201
|
Parallel analysis of tetramerization domain mutants of the human p53 protein using PCR colonies. Genomic Med 2007; 1:113-24. [PMID: 18923936 DOI: 10.1007/s11568-007-9011-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Accepted: 08/01/2007] [Indexed: 01/21/2023] Open
Abstract
A highly-parallel yeast functional assay, capable of screening approximately 100-1,000 mutants in parallel and designed to screen the activity of transcription activator proteins, was utilized to functionally characterize tetramerization domain mutants of the human p53 transcription factor and tumor suppressor protein. A library containing each of the 19 possible single amino acid substitutions (57 mutants) at three positions in the tetramerization domain of the human p53 protein, was functionally screened in Saccharomyces cerevisiae. Amino acids Leu330 and Ile332, whose side chains form a portion of a hydrophobic pocket that stabilizes the active p53 tetramer, were found to tolerate most hydrophobic amino acid substitutions while hydrophilic substitutions resulted in the inactivation of the protein. Amino acid Gln331 tolerated essentially all mutations. Importantly, highly parallel mutagenesis and cloning techniques were utilized which, in conjunction with recently reported highly parallel DNA sequencing methods, would be capable of increasing throughput an additional 2-3 orders of magnitude.
Collapse
|
202
|
Das S, Raj L, Zhao B, Bernstein A, Aaronson SA, Lee SW. Hzf Determines cell survival upon genotoxic stress by modulating p53 transactivation. Cell 2007; 130:624-37. [PMID: 17719541 PMCID: PMC2779720 DOI: 10.1016/j.cell.2007.06.013] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 05/01/2007] [Accepted: 06/05/2007] [Indexed: 01/30/2023]
Abstract
A critical unresolved issue about the genotoxic stress response is how the resulting activation of the p53 tumor suppressor can lead either to cell-cycle arrest and DNA repair or to apoptosis. We show here that hematopoietic zinc finger (Hzf), a zinc-finger-containing p53 target gene, modulates p53 transactivation functions in an autoregulatory feedback loop. Hzf is induced by p53 and binds to its DNA-binding domain, resulting in preferential transactivation of proarrest p53 target genes over its proapoptotic target genes. Thus, p53 activation results in cell-cycle arrest in Hzf wild-type MEFs, while in Hzf(-/-) MEFs, apoptosis is induced. Exposure of Hzf null mice to ionizing radiation resulted in enhanced apoptosis in several organs, as compared to in wild-type mice. These findings provide novel insights into the regulation of p53 transactivation function and suggest that Hzf functions as a key player in regulating cell fate decisions in response to genotoxic stress.
Collapse
Affiliation(s)
- Sanjeev Das
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Lakshmi Raj
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Bo Zhao
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, New York 10029, USA
| | - Alan Bernstein
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, Canada M5G IX5
| | - Stuart A. Aaronson
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, New York 10029, USA
| | - Sam W. Lee
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| |
Collapse
|
203
|
Wang J, Liu YX, Hande MP, Wong AC, Jin YJ, Yin Y. TAp73 is a downstream target of p53 in controlling the cellular defense against stress. J Biol Chem 2007; 282:29152-62. [PMID: 17693405 DOI: 10.1074/jbc.m703408200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TAp73 is a p53 tumor suppressor gene homologue that is known to be mainly involved in apoptosis. We report here that TAp73 is necessary for the cellular response to oxidative stress and that TAp73 functions as a downstream target of p53 in this process. We show that p53 physically interacts with the TAp73 promoter under stress conditions that lead to cell death. Particularly, p53 binds to a palindromic site in the TAp73 promoter, activates the promoter of TAp73, and selectively induces TAp73 transcription. TAp73 expression is highly increased under oxidative stress in a p53-dependent manner. Furthermore, knock-down of TAp73 expression inhibits the cellular apoptotic response to oxidative damage. In contrast, the ectopic expression of TAp73 in p53(-/-) mouse embryonic fibroblasts induces oxidative cell death. Our findings demonstrate that p53 is a direct transcriptional regulator of TAp73. Our data reveal a new pathway for cellular protection against oxidative damage and provide evidence that TAp73 is a stress-response gene and a downstream effector in the p53 pathway.
Collapse
Affiliation(s)
- Jianli Wang
- Department of Radiation Oncology, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | |
Collapse
|
204
|
Tidow H, Melero R, Mylonas E, Freund SMV, Grossmann JG, Carazo JM, Svergun DI, Valle M, Fersht AR. Quaternary structures of tumor suppressor p53 and a specific p53 DNA complex. Proc Natl Acad Sci U S A 2007; 104:12324-9. [PMID: 17620598 PMCID: PMC1941468 DOI: 10.1073/pnas.0705069104] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Indexed: 12/31/2022] Open
Abstract
The homotetrameric tumor suppressor p53 consists of folded core and tetramerization domains, linked and flanked by intrinsically disordered segments that impede structure analysis by x-ray crystallography and NMR. Here, we solved the quaternary structure of human p53 in solution by a combination of small-angle x-ray scattering, which defined its shape, and NMR, which identified the core domain interfaces and showed that the folded domains had the same structure in the intact protein as in fragments. We combined the solution data with electron microscopy on immobilized samples that provided medium resolution 3D maps. Ab initio and rigid body modeling of scattering data revealed an elongated cross-shaped structure with a pair of loosely coupled core domain dimers at the ends, which are accessible for binding to DNA and partner proteins. The core domains in that open conformation closed around a specific DNA response element to form a compact complex whose structure was independently determined by electron microscopy. The structure of the DNA complex is consistent with that of the complex of four separate core domains and response element fragments solved by x-ray crystallography and contacts identified by NMR. Electron microscopy on the conformationally mobile, unbound p53 selected a minor compact conformation, which resembled the closed conformation, from the ensemble of predominantly open conformations. A multipronged structural approach could be generally useful for the structural characterization of the rapidly growing number of multidomain proteins with intrinsically disordered regions.
Collapse
Affiliation(s)
- Henning Tidow
- *Medical Research Council Centre for Protein Engineering, Hills Road, Cambridge CB2 0QH, United Kingdom
| | - Roberto Melero
- Centro Nacional de Biotecnología, Darwin 3, Cantoblanco 28049 Madrid, Spain
| | - Efstratios Mylonas
- European Molecular Biology Laboratory, Hamburg Outstation, Notkestrasse 85, 22603 Hamburg, Germany
| | - Stefan M. V. Freund
- *Medical Research Council Centre for Protein Engineering, Hills Road, Cambridge CB2 0QH, United Kingdom
| | - J. Guenter Grossmann
- Molecular Biophysics Group, Council for the Central Laboratory of the Research Councils (CCLRC) Daresbury Laboratory, Warrington, Cheshire WA4 4AD, United Kingdom
| | - José María Carazo
- Centro Nacional de Biotecnología, Darwin 3, Cantoblanco 28049 Madrid, Spain
| | - Dmitri I. Svergun
- European Molecular Biology Laboratory, Hamburg Outstation, Notkestrasse 85, 22603 Hamburg, Germany
- Institute of Crystallography, Russian Academy of Sciences, Leninsky pr. 59, 117333 Moscow, Russia; and
| | - Mikel Valle
- Centro Nacional de Biotecnología, Darwin 3, Cantoblanco 28049 Madrid, Spain
- **CIC-bioGUNE, Parque Tecnológico de Bizkaia, 48160 Derio, Spain
| | - Alan R. Fersht
- *Medical Research Council Centre for Protein Engineering, Hills Road, Cambridge CB2 0QH, United Kingdom
| |
Collapse
|
205
|
Klassen SS, Rabkin SW. The Role of p53 in Nitric Oxide–Induced Cardiomyocyte Cell Death. DNA Cell Biol 2007; 26:465-75. [PMID: 17630850 DOI: 10.1089/dna.2007.0576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The role of p53 in mediating nitric oxide (NO)-induced cell death remains uncertain. The exogenous NO donor S-nitrosoglutathione (GSNO) produced a concentration-dependent reduction in cell viability in embryonic chick cardiomyocytes in culture. Western blotting and immunocytochemistry for p53 showed that p53 was increased in whole cell lysates by GSNO: 0.001 mM GSNO led to 1.3 +/- 0.5-fold increase compared to control, and significantly (p < 0.05) increased to 1.6 +/- 0.2-fold after 0.01 mM GSNO. Higher GSNO concentrations did not further increase p53 protein expression despite producing significant increases in cell death. The p53 inhibitor pifithrin did not block GSNO-induced cell death. GSNO induced morphological changes of DNA fragmentation, nuclear condensation, and cell shrinkage. Pifithrin failed to block these morphologic changes, while it antagonized the similar cellular changes induced by adriamycin, which operates in part through p53. NO induced a concentration-dependent DNA damage. When assessed by the comet assay, the damage was 2.1 +/- 0.3-fold and 2.6 +/- 0.5-fold more than the control following 0.01 mM and 1.0 mM GSNO treatments, respectively. The DNA damage was not reduced by treatment with the pifithrin, which markedly reduced DNA damage induced by adriamycin. There was no p53 translocation to mitochondria, any major cytochrome c release from mitochondria, or change in mitochondrial membrane potential. Furthermore, cyclosporin A, which inhibits mitochondrial pore opening and cytochrome c loss, did not alter NO-induced cell death. Translocation of p53 from the cytosol to the nucleus occurred with a maximal increase of 2.9-fold in the nucleus following 1.0 mM GSNO for 24 h. These data indicate that in cardiomyocytes, NO induced marked DNA damage and translocation of p53 to the nucleus, suggesting that p53 is involved in the cellular response to NO, perhaps to modulate the genomic response to NO-induced cellular toxicity. NO-induced cell death, however, operates through p53-independent pathways, including a mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Shaun S Klassen
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
206
|
Abstract
Convergent studies demonstrated that p53 regulates homologous recombination (HR) independently of its classic tumour-suppressor functions in transcriptionally transactivating cellular target genes that are implicated in growth control and apoptosis. In this review, we summarise the analyses of the involvement of p53 in spontaneous and double-strand break (DSB)-triggered HR and in alternative DSB repair routes. Molecular characterisation indicated that p53 controls the fidelity of Rad51-dependent HR and represses aberrant processing of replication forks after stalling at unrepaired DNA lesions. These findings established a genome stabilising role of p53 in counteracting error-prone DSB repair. However, recent work has also unveiled a stimulatory role for p53 in topoisomerase I-induced recombinative repair events that may have implications for a gain-of-function phenotype of cancer-related p53 mutants. Additional evidence will be discussed which suggests that p53 and/or p53-regulated gene products also contribute to nucleotide excision, base excision, and mismatch repair.
Collapse
Affiliation(s)
- S A Gatz
- Universitätsklinik für Kinder- und Jugendmedizin, Eythstr. 24, 89075 Ulm, Germany
| | | |
Collapse
|
207
|
Raver-Shapira N, Marciano E, Meiri E, Spector Y, Rosenfeld N, Moskovits N, Bentwich Z, Oren M. Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol Cell 2007; 26:731-43. [PMID: 17540598 DOI: 10.1016/j.molcel.2007.05.017] [Citation(s) in RCA: 1030] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 05/05/2007] [Accepted: 05/17/2007] [Indexed: 02/07/2023]
Abstract
p53 is a potent tumor suppressor, whose biological effects are largely due to its function as a transcriptional regulator. Here we report that, in addition to regulating the expression of hundreds of protein-coding genes, p53 also modulates the levels of microRNAs (miRNAs). Specifically, p53 can induce expression of microRNA-34a (miR-34a) in cultured cells as well as in irradiated mice, by binding to a perfect p53 binding site located within the gene that gives rise to miR-34a. Processing of the primary transcript into mature miR-34a involves the excision of a 30 kb intron. Notably, inactivation of miR-34a strongly attenuates p53-mediated apoptosis in cells exposed to genotoxic stress, whereas overexpression of miR-34a mildly increases apoptosis. Hence, miR-34a is a direct proapoptotic transcriptional target of p53 that can mediate some of p53's biological effects. Perturbation of miR-34a expression, as occurs in some human cancers, may thus contribute to tumorigenesis by attenuating p53-dependent apoptosis.
Collapse
Affiliation(s)
- Nina Raver-Shapira
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | | | |
Collapse
|
208
|
Kochetkov DV, Ilyinskaya GV, Komarov PG, Strom E, Agapova LS, Ivanov AV, Budanov AV, Frolova EI, Chumakov PM. Transcriptional inhibition of the human papilloma virus reactivates tumor suppressor p53 in cervical carcinoma cells. Mol Biol 2007. [DOI: 10.1134/s0026893307030120] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
209
|
Karni-Schmidt O, Friedler A, Zupnick A, McKinney K, Mattia M, Beckerman R, Bouvet P, Sheetz M, Fersht A, Prives C. Energy-dependent nucleolar localization of p53 in vitro requires two discrete regions within the p53 carboxyl terminus. Oncogene 2007; 26:3878-91. [PMID: 17237827 DOI: 10.1038/sj.onc.1210162] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 09/22/2006] [Accepted: 10/23/2006] [Indexed: 12/19/2022]
Abstract
The p53 tumor suppressor is a nucleocytoplasmic shuttling protein that is found predominantly in the nucleus of cells. In addition to mutation, abnormal p53 cellular localization is one of the mechanisms that inactivate p53 function. To further understand features of p53 that contribute to the regulation of its trafficking within the cell, we analysed the subnuclear localization of wild-type and mutant p53 in human cells that were either permeabilized with detergent or treated with the proteasome inhibitor MG132. We, here, show that either endogenously expressed or exogenously added p53 protein localizes to the nucleolus in detergent-permeabilized cells in a concentration- and ATP hydrolysis-dependent manner. Two discrete regions within the carboxyl terminus of p53 are essential for nucleolar localization in permeabilized cells. Similarly, localization of p53 to the nucleolus after proteasome inhibition in unpermeabilized cells requires sequences within the carboxyl terminus of p53. Interestingly, genotoxic stress markedly decreases the association of p53 with the nucleolus, and phosphorylation of p53 at S392, a site that is modified by such stress, partially impairs its nucleolar localization. The possible significance of these findings is discussed.
Collapse
Affiliation(s)
- O Karni-Schmidt
- Department of Biological Sciences, Columbia University, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Yang D, Qi Y, Chen Q, Wang Z, Jin X, Gao J, Fu J, Xiao X, Zhou Z. The over-expression of p53 H179Y residue mutation causes the increase of cyclin A1 and Cdk4 expression in HELF cells. Mol Cell Biochem 2007; 304:219-26. [PMID: 17530187 DOI: 10.1007/s11010-007-9503-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Accepted: 04/27/2007] [Indexed: 01/10/2023]
Abstract
Down-regulation of p53 expression has been found in a broad range of human cancers and cell proliferation disorders, indicating that p53 plays a key role in cell cycle regulation and tumor suppression. In our current study, we transfected human embryonic lung fibroblast (HELF) cells with pcDNA3-wild-type p53 (pcDNA3-wtp53) plasmid, or pcDNA3-H179Y-mutated p53 (pcDNA3-mtp53) plasmid that mimics the mutation found in some human lung tumors, and further studied the role of p53 in the regulation of cell proliferation. Over expression of wild-type p53 caused cell cycle arrest at G1 phase with reduced cell size, decreased expression of cyclin D3, cyclin E, Cdk2 and Cdk4, and increased expression of p21. In contrast, over expression of H179Y-mutant p53 promoted G1 to S phase transition with enlarged cell size and increased cyclin A1 and Cdk4 expression in HELF cells. These results indicate that mutation at the p53 H179Y residue up-regulates cyclin A1 and Cdk4 expression, and promotes HELF cell proliferation.
Collapse
Affiliation(s)
- Di Yang
- Department of Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100094, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
211
|
Rad9 modulates the P21WAF1 pathway by direct association with p53. BMC Mol Biol 2007; 8:37. [PMID: 17511890 PMCID: PMC1885445 DOI: 10.1186/1471-2199-8-37] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Accepted: 05/21/2007] [Indexed: 11/24/2022] Open
Abstract
Background Previous studies suggest that human RAD9 (hRad9), encoding a DNA damage checkpoint molecule, which is frequently amplified in epithelial tumor cells of breast, lung, head and neck cancer, participates in regulation of the tumor suppressor p53-dependent transactivation of pro-survival P21WAF1. This study examined the exact mechanism of the hRad9 function, especially through the phosphorylation of the C-terminus, in the transcription regulation of P21WAF1. Results The transfection of phosphorylation-defective hRAD9 mutants of C-terminus resulted in reduction of the p53-dependent P21WAF1 transactivation; the knockdown of total hRad9 elicited an increased P21WAF1 mRNA expression. Immunoprecipitation and a ChIP assay showed that hRad9 and p53 formed a complex and both were associated with two p53-consensus DNA-binding sequences in the 5' region of P21WAF1 gene. The association was reduced in the experiment of phosphorylation-defective hRAD9 mutants. Conclusion The present study indicates the direct involvement of hRad9 in the p53-dependent P21WAF1 transcriptional mechanism, presumably via the phosphorylation sites, and alterations of the hRad9 pathway might therefore contribute to the perturbation of checkpoint activation in cancer cells.
Collapse
|
212
|
Zhao Y, Katzman RB, Delmolino LM, Bhat I, Zhang Y, Gurumurthy CB, Germaniuk-Kurowska A, Reddi HV, Solomon A, Zeng MS, Kung A, Ma H, Gao Q, Dimri G, Stanculescu A, Miele L, Wu L, Griffin JD, Wazer DE, Band H, Band V. The notch regulator MAML1 interacts with p53 and functions as a coactivator. J Biol Chem 2007; 282:11969-81. [PMID: 17317671 DOI: 10.1074/jbc.m608974200] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Members of the evolutionarily conserved Mastermind (MAM) protein family, including the three related mammalian Mastermind-like (MAML) proteins MAML1-3, function as crucial coactivators of Notch-mediated transcriptional activation. Given the recent evidence of cross-talk between the p53 and Notch signal transduction pathways, we have investigated whether MAML1 may also be a transcriptional coactivator of p53. Indeed, we show here that MAML1 is able to interact with p53. We show that MAML1-p53 interaction involves the N-terminal region of MAML1 and the DNA-binding domain of p53, and we use a chromatin immunoprecipitation assay to show that MAML1 is part of the activator complex that binds to native p53-response elements within the promoter of the p53 target genes. Overexpression of wild-type MAML1 as well as a mutant, defective in Notch signaling, enhanced the p53-dependent gene induction in mammalian cells, whereas MAML1 knockdown reduced the p53-dependent gene expression. MAML1 increases the half-life of p53 protein and enhances its phosphorylation/acetylation upon DNA damage of cells. Finally, RNA interference-mediated knockdown of the single Caenorhabditis elegans MAML homolog, Lag-3, led to substantial abrogation of p53-mediated germ-cell apoptotic response to DNA damage and markedly reduced the expression of Ced-13 and Egl-1, downstream pro-apoptotic targets of the C. elegans p53 homolog Cep-1. Thus, we present evidence for a novel coactivator function of MAML1 for p53, independent of its function as a coactivator of Notch signaling pathway.
Collapse
Affiliation(s)
- Yongtong Zhao
- Division of Cancer Biology, Department of Medicine, ENH Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, Illinois 60201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Cui R, Widlund HR, Feige E, Lin JY, Wilensky DL, Igras VE, D'Orazio J, Fung CY, Schanbacher CF, Granter SR, Fisher DE. Central role of p53 in the suntan response and pathologic hyperpigmentation. Cell 2007; 128:853-64. [PMID: 17350573 DOI: 10.1016/j.cell.2006.12.045] [Citation(s) in RCA: 465] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Revised: 11/23/2006] [Accepted: 12/28/2006] [Indexed: 10/23/2022]
Abstract
UV-induced pigmentation (suntanning) requires induction of alpha-melanocyte-stimulating hormone (alpha-MSH) secretion by keratinocytes. alpha-MSH and other bioactive peptides are cleavage products of pro-opiomelanocortin (POMC). Here we provide biochemical and genetic evidence demonstrating that UV induction of POMC/MSH in skin is directly controlled by p53. Whereas p53 potently stimulates the POMC promoter in response to UV, the absence of p53, as in knockout mice, is associated with absence of the UV-tanning response. The same pathway produces beta-endorphin, another POMC derivative, which potentially contributes to sun-seeking behaviors. Furthermore, several instances of UV-independent pathologic pigmentation are shown to involve p53 "mimicking" the tanning response. p53 thus functions as a sensor/effector for UV pigmentation, which is a nearly constant environmental exposure. Moreover, this pathway is activated in numerous conditions of pathologic pigmentation and thus mimics the tanning response.
Collapse
Affiliation(s)
- Rutao Cui
- Melanoma Program in Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Contasta I, Pellegrini P, Berghella AM, Del Beato T, Adorno D. Colon cancer and gene alterations: their immunological implications and suggestions for prognostic indices and improvements in biotherapy. Cancer Biother Radiopharm 2007; 21:488-505. [PMID: 17105421 DOI: 10.1089/cbr.2006.21.488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Studies have shown that changes occur in c-Ki-ras, p53, and Bcl2 gene structure and function during the various stages of human colon carcinogenesis. Alterations of these genes are responsible for the establishment of a state of continuous stimulus for cell division and apoptotic inhibition at physiological and pharmacological levels. This paper focuses on the results of our research aimed at investigating how these gene alterations influence tumoral mechanisms on an immunological level and how immunological parameters can be used as prognostic markers for the passage of normal tissue to adenoma and adenoma to carcinoma. Overall, our data suggest that an alteration in the c-Ki-ras gene results in a switch to a suppressive type of immune response, determining an impairment of immune cell activation at both antigen- presenting-cell and T-cell levels. c-Ki-ras gene mutations, p53 deletions, and Bc12 expression, on the other hand, can be used as prognostic markers for the passage of normal tissue to adenoma and adenoma to carcinoma. The p53 oncogene does not appear to impair patients' immunological response further. In conclusion, an evaluation of c-Ki-ras, rather than p53 gene alterations, would seem to be more relevant in colon cancer prevention programs and biotherapy improvement.
Collapse
Affiliation(s)
- Ida Contasta
- CNR Institute of Organ Transplantation and Immunocytology (ITOI), L'Aquila, Italy.
| | | | | | | | | |
Collapse
|
215
|
Tang X, Molina M, Amar S. p53 short peptide (p53pep164) regulates lipopolysaccharide-induced tumor necrosis factor-alpha factor/cytokine expression. Cancer Res 2007; 67:1308-16. [PMID: 17283168 DOI: 10.1158/0008-5472.can-06-1600] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The p53 protein is a sequence-specific DNA-binding factor that can induce apoptosis or activate genes whose dysregulation is involved in cancer. By using serial analysis of gene expression technique, p53-induced genes (PIGs) have been identified, one of which was lipopolysaccharide (LPS)-induced tumor necrosis factor-alpha (TNF-alpha) factor (LITAF/PIG7). LITAF regulates the transcription of cytokines such as TNF-alpha. To further elucidate the role of p53 in LITAF expression, LITAF promoter activity was carefully dissected. In this study, we found that the element required for transcriptional activity is mainly located in the region from -990 to -500 of the LITAF promoter; the specific site required for p53 protein-DNA binding is located between -550 and -500. We also found that transient transfection of either a p53 short DNA sequence, called p53LFB12, or its corresponding 7-amino-acid synthetic peptide from amino acids 164 to 170 (K164Q165S166Q167H168M169T170), named p53pep164, significantly reduced LITAF promoter activity to 15% in p53-null H1299 cells. Transfection of p53pep164 into H1299 cells significantly down-regulated LPS-induced LITAF expression as well. Furthermore, transfection of p53pep164 into human monocytes resulted in down-regulation of nine proinflammatory cytokines, including TNF-alpha. We also found that the LPS-activated p53 is a short-lived protein, and that p53-orchestrated apoptosis occurs shortly after the initiation stage following LPS stimulation and lasts a short time. Once p53 levels return to baseline, the p53-mediated inhibition of LITAF is released, and LITAF-mediated cytokine production can proceed. The present finding proposes a novel link between p53 and the inflammatory processes and highlights potential interventional approaches to control p53-associated inflammatory processes.
Collapse
Affiliation(s)
- Xiaoren Tang
- Department of Periodontology and Oral Biology, School of Dental Medicine, Boston University Medical Center, 700 Albany Street, Boston, MA 02118, USA
| | | | | |
Collapse
|
216
|
Wang P, Yu J, Zhang L. The nuclear function of p53 is required for PUMA-mediated apoptosis induced by DNA damage. Proc Natl Acad Sci U S A 2007; 104:4054-9. [PMID: 17360476 PMCID: PMC1820707 DOI: 10.1073/pnas.0700020104] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The tumor suppressor p53 can induce apoptosis by activating gene expression in the nucleus, or by directly permeabilizing mitochondria in the cytoplasm. It has been shown that PUMA, a downstream target of p53 and a BH3-only Bcl-2 family member, plays an essential role in apoptosis induced by both nuclear and cytoplasmic p53. To understand how PUMA does so, we used homologous recombination to delete the binding sites of p53 in the promoter of PUMA in human colorectal cancer cells. As a result, the induction of PUMA and apoptosis in response to p53 and DNA-damaging agents were abrogated. Transcription coactivator recruitment and histone modifications in the PUMA promoter were suppressed. However, induction of PUMA and apoptosis in response to non-DNA-damaging stimuli were unaffected. These results indicate that the binding of nuclear p53 to the specific sites within the PUMA promoter is essential for its ability to induce apoptosis and is likely to be required for its tumor suppressive capacity.
Collapse
Affiliation(s)
- Peng Wang
- Departments of Pharmacology and Pathology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213
| | - Jian Yu
- Departments of Pharmacology and Pathology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213
| | - Lin Zhang
- Departments of Pharmacology and Pathology, University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
217
|
Dorsey WC, Tchounwou PB, Ford BD. Neuregulin 1-Beta cytoprotective role in AML 12 mouse hepatocytes exposed to pentachlorophenol. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2007; 3:11-22. [PMID: 16823072 PMCID: PMC3785675 DOI: 10.3390/ijerph2006030002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuregulins are a family of growth factor domain proteins that are structurally related to the epidermal growth factor. Accumulating evidence has shown that neuregulins have cyto- and neuroprotective properties in various cell types. In particular, the neuregulin-1 Beta (NRG1-Beta) isoform is well documented for its antiinflammatory properties in rat brain after acute stroke episodes. Pentachlorophenol (PCP) is an organochlorine compound that has been widely used as a biocide in several industrial, agricultural, and domestic applications. Previous investigations from our laboratory have demonstrated that PCP exerts both cytotoxic and mitogenic effects in human liver carcinoma (HepG2) cells, primary catfish hepatocytes and AML 12 mouse hepatocytes. We have also shown that in HepG2 cells, PCP has the ability to induce stress genes that may play a role in the molecular events leading to toxicity and tumorigenesis. In the present study, we hypothesize that NRG1-Beta will exert its cytoprotective effects in PCP-treated AML 12 mouse hepatocytes by its ability to suppress the toxic effects of PCP. To test this hypothesis, we performed the MTT-cell respiration assay to assess cell viability, and Western-blot analysis to assess stress-related proteins as a consequence of PCP exposure. Data obtained from 48 h-viability studies demonstrated a biphasic response; showing a dose-dependent increase in cell viability within the range of 0 to 3.87 microg/mL, and a gradual decrease within the concentration range of 7.75 to 31.0 microg/mL in concomitant treatments of NRG1-Beta+PCP and PCP. Cell viability percentages indicated that NRG1-Beta+PCPtreated cells were not significantly impaired, while PCP-treated cells were appreciably affected; suggesting that NRG1-Beta has the ability to suppress the toxic effects of PCP. Western Blot analysis demonstrated the potential of PCP to induce oxidative stress and inflammatory response (c-fos), growth arrest and DNA damage (GADD153), proteotoxic effects (HSP70), cell cycle arrest as consequence of DNA damage (p53), mitogenic response (cyclin- D1), and apoptosis (caspase-3). NRG1-Beta exposure attenuated stress-related protein expression in PCP-treated AML 12 mouse hepatocytes. Here we provide clear evidence that NRG1-Beta exerts cytoprotective effects in AML 12 mouse hepatocytes exposed to PCP.
Collapse
Affiliation(s)
- Waneene C. Dorsey
- Molecular Toxicology Research Laboratory, Grambling State University, Grambling, LA,
USA
| | - Paul B. Tchounwou
- Molecular Toxicology Research Laboratory, NIH-Center for Environmental Health, College of Science, Engineering, and Technology, Jackson State University, Jackson, MS,
USA
- Correspondence to Dr. Paul B. Tchounwou.
| | - Byron D. Ford
- Department of Anatomy and Neurobiology, Morehouse School of Medicine, Atlanta, GA,
USA
| |
Collapse
|
218
|
Magrini R, Bakker A, Gaviraghi G, Terstappen GC. Targeting the p53 tumor suppressor gene function in glioblastomas using small chemical molecules. Drug Dev Res 2007. [DOI: 10.1002/ddr.20151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
219
|
Matsumoto M, Furihata M, Ohtsuki Y. Posttranslational phosphorylation of mutant p53 protein in tumor development. Med Mol Morphol 2006; 39:79-87. [PMID: 16821145 DOI: 10.1007/s00795-006-0320-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2006] [Accepted: 04/20/2006] [Indexed: 01/10/2023]
Abstract
p53 has been called the "cellular gatekeeper" and the "genome guard," because in response to exposure to DNA-damaging agents, it induces cell-cycle arrest in G1 or apoptosis and also directly affects DNA replication. Multiple mechanisms regulate p53 activity and posttranslational modification, including multisite phosphorylation of wild-type p53, in particular. Normal functions of wild-type p53 are abrogated by mutation of this gene, and oncogenic studies have revealed that p53 mutation is among the most common genetic alteration in human cancers. It is generally accepted that mutant p53 protein may not only lose the tumor suppressor functions of wild-type p53 but also acquire additional tumorigenetic roles, including dominant-negative effects and gain of function. Although many studies have revealed such aberrant functions of mutant p53, less is known about the posttranslational phosphorylation status of mutant p53 and novel biological functions of phosphorylation in carcinogenesis.
Collapse
Affiliation(s)
- Manabu Matsumoto
- Department of Clinical Laboratory, Kochi Medical School Hospital, Nankoku, Kochi, 783-8305, Japan.
| | | | | |
Collapse
|
220
|
Okorokov AL, Sherman MB, Plisson C, Grinkevich V, Sigmundsson K, Selivanova G, Milner J, Orlova EV. The structure of p53 tumour suppressor protein reveals the basis for its functional plasticity. EMBO J 2006; 25:5191-200. [PMID: 17053786 PMCID: PMC1630404 DOI: 10.1038/sj.emboj.7601382] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Accepted: 09/13/2006] [Indexed: 01/11/2023] Open
Abstract
p53 major tumour suppressor protein has presented a challenge for structural biology for two decades. The intact and complete p53 molecule has eluded previous attempts to obtain its structure, largely due to the intrinsic flexibility of the protein. Using ATP-stabilised p53, we have employed cryoelectron microscopy and single particle analysis to solve the first three-dimensional structure of the full-length p53 tetramer (resolution 13.7 A). The p53 molecule is a D2 tetramer, resembling a hollow skewed cube with node-like vertices of two sizes. Four larger nodes accommodate central core domains, as was demonstrated by fitting of its X-ray structure. The p53 monomers are connected via their juxtaposed N- and C-termini within smaller N/C nodes to form dimers. The dimers form tetramers through the contacts between core nodes and N/C nodes. This structure revolutionises existing concepts of p53's molecular organisation and resolves conflicting data relating to its biochemical properties. This architecture of p53 in toto suggests novel mechanisms for structural plasticity, which enables the protein to bind variably spaced DNA target sequences, essential for p53 transactivation and tumour suppressor functions.
Collapse
Affiliation(s)
- Andrei L Okorokov
- Department of Pathology, Royal Free and University College Medical School, University College London, London, UK
- Wolfson Institute for Biomedical Research, University College London, London, UK
- Department of Pathology, Royal Free and University College Medical School, University College London, London WCIE 6JJ, UK. Tel.: +44 20 7679 0959; Fax: +44 20 7388 4408; E-mail:
| | - Michael B Sherman
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Celia Plisson
- School of Crystallography, Birkbeck College, London, UK
| | - Vera Grinkevich
- Microbiology and Tumor Biology Center, Karolinska Institutet, Stockholm, Sweden
| | | | - Galina Selivanova
- Microbiology and Tumor Biology Center, Karolinska Institutet, Stockholm, Sweden
| | - Jo Milner
- YCR p53 Laboratory, Department of Biology, University of York, York, UK
| | - Elena V Orlova
- School of Crystallography, Birkbeck College, London, UK
- Department of Crystallography, Birkbeck College, Malet Street, London WC1E 7HX, UK. Tel.: +44 20 7631 6845; Fax: +44 20 7631 6803; E-mail:
| |
Collapse
|
221
|
Ohnishi K, Scuric Z, Schiestl RH, Okamoto N, Takahashi A, Ohnishi T. siRNA targeting NBS1 or XIAP increases radiation sensitivity of human cancer cells independent of TP53 status. Radiat Res 2006; 166:454-62. [PMID: 16972754 DOI: 10.1667/rr3606.1] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2006] [Accepted: 05/02/2006] [Indexed: 11/03/2022]
Abstract
NBS1 is essential for the repair of radiation-induced DNA double-strand breaks (DSBs) in yeast and higher vertebrate cells. In this study, we examined whether suppressed NBS1 expression by small interference RNA (siRNA) could enhance radiation sensitivity in cancer cells with different TP53 status. We used human non-small cell lung cancer cells differing in TP53 gene status (H1299/wtp53 cells bearing wild-type TP53 or H1299/mp53 cells bearing mutant TP53). A DNA cassette expressing siRNA targeted for the NBS1 gene was transfected into those cell lines, and radiation sensitivity was examined with a colony-forming assay. Cellular levels of NBS1 and other proteins were analyzed using Western blotting. We found that the radiation sensitivity of H1299/wtp53 and H1299/mp53 cells was enhanced by transfection of the DNA cassette. In the NBS1-siRNA-transfected cells, we observed decreased constitutive expression of NBS1 protein and decreased radiation-induced accumulation of phosphorylated NBS1 protein. In addition, radiation-induced expression of the transcription factor NF-kappaB (NFKB) and XIAP (X-chromosome-linked inhibitor of apoptosis protein) was suppressed by NBS1-siRNA. Enhanced X-ray sensitivity after NBS1-siRNA transfection was achieved in TP53 wild-type cells and sensitivity was even more pronounced in TP53 mutant cells. The transfection of siRNA targeted for XIAP also enhanced X-ray sensitivity even more for TP53 mutant cells compared to TP53 wild-type cells. Our data suggest that the sensitization to radiation results from NBS1-siRNA-mediated suppression of DNA repair and/ or X-ray-induced cell survival signaling pathways through NFKB and XIAP. siRNA targeting appears to be a novel radiation-sensitizing agent, particularly in human TP53 mutant cancer cells.
Collapse
Affiliation(s)
- Ken Ohnishi
- Department of Biology, Nara Medical University School of Medicine, Kashihara, Nara, Japan
| | | | | | | | | | | |
Collapse
|
222
|
Lu X, Feki A. Phenotypic features with p53 alterations related to human papillomavirus and prognostic evaluation in cervical cancer. Int J Gynecol Cancer 2006; 16:708-17. [PMID: 16681751 DOI: 10.1111/j.1525-1438.2006.00591.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cervical cancer is one of the most common tumor affecting women worldwide. Human papillomavirus (HPV) was found to have a causal relationship with cervical cancer and its precursors. The interaction between HPV E6 protein and p53 was identified in in vitro studies. The aim of the study was to evaluate the prevalence of p53 alterations related to HPV infection and the prognostic significance of p53 alterations in cervical cancer. Studies were identified by a MEDLINE search, and all relevant articles were retrieved from 1991 to March 2004. The prevalence of p53 mutations is a rare event in cervical cancer. The correlation between p53 mutations and HPV or prognosis is controversial. Loss of heterozygosity (LOH) of p53 is more commonly found in cervical cancer and is related with the prognosis of this disease. There is no significant correlation between p53 polymorphism and development of cervical cancer. The p53 mutations were not commonly found in cervical cancer. LOH of p53 may contribute to the progression of this malignancy. p53 polymorphism failed to be an independent prognostic factor in predicting the outcome of patients with cervical cancer. Further, epidemiologic surveys should be undertaken in larger populations and in different geographical regions.
Collapse
Affiliation(s)
- X Lu
- Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China.
| | | |
Collapse
|
223
|
Yamagata T, Maki K, Waga K, Mitani K. TEL/ETV6 induces apoptosis in 32D cells through p53-dependent pathways. Biochem Biophys Res Commun 2006; 347:517-26. [PMID: 16828711 DOI: 10.1016/j.bbrc.2006.06.127] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2006] [Accepted: 06/22/2006] [Indexed: 11/23/2022]
Abstract
TEL is an ETS family transcription factor that is critical for maintaining hematopoietic stem cells in adult bone marrow. To investigate the roles of TEL in myeloid proliferation and differentiation, we introduced TEL cDNA into mouse myeloid 32Dcl3 cells. Overexpression of TEL repressed interleukin-3-dependent proliferation through blocking cell cycle progression. Also, the presence of TEL triggered apoptosis through the mitochondrial intrinsic pathway on exposure to granulocyte colony-stimulating factor. We found an increase in p53 protein and its DNA binding in the TEL-overexpressing cells. Forced expression of TEL stimulated transcription via the p53-responsive element and increased the expression of cellular target genes for p53 such as cell cycle regulator p21 and apoptosis inducer Puma. Consistently, induction of apoptosis was delayed by pifithrin-alpha treatment and completely blocked by increased expression of Bcl-2 in the TEL-overexpressing cells. These data collectively suggest that TEL exerts a tumor suppressive function through augmenting the p53 pathway and facilitates normal development of myelopoiesis.
Collapse
Affiliation(s)
- Tetsuya Yamagata
- Department of Hematology, Dokkyo Medical University School of Medicine, Tochigi 321-0293, Japan
| | | | | | | |
Collapse
|
224
|
Takagi M, Takeda T, Asada Y, Sugimoto C, Onuma M, Ohashi K. The presence of a short form of p53 in chicken lymphoblastoid cell lines during apoptosis. J Vet Med Sci 2006; 68:561-6. [PMID: 16820712 DOI: 10.1292/jvms.68.561] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To examine the roles of a short form of p53 in the regulation of apoptosis in chicken lymphoblastoid tumor cell lines derived from Marek's disease (MD) and avian leukosis (AL), the expressions of the p53 proteins were analyzed in these cell lines in which apoptosis was chemically induced. In MSB1-O derived from MD, the expression of a 40 kDa protein of p53 was decreased and that of a 32 kDa protein, a short form of p53, was increased during apoptosis induced by actinomycin D. In 1104B1 derived from AL, the expressions of 42 and 32 kDa of p53 were increased during the apoptosis. The short form of p53 was undetectable in these cell lines when apoptosis was blocked by the pretreatment with endonuclease inhibitor, Zn2+, protease inhibitors, TPCK and TLCK, or caspase inhibitor, Z-VAD-FMK. In the transcriptional level, the expressions of bcl-2 and IAP were decreased in these cell lines during actinomycin D-induced apoptosis, but no change was detected in the expression level of p53. These results suggest that, in these chicken tumors, the short form of p53 could play a role in the initiation of apoptosis induced by the chemotherapeutic compound, and that the regulation of its expression may be important for the maintenance of transformation status.
Collapse
Affiliation(s)
- Michihiro Takagi
- Department of Microbiology and Immunology, Faculty of Agriculture, Kobe University, Japan
| | | | | | | | | | | |
Collapse
|
225
|
Duan W, Gao L, Wu X, Zhang Y, Otterson GA, Villalona-Calero MA. Differential response between the p53 ubiquitin-protein ligases Pirh2 and MdM2 following DNA damage in human cancer cells. Exp Cell Res 2006; 312:3370-8. [PMID: 16934800 DOI: 10.1016/j.yexcr.2006.07.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2006] [Revised: 07/03/2006] [Accepted: 07/10/2006] [Indexed: 12/13/2022]
Abstract
Pirh2, a recently identified ubiquitin-protein ligase, has been reported to promote p53 degradation. Pirh2 physically interacts with p53 and promotes ubiquitination of p53 independently of MDM2. Like MDM2, Pirh2 is thought to participate in an autoregulatory feedback loop that controls p53 function. We have previously reported that Pirh2 was overexpressed in human and murine lung cancers as compared to uninvolved lung tissue. Pirh2 increase could potentially cause degradation of wildtype p53 and reduce its tumor suppression function in the lung tumor cells. Since Pirh2 has been reported to be transactivated by p53, however, the mechanisms by which a high level of Pirh2 expression is maintained in tumor cells despite low level of wildtype p53 protein are unclear. In order to evaluate p53 involvement in the transactivation of Pirh2, we evaluated Pirh2, MDM2, p53 and p21 expression with Western blot analysis and real time PCR after gamma irradiation or cisplatin DNA damage treatment using human cancer cell lines containing wildtype (A549, MCF-7), mutant (H719) and null (H1299) p53. Surprisingly, Pirh2 expression was not affected by the presence of wildtype p53 in the cancer cells. In contrast, MDM2 was upregulated by wildtype p53 in A549 and MCF-7 cells and was absent from the H1299 and the H719 cells. We conclude that Pirh2 operates in a distinct manner from MDM2 in response to DNA damage in cancer cells. Pirh2 elevation in p53 null cells indicates the existence of additional molecular mechanisms for Pirh2 upregulation and suggests that p53 is not the sole target of Pirh2 ubiquitin ligase activity.
Collapse
Affiliation(s)
- Wenrui Duan
- Comprehensive Cancer Center and Department of Internal Medicine at The Ohio State University College of Medicine and Public Health, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
226
|
Zhang Y, Chen J, Gurumurthy CB, Kim J, Bhat I, Gao Q, Dimri G, Lee SW, Band H, Band V. The human orthologue of Drosophila ecdysoneless protein interacts with p53 and regulates its function. Cancer Res 2006; 66:7167-75. [PMID: 16849563 DOI: 10.1158/0008-5472.can-06-0722] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Biochemical mechanisms that control the levels and function of key tumor suppressor proteins are of great interest as their alterations can lead to oncogenic transformation. Here, we identify the human orthologue of Drosophila melanogaster ecdysoneless (hEcd) as a novel p53-interacting protein. Overexpression of hEcd increases the levels of p53 and enhances p53 target gene transcription whereas hEcd knockdown has the opposite effects on p53 levels and target gene expression. Furthermore, hEcd interacts with murine double minute-2 and stabilizes p53 by inhibiting murine double minute-2-mediated degradation of p53. Thus, hEcd protein represents a novel regulator of p53 stability and function. Our studies also represent the first demonstration of a biochemical function for hEcd protein and raise the possibility that altered hEcd levels and/or function may contribute to oncogenesis.
Collapse
Affiliation(s)
- Ying Zhang
- Division of Cancer Biology and Molecular Oncology, Department of Medicine, Evanston Northwestern Healthcare Research Institute and Feinberg School of Medicine, and Robert H. Lurie Comprehensive Cancer Center, Evanston, Illinois 60201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
227
|
Nakamura Y, Futamura M, Kamino H, Yoshida K, Nakamura Y, Arakawa H. Identification of p53-46F as a super p53 with an enhanced ability to induce p53-dependent apoptosis. Cancer Sci 2006; 97:633-41. [PMID: 16827804 PMCID: PMC11158242 DOI: 10.1111/j.1349-7006.2006.00214.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
More than half of human cancers contain mutations in the tumor suppressor protein p53, most of which accumulate in the DNA binding domain of the protein. Here we report the identification of a mutant p53, designated p53-46F, in which Ser-46 is replaced with phenylalanine. In vitro, adenovirus-mediated transduction of the p53-46F gene induced apoptosis more efficiently than wild-type p53 in a number of cancer cell lines, whereas Ser-15 phosphorylation of p53-46F was enhanced in all cancer cell lines examined. Moreover, the expression level of the cell cycle inhibitor p21/WAF1 was decreased in cell lines infected with adenovirus p53-46F (Ad-p53-46F). p53-46F caused a more enhanced level of transcriptional activation of several p53-target genes, including Noxa, p53AIP1 and p53RFP, compared with wild-type p53. In vivo, adenovirus-mediated gene transfer of p53-46F enhanced apoptosis, thus suppressing tumor growth of a lung cancer cell line more effectively than wild-type p53 or p53-121F, another p53 mutant. Collectively, our data suggest that p53-46F is an active version of p53 that demonstrates enhanced induction of p53-dependent apoptosis. This is probably mediated by upregulated transactivation of genes downstream of p53, increased Ser-15 phosphorylation and a decrease in p21/WAF1 levels. We propose p53-46F as an alternative candidate to wild-type p53 for use in developing new therapeutic strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Yasuyuki Nakamura
- Cancer Medicine and Biophysics Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | | | | | | | | | | |
Collapse
|
228
|
De Galitiis F, Cannita K, Tessitore A, Martella F, Di Rocco ZC, Russo A, Adamo V, Iacobelli S, Martinotti S, Marchetti P, Ficorella C, Ricevuto E. Novel P53 mutations detected by FAMA in colorectal cancers. Ann Oncol 2006; 17 Suppl 7:vii78-83. [PMID: 16760300 DOI: 10.1093/annonc/mdl957] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The aim of the study was to identify p53 gene mutations by FAMA (fluorescence-assisted mismatch analysis) in colorectal cancers. PATIENTS AND METHODS Analytical scanning of the p53 gene (exons 5-9) was performed in colon cancer samples from 44 consecutive patients by FAMA. FAMA is a semiautomatic scanning approach based on the chemical cleavage of the mismatch in fluorescently labeled heteroduplex DNA, obtained from the combination of a normal and a mutated allele. FAMA has already shown optimal levels of diagnostic accuracy and sensitivity in detecting gene mutations (nucleotide substitutions, insertions/deletions) both at the germline and somatic level. The peculiar feature of FAMA is its ability to detect and localize mutations, by a redundant pattern of signals due to fluorescent DNA fragments generated by chemical cleavage. Moreover, previous data have demonstrated that normal contaminating DNA from stromal cells in the sample does not affect the sensitivity of the procedure, leading to the identification of the mutation even when the ratio mutant/normal allele is 10%. RESULTS Eighteen mutations (12 missense, one nonsense, two deletions, three nucleotide substitutions at the level of the splice-junctions) and two polymorphisms were detected by FAMA in 17 patients (39%) and then confirmed by automated sequence analysis. Six of 18 mutations (33%) were not previously reported for colon cancer samples and two of 18 lesions (11%) were identified as novel p53 mutations. CONCLUSIONS Analytical scanning of the p53 gene by FAMA in DNA from colon cancer samples provides a sensitive, accurate and specific diagnostic procedure for routine clinical application.
Collapse
Affiliation(s)
- F De Galitiis
- U.O. Medical Oncology, San Salvatore Hospital, University of L'Aquila, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Zhang J, Yan W, Chen X. p53 is required for nerve growth factor-mediated differentiation of PC12 cells via regulation of TrkA levels. Cell Death Differ 2006; 13:2118-28. [PMID: 16729028 DOI: 10.1038/sj.cdd.4401972] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
p53 is necessary for the elimination of neural cells inappropriately differentiated or in response to stimuli. However, the role of p53 in neuronal differentiation is not certain. Here, we showed that nerve growth factor (NGF)-mediated differentiation in PC12 cells is enhanced by overexpression of wild-type p53 but inhibited by mutant p53 or knockdown of endogenous wild-type p53, the latter of which can be rescued by expression of exogenous wild-type p53. Interestingly, p53 knockdown or overexpression of mutant p53 attenuates NGF-mediated activation of TrkA, the high-affinity receptor for NGF and a tyrosine kinase, and activation of the mitogen-activated protein kinase pathway. In addition, p53 knockdown reduces the constitutive levels of TrkA, which renders PC12 cells inert to NGF. And finally, we showed that both constitutive and stimuli-induced expressions of TrkA are regulated by p53 and that induction of TrkA by activated endogenous p53 enhances NGF-mediated differentiation. Taken together, our data demonstrate that p53 plays a critical role in NGF-mediated neuronal differentiation in PC12 cells at least in part via regulation of TrkA levels.
Collapse
Affiliation(s)
- J Zhang
- Department of Cell Biology, The University of Alabama at Birmingham, Birmingham, 1530 3rd Avenue South, AL 35294-0005, USA
| | | | | |
Collapse
|
230
|
Espinoza-Fonseca LM, Trujillo-Ferrara JG. Transient stability of the helical pattern of region F19–L22 of the N-terminal domain of p53: A molecular dynamics simulation study. Biochem Biophys Res Commun 2006; 343:110-6. [PMID: 16530164 DOI: 10.1016/j.bbrc.2006.02.129] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2006] [Accepted: 02/22/2006] [Indexed: 12/29/2022]
Abstract
Two molecular dynamics simulations of the region E17-N29 of p53 (p53(17-29)) at different temperatures were performed for a total time of 0.2 micros, to study the conformational landscape of this region. Previous studies have suggested that this region displays different structural motifs, such as helix of a double beta-turn, and that its secondary structure might be transiently stable. Interestingly, in this study it was found that the region F19-L25, and particularly its fragment F19-L22, display a stable, transient helical pattern at sub-microsecond periods. The region F19-L22, which contains one of the most important residues needed for the interaction of p53 with MDM2, seems to be formed and stabilized by the existence of one hydrophobic and one aromatic cluster. The main function of these clusters is to help their surrounding area to desolvate, to allow the hydrogen bond network, therefore favoring the formation of a stable helix. This preliminary study would be useful for a better understanding of the structure and function of the N-terminal domain of p53 and its implications for the control of different types of cancer.
Collapse
Affiliation(s)
- L Michel Espinoza-Fonseca
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
231
|
Abstract
Pancreatic cancer is fundamentally a disease of inherited and acquired mutations in cancer-related genes. The genes targeted in pancreatic cancer include tumor-suppressor genes (p16/CDKN2A, TP53 and SMAD4), oncogenes (KRAS, BRAF, AKT2, MYB, and AIB1), and genome-maintenance genes (MLH1, MSH2, BRAC2 and other Fanconi anemia genes). An understanding of the cancer-related genes that are altered in pancreatic cancer has a number of clinical applications including genetic counseling for individuals with a family history of cancer, early detection of pancreatic neoplasia, and mechanism-based therapies for patients with advanced disease. This chapter will provide an overview of the molecular pathogenesis of pancreatic cancer with emphasis on clinical applications.
Collapse
Affiliation(s)
- Anirban Maitra
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA.
| | | | | |
Collapse
|
232
|
Chen GD, Chou CM, Hwang SPL, Wang FF, Chen YC, Hung CC, Chen JY, Huang CJ. Requirement of nuclear localization and transcriptional activity of p53 for its targeting to the yolk syncytial layer (YSL) nuclei in zebrafish embryo and its use for apoptosis assay. Biochem Biophys Res Commun 2006; 344:272-82. [PMID: 16616005 DOI: 10.1016/j.bbrc.2006.03.136] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2006] [Accepted: 03/20/2006] [Indexed: 11/28/2022]
Abstract
We expressed zebrafish p53 protein fused to GFP by a neuron-specific HuC promoter in zebrafish embryos. Instead of displaying neuronal expression patterns, p53-GFP was targeted to zebrafish YSL nuclei. This YSL targeting is p53 sequence-specific because GFP fusion proteins of p63 and p73 displayed neuronal-specific patterns. To dissect the underlying mechanisms, various constructs encoding a series of p53 mutant proteins under the control of different promoters were generated. Our results showed that expression of p53, in early zebrafish embryo, is preferentially targeted to the nuclei of YSL, which is mediated by importin. Similarly, this targeting is abrogated when p53 nuclear localization signal is disrupted. In addition, the transcriptional activity of p53 is required for this targeting. We further showed that fusion of pro-apoptotic BAD protein to p53-GFP led to apoptosis of YSL cells, and subsequent imperfect microtubule formation and abnormal blastomere movements.
Collapse
Affiliation(s)
- Gen-Der Chen
- Institute of Biochemistry, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
233
|
Gomes NP, Bjerke G, Llorente B, Szostek SA, Emerson BM, Espinosa JM. Gene-specific requirement for P-TEFb activity and RNA polymerase II phosphorylation within the p53 transcriptional program. Genes Dev 2006; 20:601-12. [PMID: 16510875 PMCID: PMC1410802 DOI: 10.1101/gad.1398206] [Citation(s) in RCA: 225] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activation of the p53 pathway mediates cellular responses to diverse forms of stress. Here we report that the p53 target gene p21(CIP1) is regulated by stress at post-initiation steps through conversion of paused RNA polymerase II (RNAP II) into an elongating form. High-resolution chromatin immunoprecipitation assays (ChIP) demonstrate that p53-dependent activation of p21(CIP1) transcription after DNA damage occurs concomitantly with changes in RNAP II phosphorylation status and recruitment of the elongation factors DSIF (DRB Sensitivity-Inducing Factor), P-TEFb (Positive Transcription Elongation Factor b), TFIIH, TFIIF, and FACT (Facilitates Chromatin Transcription) to distinct regions of the p21(CIP1) locus. Paradoxically, pharmacological inhibition of P-TEFb leads to global inhibition of mRNA synthesis but activation of the p53 pathway through p53 accumulation, expression of specific p53 target genes, and p53-dependent apoptosis. ChIP analyses of p21(CIP1) activation in the absence of functional P-TEFb reveals the existence of two distinct kinases that phosphorylate Ser5 of the RNAP II C-terminal domain (CTD). Importantly, CTD phosphorylation at Ser2 is not required for p21(CIP1) transcription, mRNA cleavage, or polyadenylation. Furthermore, recruitment of FACT requires CTD kinases, yet FACT is dispensable for p21(CIP1) expression. Thus, select genes within the p53 pathway bypass the requirement for P-TEFb and RNAP II phosphorylation to trigger a cellular response to inhibition of global mRNA synthesis.
Collapse
Affiliation(s)
- Nathan P Gomes
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
234
|
di Masi A, Antoccia A, Dimauro I, Argentino-Storino A, Mosiello A, Mango R, Novelli G, Tanzarella C. Gene expression and apoptosis induction in p53-heterozygous irradiated mice. Mutat Res 2006; 594:49-62. [PMID: 16169021 DOI: 10.1016/j.mrfmmm.2005.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Revised: 07/11/2005] [Accepted: 07/29/2005] [Indexed: 05/04/2023]
Abstract
The role of the p53-genetic background in the expression of genes involved in either cell cycle checkpoint activation or apoptosis was evaluated in p53+/+ and p53+/- mouse strains at both basal levels and after DNA-induced damage. The spleen, colon, kidneys, lungs and liver of both strains were harvested from untreated animals and from mice exposed to 7.5 Gy of X-rays and sacrificed after 5 h. No significant differences were observed in the basal levels of p53 protein, CDKN1A and bax mRNA and spontaneous apoptosis, neither among the different organs within the same strain, nor between the same organ in the p53+/+ and p53+/- strains. After X-ray exposure, p53-dependent regulation was strikingly tissue-specific. In wild-type irradiated mice, p53 protein level increased after radiation treatment in all the organs analysed, whereas both CDKN1A and bax genes transcription increased in the spleen, colon and lungs, as assessed by means of quantitative RT-PCR. In p53+/- irradiated mice, on the contrary, a significant p53 induction was detected only in the spleen, while CDKN1A and bax genes levels increased in the spleen, colon and lungs, revealing the existence of different mechanisms of gene regulation in different organs. Apoptosis induction was observed in the spleen and colon of both strains, even if to lower extent in p53+/- mice compared to p53+/+ animals. In conclusion, in the spleen and colon, target gene transcription and apoptosis may be related to p53 genotype after DNA damage-induction. Moreover, our findings highlight the selectivity of p53 in transactivation following DNA damage in vivo, resulting in tissue-specific responses.
Collapse
Affiliation(s)
- Alessandra di Masi
- Department of Biology, University of Rome Roma Tre, Viale G. Marconi, 446, 00146 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
235
|
Barbieri CE, Pietenpol JA. p63 and epithelial biology. Exp Cell Res 2006; 312:695-706. [PMID: 16406339 DOI: 10.1016/j.yexcr.2005.11.028] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 11/15/2005] [Accepted: 11/17/2005] [Indexed: 02/06/2023]
Abstract
The transcription factor p63 is a homologue of the tumor suppressor p53. Unlike p53, which is dispensable for normal development, p63 is critical for the development of stratified epithelial tissues such as epidermis, breast, and prostate. p63 encodes multiple protein isoforms with both transactivating and transcriptional repressor activities that can regulate a wide spectrum of target genes. p63 is also implicated in tumor formation and progression in stratified epithelia, with evidence for both tumor suppressive and oncogenic properties. This review will examine current data and hypotheses regarding the role of p63 in the development, maintenance, and tumorigenesis of stratified epithelium.
Collapse
Affiliation(s)
- Christopher E Barbieri
- Department of Biochemistry, Center in Molecular Toxicology, 652 Preston Research Building, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | |
Collapse
|
236
|
Strauss BE, Bajgelman MC, Costanzi-Strauss E. A novel gene transfer strategy that combines promoter and transgene activities for improved tumor cell inhibition. Cancer Gene Ther 2005; 12:935-46. [PMID: 15905860 DOI: 10.1038/sj.cgt.7700846] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2004] [Indexed: 11/09/2022]
Abstract
Typically, gene transfer strategies utilize a promoter/transgene arrangement that treat these elements independently and do not offer any interplay between them. Our goal was to establish a promoter/transgene combination that would result in improvement in both expression and therapeutic effect by utilizing the transcriptional properties of p53 to drive its own expression as well as act as a tumor suppressor. The pCL retroviral system was modified in the U3 region of the 3' LTR by the addition of a p53-responsive sequence (the PG element), creating the pCLPG system. Upon reverse transcription, the 5' LTR is converted, as shown here, to a p53-dependent promoter. We also show, using a temperature-sensitive model, that the pCLPG system could be driven by p53 encoded within the virus construct and expression was modulated depending on the p53 phenotype, demonstrating a regulatory feedback loop. Moreover, the pCLPG system was shown to express the transgene at a higher level and to inhibit tumor cell proliferation more robustly than the original pCL system. This novel system employs the transgene to serve two purposes, drive viral expression and inhibit tumor cell proliferation. The pCLPG vectors represent a new gene transfer strategy of synergizing the promoter and transgene activities.
Collapse
Affiliation(s)
- Bryan E Strauss
- Heart Institute, InCor, University of São Paulo School of Medicine, Av. De Eneas de Carvalho Aguiar 44, Building II 10th Floor, São Paulo (SP), CEP 05403-000 Brazil.
| | | | | |
Collapse
|
237
|
Hearnes JM, Mays DJ, Schavolt KL, Tang L, Jiang X, Pietenpol JA. Chromatin immunoprecipitation-based screen to identify functional genomic binding sites for sequence-specific transactivators. Mol Cell Biol 2005; 25:10148-58. [PMID: 16260627 PMCID: PMC1280257 DOI: 10.1128/mcb.25.22.10148-10158.2005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2005] [Revised: 06/06/2005] [Accepted: 08/28/2005] [Indexed: 02/05/2023] Open
Abstract
In various human diseases, altered gene expression patterns are often the result of deregulated gene-specific transcription factor activity. To further understand disease on a molecular basis, the comprehensive analysis of transcription factor signaling networks is required. We developed an experimental approach, combining chromatin immunoprecipitation (ChIP) with a yeast-based assay, to screen the genome for transcription factor binding sites that link to transcriptionally regulated target genes. We used the tumor suppressor p53 to demonstrate the effectiveness of the method. Using primary and immortalized, nontransformed cultures of human mammary epithelial cells, we isolated over 100 genomic DNA fragments that contain novel p53 binding sites. This approach led to the identification and validation of novel p53 target genes involved in diverse signaling pathways, including growth factor signaling, protein kinase/phosphatase signaling, and RNA binding. Our results yield a more complete understanding of p53-regulated signaling pathways, and this approach could be applied to any number of transcription factors to further elucidate complex transcriptional networks.
Collapse
Affiliation(s)
- Jamie M Hearnes
- Department of Biochemistry, Center for Molecular Toxicology, Vanderbilt-Ingram Cancer Center, Nashville, TN 37232-6838, USA
| | | | | | | | | | | |
Collapse
|
238
|
Bjørling-Poulsen M, Siehler S, Wiesmüller L, Meek D, Niefind K, Issinger OG. The 'regulatory' beta-subunit of protein kinase CK2 negatively influences p53-mediated allosteric effects on Chk2 activation. Oncogene 2005; 24:6194-200. [PMID: 15940255 DOI: 10.1038/sj.onc.1208762] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The 'regulatory' beta-subunit of protein kinase CK2 has previously been shown to interact with protein kinases such as A-Raf, c-Mos, Lyn and Chk1 in addition to the catalytic subunit of CK2. Sequence alignments suggest that these interactions have a structural basis, and hence other protein kinases harboring corresponding sequences may be potential interaction partners for CK2beta. We show here that Chk2 specifically interacts with CK2beta in vitro and in cultured cells, and that activation of Chk2 leads to a reduction of this interaction. Additionally, we show that the presence of the CK2beta-subunit significantly reduces the Chk2-catalysed phosphorylation of p53 in vitro. These findings support the notion that CK2beta can act as a general modulator of remote docking sites in protein kinase--substrate interactions.
Collapse
|
239
|
Spyres L, Gaddis S, Bedford E, Arantes S, Liburd N, Powell KL, Thames H, Mitchell D, Walborg E, Rouabhia M, Aldaz CM, MacLeod MC. Quantitative high-throughput measurement of gene expression with sub-zeptomole sensitivity by capillary electrophoresis. Anal Biochem 2005; 345:284-95. [PMID: 16125665 DOI: 10.1016/j.ab.2005.07.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Revised: 07/06/2005] [Accepted: 07/13/2005] [Indexed: 11/17/2022]
Abstract
Microarray technologies have provided the ability to monitor the expression of whole genomes rapidly. However, concerns persist with regard to quantitation and reproducibility, and the detection limits for individual genes in particular arrays are generally unknown. This article describes a semiautomated PCR-based technology, Q-RAGE, which rapidly provides measurements of mRNA abundance with extremely high sensitivity using fluorescent detection of specific products separated by capillary electrophoresis. A linear relationship between template concentration and fluorescent signal can be demonstrated down to template concentrations in the low aM region, corresponding to approximately 0.04 zmol (24 molecules) per reaction. The technique is shown to be quantitative over five orders of magnitude of template concentration, and average mRNA abundances of approximately 0.01 molecule per cell can be detected. A single predefined set of 320 primers provides 90-95% coverage of all eukaryotic genomes. Analysis of a set of 19 p53-regulated genes in untreated cultures of normal human epithelial cells, derived from three different tissues, revealed a 600-fold range of apparent constitutive expression levels. For most of the genes assayed, good correlations were observed among the expression levels in normal mammary, bronchial, and epidermal epithelial cells.
Collapse
Affiliation(s)
- Lea Spyres
- Department of Carcinogenesis, University of Texas M. D. Anderson Cancer Center, Smithville, TX 78957, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Warburton HE, Brady M, Vlatković N, Linehan WM, Parsons K, Boyd MT. p53 Regulation and Function in Renal Cell Carcinoma. Cancer Res 2005; 65:6498-503. [PMID: 16061625 DOI: 10.1158/0008-5472.can-05-0017] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Loss of p53 function is a critical event in tumor evolution. This occurs through a range of molecular events, typically a missense p53 mutation followed by loss of heterozygosity. In many cancers, there is compelling evidence that cells that can compromise p53 function have a selective advantage. The situation in renal cell carcinoma is unclear. It has recently been suggested that p53 function is unusually compromised in renal carcinoma cells by a novel dominant, MDM2/p14ARF-independent mechanism. This is hard to reconcile with other recent studies that have identified p53 as an important prognostic indicator. Indeed, one of these latter studies found that the best predictor of poor outcome was the presence of high levels of both p53 (usually indicative of p53 mutation) and MDM2. Thus, it is important that we gain a clearer understanding of the regulation of p53 and the role of MDM2 in renal cell cancer. To address this, we have investigated the transcriptional activity of p53 in a panel of renal cell carcinoma cell lines and the contribution of MDM2 and p14ARF to p53 regulation. We have found that p53 is functional in p53 wild-type renal cell carcinoma cells and that this activity is significantly regulated by MDM2 and to a much lesser extent by p14ARF. Moreover, following induction of DNA damage with UV, the p53 response in these cells is intact. Thus, future studies of renal cell carcinoma that focus on p53 and MDM2 and their role in determining disease outcome will be required to create a better understanding of this notoriously difficult to manage disease.
Collapse
Affiliation(s)
- Hazel E Warburton
- MDM2/p53 Laboratory, Division of Surgery and Oncology, University of Liverpool
| | | | | | | | | | | |
Collapse
|
241
|
Woo GH, Bak EJ, Nakayama H, Doi K. Hydroxyurea (HU)-induced apoptosis in the mouse fetal lung. Exp Mol Pathol 2005; 79:59-67. [PMID: 16005713 DOI: 10.1016/j.yexmp.2005.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Accepted: 02/25/2005] [Indexed: 10/25/2022]
Abstract
In this study, the cytotoxicologic effects of HU on the fetal lung were assessed by exposing pregnant mice to HU on day 13 of gestation. The number of TUNEL-positive cells, i.e., apoptotic cells, in the fetal lung began to increase at 3 h after treatment (h), peaked at 6 h, and decreased thereafter, and the sequence of the number of cleaved caspase 3-positive cells corresponded to that of TUNEL-positive cells. Such positive reactivity for TUNEL and cleaved caspase 3 was mainly seen in pulmonary mesenchymal cells. Prior to the induction of apoptosis, the number of p53-positive cells in the fetal lung prominently increased at 1 and 3 h, and decreased thereafter. Among p53 transcriptional target genes (p21, fas, bax, apaf1, cyclin G, mdm2, and gad 45) examined, the expression levels of p21, bax, and cyclin G mRNAs were significantly elevated. In addition, the expression of fas mRNA tended to show higher levels compared with controls until 24 h. In addition, the results of flow cytometric analysis suggested that cell cycle arrest might be induced in S phase at 3 h. The present results suggest that HU-induced apoptosis in the mouse fetal lung may be closely related with the induction of p53.
Collapse
Affiliation(s)
- Gye-Hyeong Woo
- Department of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | |
Collapse
|
242
|
Boehden GS, Baumann C, Siehler S, Wiesmüller L. Wild-type p53 stimulates homologous recombination upon sequence-specific binding to the ribosomal gene cluster repeat. Oncogene 2005; 24:4183-92. [PMID: 15782112 DOI: 10.1038/sj.onc.1208592] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
p53 plays a central role in the maintenance of the genome integrity, both as a gatekeeper and a caretaker. Sequence-specific recognition of DNA is underlying the ability of p53 to transcriptionally transactivate target genes during checkpoint control and to regulate DNA replication at the TGCCT repeat from the ribosomal gene cluster (RGC). In contrast, suppression of recombination by p53 has been observed with nonconsensus DNA sequences. In this study, we discovered that wild-type p53 stimulates homologous recombination adjacent to the RGC repeat, whereas downregulation is seen with a mutated version thereof and with a microsatellite repeat sequence. Analysis of the causes possibly underlying the enhancement of homologous recombination revealed that p53 binding to the RGC element delays DNA synthesis. This was demonstrated after integration of the corresponding DNA fragments into our Simian virus 40-based model system, which was used to study recombination on replicating minichromosomes. Differently, with plasmid-based substrates, p53 did not stimulate recombination at the RGC sequence. Thus, in combination with our previous findings, p53 may promote homologous recombination by two separate mechanisms involving either molecular interactions with topoisomerase I or/and by specific binding to certain genomic regions, thereby causing replication fork stalling and recombination.
Collapse
Affiliation(s)
- Gisa S Boehden
- Universitätsfrauenklinik, Prittwitzstrasse 43, D-89075 Ulm, Germany
| | | | | | | |
Collapse
|
243
|
Yu J, Zhang L. The transcriptional targets of p53 in apoptosis control. Biochem Biophys Res Commun 2005; 331:851-8. [PMID: 15865941 DOI: 10.1016/j.bbrc.2005.03.189] [Citation(s) in RCA: 318] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2005] [Indexed: 01/12/2023]
Abstract
Induction of apoptosis is an essential function of p53 as a tumor suppressor. p53 can activate its downstream targets in a sequence specific manner to induce apoptosis. Most tumor derived p53 mutants are deficient in transcription activation as well as apoptosis induction. p53 can activate genes in the extrinsic and intrinsic pathways through transcription-dependent mechanisms or induce apoptosis through transcription-independent mechanisms. Several proapoptotic Bcl-2 family proteins, such as PUMA and Noxa, are shown to be critical mediators of p53-dependent apoptosis. The selective activation of the apoptotic targets of p53 is modulated by transcription coactivators. The induction of apoptotic genes alone sometimes is not sufficient to induce apoptosis, as the cell cycle arrest mediated by the cell cycle inhibitors dominates apoptosis. Preventing the induction of p21 under these conditions can drive the cells towards apoptosis. Understanding how p53 controls apoptosis through its targets may lead to discoveries of novel therapeutics to combat cancer and other diseases.
Collapse
Affiliation(s)
- Jian Yu
- The Department of Pathology, University of Pittsburgh School of Medicine, The University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
244
|
Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, Rustgi AK, Chang S, Tuveson DA. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 2005; 7:469-83. [PMID: 15894267 DOI: 10.1016/j.ccr.2005.04.023] [Citation(s) in RCA: 1955] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Revised: 03/16/2005] [Accepted: 04/25/2005] [Indexed: 11/26/2022]
Abstract
To define the genetic requirements for pancreatic ductal adenocarcinoma (PDA), we have targeted concomitant endogenous expression of Trp53(R172H) and Kras(G12D) to the mouse pancreas, revealing the cooperative development of invasive and widely metastatic carcinoma that recapitulates the human disease. The primary carcinomas and metastases demonstrate a high degree of genomic instability manifested by nonreciprocal translocations without obvious telomere erosion-hallmarks of human carcinomas not typically observed in mice. No mutations were discovered in other cardinal tumor suppressor gene pathways, which, together with previous results, suggests that there are distinct genetic pathways to PDA with different biological behaviors. These findings have clear implications for understanding mechanisms of disease pathogenesis, and for the development of detection and targeted treatment strategies.
Collapse
MESH Headings
- Animals
- Cadherins/metabolism
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Centrosome/pathology
- Chromosomal Instability/genetics
- Chromosome Aberrations
- Cytogenetic Analysis
- Disease Progression
- Gene Expression/genetics
- Gene Expression Regulation/genetics
- Gene Rearrangement/genetics
- Genes, Tumor Suppressor
- Homeodomain Proteins/genetics
- Integrases/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Mice, Mutant Strains
- Mice, Transgenic
- Mutation, Missense
- Neoplasm Metastasis
- Oncogene Proteins v-erbB/metabolism
- Proto-Oncogene Proteins p21(ras)
- Survival Analysis
- Telomere/genetics
- Trans-Activators/genetics
- Translocation, Genetic
- Tumor Suppressor Protein p53/genetics
- ras Proteins/genetics
Collapse
Affiliation(s)
- Sunil R Hingorani
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Abstract
As a transcription factor, p53 recognizes a specific consensus DNA sequence and activates the expression of the target genes involved in either growth arrest or apoptosis. Despite our wealth of knowledge on the genes that are targeted by p53 in growth arrest and apoptosis, relatively little is known about the promoter specificity triggered by p53 in these processes. Here we show that interaction with c-Abl stabilized p53 tetrameric conformation, and as a consequence c-Abl stimulated p53 DNA binding only when all quarter binding sites (a perfect binding sequence) on p53-responsive promoters were present. This result suggests that in response to DNA damage, c-Abl binding may specifically stimulate p53 DNA binding on the promoters with perfect binding sequences. A sequence comparison of several known p53-responsive elements illustrates the presence of the perfect binding sequences on the p21 but not the Bax promoter. Significantly, we show that c-Abl indeed enhanced p53 DNA binding and transcription from p21 but not Bax. These results suggest that the promoter specificity plays an important role in selective activation of p53 DNA binding by c-Abl. The implications of this with relation to selective activation of p53 target genes involved in either growth arrest or apoptosis are discussed.
Collapse
Affiliation(s)
- Gang Wei
- Department of Biochemistry, University of California, Riverside, California 92521, USA
| | | | | |
Collapse
|
246
|
Park JK, Feuerman MH. Afr2 regulation occurs cell-autonomously in vitro but is not conferred on episomal DNA in transient assays. DNA Cell Biol 2005; 24:189-98. [PMID: 15767785 DOI: 10.1089/dna.2005.24.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oncofetal antigens such as alpha-fetoprotein (AFP) are expressed in regenerating liver. The level of AFP gene expression during liver regeneration is regulated by the unlinked, autosomal gene, Alpha-fetoprotein regulator 2 (Afr2). C3H/HeJ (Afr2A/A) mice express 10-fold higher levels of AFP than C57BL/6J (Afr2B/B) mice. Here we show that primary hepatocytes isolated from C3H/HeJ and C57BL/6J mice exhibit differential expression of the endogenous AFP gene, which was attributed to the Afr2 gene locus and indicative of a cell-autonomous mechanism. We show that the Afr2-Response Element (ARE), between 1010 and 838 base pairs upstream of the AFP transcriptional start site, did not modulate reporter gene expression in transfection assays of Hep G2, Hep 3B, Hepa 1.6, and HeLa cell lines. Reporter gene expression in transiently transfected primary hepatocytes was also ARE-independent. Finally, gene expression from reporter constructs delivered by hydrodynamics-based transfection to the livers of C3H/HeJ and C57BL/6J mice after CCl4-induced liver regeneration was ARE-independent. In conclusion, ARE-dependent transcription was not found in transient assays performed in three different systems, two of which retained regulation of the endogenous AFP gene, suggesting that the ARE may not function as a simple transcription factor recognition site.
Collapse
Affiliation(s)
- James K Park
- Graduate Program in Molecular and Cellular Biology and Department of Biochemistry, State University of New York-Downstate Medical Center, Brooklyn, New York 11203, USA
| | | |
Collapse
|
247
|
Castro ME, del Valle Guijarro M, Moneo V, Carnero A. Cellular senescence induced by p53-ras cooperation is independent of p21waf1 in murine embryo fibroblasts. J Cell Biochem 2004; 92:514-24. [PMID: 15156563 DOI: 10.1002/jcb.20079] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Oncogenic activation in primary murine fibroblasts initiates a senescence-like cell cycle arrest that depends on the p53 tumor suppressor pathway. Conditional p53 activation efficiently induced a reversible cell cycle arrest but was unable to induce features of senescence. In contrast, coexpression of oncogenic ras with p53 produced an irreversible cell cycle arrest that displayed features of cellular senescence. Introduction of a conditional murine p53 allele (p53val135) into double p53/p21-null mouse embryonic fibroblasts showed that p21waf1 was not required for this effect, since p53-/-;p21-/- double-null cells undergo terminal growth arrest with features of senescence following coexpression of oncogenic Ras and p53. Our results indicate that oncogenic activation of the Ras pathway in murine fibroblasts converts p53 into a senescence inducer through a p21waf1-independent mechanism.
Collapse
Affiliation(s)
- Maria E Castro
- Experimental Therapeutics Program, Centro Nacional de Investigaciones Oncologicas (CNIO), Melchor Fernandez Almagro, 3, 28029, Madrid Spain
| | | | | | | |
Collapse
|
248
|
Paz K, Socci ND, van Nimwegen E, Viale A, Darnell JE. Transformation fingerprint: induced STAT3-C, v-Src and Ha-Ras cause small initial changes but similar established profiles in mRNA. Oncogene 2004; 23:8455-63. [PMID: 15378015 DOI: 10.1038/sj.onc.1207803] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Induced transformation of mouse fibroblasts was carried out by releasing tetracycline-repressed expression of an oncogenic mutant of STAT3, STAT3-C, or of v-Src or Ha-Ras. At 15 days after derepression of each oncogene, DNA microarrays showed elevation (>3-fold) of a similar group of approximately 25 mRNAs compared to untransformed cells. RT-PCR confirmed a number of these mRNA elevations. RNA samples were then analysed at intervals during the first 24 h after doxycycline removal to determine the time of early changes. Extensive changes were not observed by array analysis, except in v-Src-expressing cells where about 10 mRNAs were elevated threefold or more. However, RT-PCR did uncover changes in each derepressed cell type that included some of the changes observed after the 15-day transformation period. In addition, STAT3-C target genes such as BclXI and cyclin D1, which were not observed on array analysis, were elevated by RT-PCR analysis. We conclude, therefore, that early after oncogene induction, transcriptional changes, including those initiated by STAT3-C, may occur only in scarce mRNA and/or to a limited extent. However, with additional time and probably additional cell division, a new epigenetic state is established that is mirrored by a changed transcriptional profile emblematic of transformation by each of three oncogenes.
Collapse
Affiliation(s)
- Keren Paz
- Laboratory of Molecular Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
249
|
Gu L, Zhu N, Findley HW, Woods WG, Zhou M. Identification and Characterization of the IKKα Promoter. J Biol Chem 2004; 279:52141-9. [PMID: 15469934 DOI: 10.1074/jbc.m407915200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
IKKalpha, a subunit of IkBalpha kinase (IKK) complex, has an important role in the activation of nuclear factor-kB (NF-kB), a key regulator of normal and tumor cell proliferation, apoptosis, and response to chemotherapy. However, little is known about the transcriptional regulation of the IKKalpha gene itself. The present study revealed that the transcriptional induction of the IKKalpha gene is positively regulated by binding ETS-1, the protein product of the ETS-1 proto-oncogene. Furthermore, ETS-1 mediated activation of IKKalpha is negatively regulated by p53 binding to ETS-1. By analyzing the genomic DNA sequence, we identified the putative IKKalpha promoter sequence in the 5'-flanking untranslated region of the IKKalpha gene. Transfection of EU-4, an acute lymphoblastic leukemia (ALL) cell line, with plasmids containing the IKKalpha 5'-untranslated region sequence upstream of the luciferase reporter showed that this region possessed major promoter activity. Induction or enforced overexpression of p53 represses IKKalpha mRNA and protein expression as well as IKKalpha promoter activity. Deletion and mutation analyses as well as chromatin immunoprecipitation and electrophoretic mobility shift assay indicated that ETS-1 binds to the core IKKalpha promoter and strongly induces its activity. Although p53 does not directly bind to the IKKalpha promoter, it physically interacts with ETS-1 and specifically inhibits ETS-1-induced IKKalpha promoter activity. These results suggest that the proximal 5'-flanking region of the IKKalpha gene contains a functional promoter reciprocally regulated by p53 and ETS-1. Furthermore, loss of p53-mediated control over ETS-1-dependent transactivation of IKKalpha may represent a novel pathway for the constitutive activation of NF-kB-mediated gene expression and therapy resistance in cancer.
Collapse
Affiliation(s)
- Lubing Gu
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
250
|
Gomez-Lazaro M, Fernandez-Gomez FJ, Jordán J. p53: Twenty five years understanding the mechanism of genome protection. J Physiol Biochem 2004; 60:287-307. [PMID: 15957248 DOI: 10.1007/bf03167075] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This year the p53 protein, also known as "guardian of the genome", turns twenty five years old. During this period the p53 knowledge have changed from an initial pro-oncogene activity to the tumorsupressor p53 function. p53 is activated upon stress signals, such as gamma irradiation, UV, hypoxia, virus infection, and DNA damage, leading to protection of cells by inducing target genes. The molecules activated by p53 induce cell cycle arrest, DNA repair to conserve the genome and apoptosis. The regulation of p53 functions is tightly controlled through several mechanisms including p53 transcription and translation, protein stability, post-translational modifications, and subcellular localization. In fact, mutations in p53 are the most frequent molecular alterations detected in human tumours. Furthermore, in some degenerative processes, fragmentation and oxidative damage in DNA take place, and in these situations p53 is involved. So, p53 is considered a pharmacological target, p53 overexpression induces apoptosis in cancer and its expression blockage protects cells against lethal insults.
Collapse
Affiliation(s)
- M Gomez-Lazaro
- Centro Regional de Investigaciones Biomédicas, Facultad de Medicina, Universidad de Castilla-La Mancha, Avda. Almansa, 02006 Albacete, Spain
| | | | | |
Collapse
|