoding the p53 tumor suppressor) is rarely mutated, although frequently inactivated, in medulloblastoma and ependymoma. Recent work in mouse models showed that the loss of p53 accelerated the development of medulloblastoma. The mechanism underlying p53 inactivation in human brain tumors is not completely understood. We show that ubiquitination factor E4B (UBE4B), an E3 and E4 ubiquitin ligase, physically interacts with p53 and Hdm2 (also known as Mdm2 in mice). UBE4B promotes p53 polyubiquitination and degradation and inhibits p53-dependent transactivation and apoptosis. Notably, silencing UBE4B expression impairs xenotransplanted tumor growth in a p53-dependent manner and overexpression of UBE4B correlates with decreased expression of p53 in these tumors. We also show that UBE4B overexpression is often associated with amplification of its gene in human brain tumors. Our data indicate that amplification and overexpression of UBE4B represent previously undescribed molecular mechanisms of inactivation of p53 in brain tumors.
Collapse
Affiliation(s)
- Hong Wu
- Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Broemer M, Tenev T, Rigbolt KTG, Hempel S, Blagoev B, Silke J, Ditzel M, Meier P. Systematic in vivo RNAi analysis identifies IAPs as NEDD8-E3 ligases. Mol Cell 2010; 40:810-22. [PMID: 21145488 DOI: 10.1016/j.molcel.2010.11.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Revised: 08/02/2010] [Accepted: 09/13/2010] [Indexed: 11/24/2022]
Abstract
The intimate relationship between mediators of the ubiquitin (Ub)-signaling system and human diseases has sparked profound interest in how Ub influences cell death and survival. While the consequence of Ub attachment is intensely studied, little is known with regards to the effects of other Ub-like proteins (UBLs), and deconjugating enzymes that remove the Ub or UBL adduct. Systematic in vivo RNAi analysis identified three NEDD8-specific isopeptidases that, when knocked down, suppress apoptosis. Consistent with the notion that attachment of NEDD8 prevents cell death, genetic ablation of deneddylase 1 (DEN1) suppresses apoptosis. Unexpectedly, we find that Drosophila and human inhibitor of apoptosis (IAP) proteins can function as E3 ligases of the NEDD8 conjugation pathway, targeting effector caspases for neddylation and inactivation. Finally, we demonstrate that DEN1 reverses this effect by removing the NEDD8 modification. Altogether, our findings indicate that IAPs not only modulate cellular processes via ubiquitylation but also through attachment of NEDD8, thereby extending the complexity of IAP-mediated signaling.
Collapse
Affiliation(s)
- Meike Broemer
- The Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, Mary-Jean Mitchell Green Building, Chester Beatty Laboratories, Fulham Road, London SW3 6JB, UK.
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Ozaki T, Nakagawara A. p53: the attractive tumor suppressor in the cancer research field. J Biomed Biotechnol 2010; 2011:603925. [PMID: 21188172 PMCID: PMC3004423 DOI: 10.1155/2011/603925] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 10/07/2010] [Indexed: 01/16/2023] Open
Abstract
p53 is one of the most studied tumor suppressors in the cancer research field. Of note, over 50% of human tumors carry loss of function mutations, and thus p53 has been considered to be a classical Knudson-type tumor suppressor. From the functional point of view, p53 is a nuclear transcription factor to transactivate a variety of its target genes implicated in the induction of cell cycle arrest, DNA repair, and apoptotic cell death. In response to cellular stresses such as DNA damage, p53 is activated and promotes cell cycle arrest followed by the replacement of DNA lesions and/or apoptotic cell death. Therefore, p53 is able to maintain the genomic integrity to prevent the accumulation of genetic alterations, and thus stands at a crossroad between cell survival and cell death. In this paper, we describe a variety of molecular mechanisms behind the regulation of p53.
Collapse
Affiliation(s)
- Toshinori Ozaki
- Laboratory of Anti-Tumor Research, Chiba Cancer Center Research Institute, Chiba 260-8717, Japan
| | - Akira Nakagawara
- Laboratory of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, 666-2 Nitona, Chuoh-ku, Chiba 260-8717, Japan
| |
Collapse
|
150
|
Fujita K, Horikawa I, Mondal AM, Miller Jenkins LM, Appella E, Vojtesek B, Bourdon JC, Lane DP, Harris CC. Positive feedback between p53 and TRF2 during telomere-damage signalling and cellular senescence. Nat Cell Biol 2010; 12:1205-12. [PMID: 21057505 PMCID: PMC3470109 DOI: 10.1038/ncb2123] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 09/24/2010] [Indexed: 12/12/2022]
Abstract
The telomere-capping complex shelterin protects functional telomeres and prevents the initiation of unwanted DNA-damage-response pathways. At the end of cellular replicative lifespan, uncapped telomeres lose this protective mechanism and DNA-damage signalling pathways are triggered that activate p53 and thereby induce replicative senescence. Here, we identify a signalling pathway involving p53, Siah1 (a p53-inducible E3 ubiquitin ligase) and TRF2 (telomere repeat binding factor 2; a component of the shelterin complex). Endogenous Siah1 and TRF2 were upregulated and downregulated, respectively, during replicative senescence with activated p53. Experimental manipulation of p53 expression demonstrated that p53 induces Siah1 and represses TRF2 protein levels. The p53-dependent ubiquitylation and proteasomal degradation of TRF2 are attributed to the E3 ligase activity of Siah1. Knockdown of Siah1 stabilized TRF2 and delayed the onset of cellular replicative senescence, suggesting a role for Siah1 and TRF2 in p53-regulated senescence. This study reveals that p53, a downstream effector of telomere-initiated damage signalling, also functions upstream of the shelterin complex.
Collapse
Affiliation(s)
- Kaori Fujita
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4258, USA
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4258, USA
| | - Abdul M. Mondal
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4258, USA
| | - Lisa M. Miller Jenkins
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4256, USA
| | - Ettore Appella
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4256, USA
| | - Borivoj Vojtesek
- Masaryk Memorial Cancer Institute, Zluty Kopec 7, 65653 Brno, Czech Republic
| | - Jean-Christophe Bourdon
- University of Dundee, Ninewells Hospital, Dept. of Surgery and Molecular Oncology, Inserm-European Associated Laboratory, Dundee, DD1 9SY, UK
| | - David P. Lane
- University of Dundee, Ninewells Hospital, Dept. of Surgery and Molecular Oncology, Inserm-European Associated Laboratory, Dundee, DD1 9SY, UK
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, Maryland 20892-4258, USA
| |
Collapse
|