201
|
Patel S, Bui TT, Drake AF, Fraternali F, Nikolova PV. The p73 DNA Binding Domain Displays Enhanced Stability Relative to Its Homologue, the Tumor Suppressor p53, and Exhibits Cooperative DNA Binding. Biochemistry 2008; 47:3235-44. [DOI: 10.1021/bi7023207] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Seema Patel
- Department of Biochemistry and Pharamceutical Sciences Research Division, School of Biomedical & Health Sciences, King’s College London, 150 Stamford Street, London SE1 9NH, United Kingdom, and Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, Kingʼs College London, London SE1 1UL, United Kingdom
| | - Tam T.T. Bui
- Department of Biochemistry and Pharamceutical Sciences Research Division, School of Biomedical & Health Sciences, King’s College London, 150 Stamford Street, London SE1 9NH, United Kingdom, and Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, Kingʼs College London, London SE1 1UL, United Kingdom
| | - Alex F. Drake
- Department of Biochemistry and Pharamceutical Sciences Research Division, School of Biomedical & Health Sciences, King’s College London, 150 Stamford Street, London SE1 9NH, United Kingdom, and Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, Kingʼs College London, London SE1 1UL, United Kingdom
| | - Franca Fraternali
- Department of Biochemistry and Pharamceutical Sciences Research Division, School of Biomedical & Health Sciences, King’s College London, 150 Stamford Street, London SE1 9NH, United Kingdom, and Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, Kingʼs College London, London SE1 1UL, United Kingdom
| | - Penka V. Nikolova
- Department of Biochemistry and Pharamceutical Sciences Research Division, School of Biomedical & Health Sciences, King’s College London, 150 Stamford Street, London SE1 9NH, United Kingdom, and Randall Division of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, Kingʼs College London, London SE1 1UL, United Kingdom
| |
Collapse
|
202
|
Beitzinger M, Hofmann L, Oswald C, Beinoraviciute-Kellner R, Sauer M, Griesmann H, Bretz AC, Burek C, Rosenwald A, Stiewe T. p73 poses a barrier to malignant transformation by limiting anchorage-independent growth. EMBO J 2008; 27:792-803. [PMID: 18239687 DOI: 10.1038/emboj.2008.13] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2007] [Accepted: 01/11/2008] [Indexed: 11/09/2022] Open
Abstract
p53 is known to prevent tumour formation by restricting the proliferation of damaged or oncogene-expressing cells. In contrast, how the p53 family member p73 suppresses tumour formation remains elusive. Using a step-wise transformation protocol for human cells, we show that, in premalignant stages, expression of the transactivation-competent p73 isoform TAp73 is triggered in response to pRB pathway alterations. TAp73 expression at this stage of transformation results in increased sensitivity to chemotherapeutic drugs and oxidative stress and inhibits proliferation and survival at high cell density. Importantly, TAp73 triggers a transcriptional programme to prevent anchorage-independent growth, which is considered a crucial hallmark of fully transformed cells. An essential suppressor of anchorage-independent growth is KCNK1, which is directly transactivated by TAp73 and commonly downregulated in glioma, melanoma and ovarian cancer. Oncogenic Ras switches p73 expression from TAp73 to the oncogenic deltaNp73 isoform in a phosphatidyl-inositol 3-kinase-dependent manner. Our results implicate TAp73 as a barrier to anchorage-independent growth and indicate that downregulation of TAp73 is a key transforming activity of oncogenic Ras mutants.
Collapse
Affiliation(s)
- Michaela Beitzinger
- Molecular Tumor Biology Group, Rudolf-Virchow-Center, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
203
|
Hallstrom TC, Mori S, Nevins JR. An E2F1-dependent gene expression program that determines the balance between proliferation and cell death. Cancer Cell 2008; 13:11-22. [PMID: 18167336 PMCID: PMC2243238 DOI: 10.1016/j.ccr.2007.11.031] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 10/10/2007] [Accepted: 11/20/2007] [Indexed: 12/17/2022]
Abstract
The Rb/E2F pathway regulates the expression of genes essential for cell proliferation but that also trigger apoptosis. During normal proliferation, PI3K/Akt signaling blocks E2F1-induced apoptosis, thus serving to balance proliferation and death. We now identify a subset of E2F1 target genes that are specifically repressed by PI3K/Akt signaling, thus distinguishing the E2F1 proliferative or apoptotic function. RNAi-mediated inhibition of several of these PI3K-repressed E2F1 target genes, including AMPK alpha 2, impairs apoptotic induction by E2F1. Activation of AMPK alpha 2 with an AMP analog further stimulates E2F1-induced apoptosis. We also show that the presence of the E2F1 apoptotic expression program in breast and ovarian tumors coincides with good prognosis, emphasizing the importance of the balance in the E2F1 proliferation/apoptotic program.
Collapse
Affiliation(s)
- Timothy C Hallstrom
- Department of Pediatrics, Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
204
|
Abstract
Tumour necrosis factor-alpha (TNF-alpha) is a cytokine that is involved in many functions, including the inflammatory response, immunity and apoptosis. Some of the responses of TNF-alpha are mediated by caspase-1, which is involved in the production of the pro-inflammatory cytokines interleukin-1beta, interleukin-18 and interleukin-33. The molecular mechanisms involved in TNF-alpha-induced caspase-1 gene expression remain poorly defined, despite the fact that signaling by TNF-alpha has been well studied. The present study was undertaken to investigate the mechanisms involved in the induction of caspase-1 gene expression by TNF-alpha. Treatment of A549 cells with TNF-alpha resulted in an increase in caspase-1 mRNA and protein expression, which was preceded by an increase in interferon regulatory factor-1 and p73 protein levels. Caspase-1 promoter reporter was activated by the treatment of cells with TNF-alpha. Mutation of the interferon regulatory factor-1 binding site resulted in the almost complete loss of basal as well as of TNF-alpha-induced caspase-1 promoter activity. Mutation of the p53/p73 responsive site resulted in reduced TNF-alpha-induced promoter activity. Blocking of p73 function by a dominant negative mutant or by a p73-directed small hairpin RNA reduced basal as well as TNF-alpha-induced caspase-1 promoter activity. TNF-alpha-induced caspase-1 mRNA and protein levels were reduced when p73 mRNA was down-regulated by small hairpin RNA. Caspase-5 gene expression was induced by TNF-alpha, which was inhibited by the small hairpin RNA-mediated down-regulation of p73. Our results show that TNF-alpha induces p73 gene expression, which, together with interferon regulatory factor-1, plays an important role in mediating caspase-1 promoter activation by TNF-alpha.
Collapse
Affiliation(s)
- Nishant Jain
- Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | |
Collapse
|
205
|
Deregulated E2f-2 underlies cell cycle and maturation defects in retinoblastoma null erythroblasts. Mol Cell Biol 2007; 27:8713-28. [PMID: 17923680 DOI: 10.1128/mcb.01118-07] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
By assessing the contribution of deregulated E2F activity to erythroid defects in Rb null mice, we have identified E2f-2 as being upregulated in end-stage red cells, where we show it is the major pRb-associated E2f and the predominant E2f detected at key target gene promoters. Consistent with its expression pattern, E2f-2 loss restored terminal erythroid maturation to Rb null red cells, including the ability to undergo enucleation. Deletion of E2f-2 also extended the life span of Rb null mice despite persistent defects in placental development, indicating that deregulated E2f-2 activity in differentiating erythroblasts contributes to the premature lethality of Rb null mice. We show that the aberrant entry of Rb null erythroblasts into S phase at times in differentiation when wild-type erythroblasts are exiting the cell cycle is inhibited by E2f-2 deletion. E2f-2 loss induced cell cycle arrest in both wild-type and Rb null erythroblasts and was associated with increased DNA double-strand breaks. These results implicate deregulated E2f-2 in the cell cycle defects observed in Rb null erythroblasts and reveal a novel role for E2f-2 during terminal red blood cell differentiation. The identification of a tissue-restricted role for E2f-2 in erythropoiesis highlights the nonredundant nature of E2f transcription factor activities in cell growth and differentiation.
Collapse
|
206
|
Abstract
In contrast with the low frequency of alterations found in the Rb gene, the pRb pathway is inactivated in the vast majority of human tumors. A similar situation takes place in mouse models of cancer, including two-stage skin tumorigenesis. This might be explained if the Rb functions are carried out, in its absence, by other proteins that are also controlled by the same upstream regulators and display similar effectors. The other Rb family members, p107 and or p130, are plausible candidates. The embryonic lethality of pRb-deficient animals, which precludes the analysis of the roles of Rb gene in mouse models, has been avoided using tissue-specific deletion of pRb. In epidermis, pRb deletion leads to altered proliferation and differentiation. However, these deficient mice do not develop spontaneous tumors, and chemical carcinogenesis experiments revealed that the absence of pRb renders fewer and smaller tumors than control animals, but showing increased malignant conversion to squamous cell carcinomas (SCC). Detailed biochemical analyses have indicated that, in the absence of pRb, multiple pathways, including the aberrant p53 activation mediated by E2F/p19(ARF), are activated leading to increased tumor apoptosis. As Rb loss in epidermis is functionally compensated by Rbl1 (p107), this might also suggest that p107 could behave as a tumor suppressor. We summarize here our findings in support of this hypothesis. The pRb-;p107-/- epidermis form spontaneous tumors, and the reduction of p107 levels restores the susceptibility of pRb-mice to chemical skin carcinogenesis experiments. Moreover, Rb-deficient keratinocytes are highly susceptible to Ha-ras-induced transformation, and this susceptibility is enhanced by p107 loss. Further functional studies have indicated that the loss of p107 in the absence of pRb produces the reduction of p53-dependent proapoptotic signals through the modulation of p63 and p73 isoforms. In addition, expression profiling analysis has revealed multiple oncogenic alterations that can contribute to tumor susceptibility in epidermis in the absence of pRb and p107.
Collapse
|
207
|
Tracy K, Dibling BC, Spike BT, Knabb JR, Schumacker P, Macleod KF. BNIP3 is an RB/E2F target gene required for hypoxia-induced autophagy. Mol Cell Biol 2007; 27:6229-42. [PMID: 17576813 PMCID: PMC1952167 DOI: 10.1128/mcb.02246-06] [Citation(s) in RCA: 308] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Revised: 12/29/2006] [Accepted: 06/10/2007] [Indexed: 12/18/2022] Open
Abstract
Hypoxia and nutrient deprivation are environmental stresses governing the survival and adaptation of tumor cells in vivo. We have identified a novel role for the Rb tumor suppressor in protecting against nonapoptotic cell death in the developing mouse fetal liver, in primary mouse embryonic fibroblasts, and in tumor cell lines. Loss of pRb resulted in derepression of BNip3, a hypoxia-inducible member of the Bcl-2 superfamily of cell death regulators. We identified BNIP3 as a direct target of pRB/E2F-mediated transcriptional repression and showed that pRB attenuates the induction of BNIP3 by hypoxia-inducible factor to prevent autophagic cell death. BNIP3 was essential for hypoxia-induced autophagy, and its ability to promote autophagosome formation was enhanced under conditions of nutrient deprivation. Knockdown of BNIP3 reduced cell death, and remaining deaths were necrotic in nature. These studies identify BNIP3 as a key regulator of hypoxia-induced autophagy and suggest a novel role for the RB tumor suppressor in preventing nonapoptotic cell death by limiting the extent of BNIP3 induction in cells.
Collapse
Affiliation(s)
- Kristin Tracy
- The Ben May Department for Cancer Research, The Gordon Center for Integrative Sciences, W-338, The University of Chicago, 929 E 57th St., Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
208
|
Tian C, Lv D, Qiao H, Zhang J, Yin YH, Qian XP, Wang YP, Zhang Y, Chen WF. TFDP3 inhibits E2F1-induced, p53-mediated apoptosis. Biochem Biophys Res Commun 2007; 361:20-5. [PMID: 17632080 DOI: 10.1016/j.bbrc.2007.06.128] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2007] [Accepted: 06/19/2007] [Indexed: 02/01/2023]
Abstract
By dimerizing with E2F proteins, TFDP has profound influence on cellular E2F activities. While TFDP1 and 2 enhance the DNA binding and the transcriptional activity of E2F, the newly identified member of the DP family, TFDP3 primarily functions as a negative regulator. To further characterize the inhibitory property of TFDP3, the present study specifically examined the modulatory role of TFDP3 on E2F1-induced cell death. HEK-293 cells underwent apoptosis following ectopic expression of E2F1. This effect was virtually abolished by co-transfection with TFDP3. In the meantime, the accumulation of p53 proteins and the increased expression of the pro-apoptotic molecules, including Bax, Puma, Noxa, and Bid were found to be suppressed. These data suggest a new mechanism for the regulation of E2F1-induced apoptosis and provide further evidence for the general involvement of TFDP3 in the regulation of E2F functions.
Collapse
Affiliation(s)
- Chan Tian
- Department of Immunology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, China
| | | | | | | | | | | | | | | | | |
Collapse
|
209
|
Lee SW, Kim EJ, Um SJ. Transcriptional regulation of the p73 gene, a member of the p53 family, by early growth response-1 (Egr-1). Biochem Biophys Res Commun 2007; 362:1044-50. [PMID: 17822668 DOI: 10.1016/j.bbrc.2007.08.128] [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] [Received: 08/10/2007] [Accepted: 08/19/2007] [Indexed: 01/20/2023]
Abstract
To elucidate the regulatory mechanism of p73 gene expression, we analyzed the human p73 promoter and found three putative Egr-1-binding sites located upstream of exon 1 (-1728, -321, and -38). The Egr-1 responsiveness of these sites was analyzed by transient transfection assays using 5'- and 3'-serial truncations of the p73 promoter, subcloned in a CAT reporter vector. The functional significance of the region was further confirmed by an electrophoretic mobility shift assay using the Egr-1 protein synthesized in vitro and a [32P]-labeled middle site sequence, followed by competition with unlabeled wild-type or mutant oligonucleotides and supershift assays using an anti-Egr-1 antibody. When induced by either the nitric oxide donor NOC-18 or the PPARgamma agonist troglitazone, Egr-1 bound to the p73 promoter, as assessed by chromatin immunoprecipitation assays, accompanied by increased expression of p73. MTT assays revealed that cell growth was significantly inhibited on treating the cells with troglitazone. Overall, our results provide direct evidence that Egr-1 positively regulated p73 expression by binding to its promoter in vivo, consistent with Egr-1 and p73 being involved in p53-independent tumor suppression.
Collapse
Affiliation(s)
- Sang-Wang Lee
- Department of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, 98 Kunja-dong, Kwangjin-gu, Seoul 143-747, Republic of Korea
| | | | | |
Collapse
|
210
|
Wang J, Yin DP, Liu YX, Baer R, Yin Y. Dual specificity phosphatase 1/CL100 is a direct transcriptional target of E2F-1 in the apoptotic response to oxidative stress. Cancer Res 2007; 67:6737-44. [PMID: 17638884 DOI: 10.1158/0008-5472.can-06-4402] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
E2F-1 mediates apoptosis through transcriptional regulation of its targets. We report here that E2F-1 acts as a direct transcriptional regulator of dual specificity phosphatase 1 (DUSP1; CL100), a threonine and tyrosine phosphatase that inhibits mitogen-activated protein (MAP) kinases. We found that DUSP1 is transcriptionally induced by ectopic E2F-1 expression and that extracellular signal-regulated kinase 1/2 are dephosphorylated in the presence of E2F-1 and DUSP1. E2F-1 mediates apoptosis in the cellular response to oxidative stress. DUSP1 levels are significantly increased in an E2F-1-dependent manner following oxidative stress but not other stresses examined. DUSP1 mediates the cellular response to oxidative stress. We found that E2F-1 binds to chromatin encompassing the DUSP1 promoter and greatly stimulates the promoter activity of the DUSP1 gene. In particular, E2F-1 physically binds to an E2F-1 consensus sequence and a palindromic motif in the DUSP1 promoter. Interestingly, E2F-1 is acetylated following oxidative stress. Our findings show that E2F-1 is a transcriptional activator of DUSP1 and that DUSP1 is a link between E2F-1 and MAP kinases.
Collapse
Affiliation(s)
- Jianli Wang
- Department of Radiation Oncology, Center for Radiological Research, College of Physicians and Surgeons, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
211
|
Lau LMS, Nugent JK, Zhao X, Irwin MS. HDM2 antagonist Nutlin-3 disrupts p73-HDM2 binding and enhances p73 function. Oncogene 2007; 27:997-1003. [PMID: 17700533 DOI: 10.1038/sj.onc.1210707] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Nutlin-3, a small molecule inhibitor, activates p53 by disrupting p53-HDM2 association. In this study, we found that Nutlin-3 suppressed cell growth and induced apoptosis in the absence of wild-type p53, suggesting a p53-independent mechanism for Nutlin-3-induced cell death. Like p53, its homolog p73 transactivates proapoptotic genes and induces cell death. Since HDM2, a key negative regulator of p53, also binds to and inhibits p73, we asked whether p73 could mediate Nutlin-3-induced apoptosis. We demonstrate that Nutlin-3 inhibits endogenous binding between the proapoptotic p73 isoform TAp73alpha and HDM2 in p53-null cells. Dissociation of p73 and HDM2 leads to increased p73 transcriptional activity with upregulation of p73 target genes noxa, puma and p21, as well as enhanced apoptosis. p73 knockdown by siRNA results in rescue of Nutlin-3-treated cells, indicating that Nutlin-3-induced apoptosis is, at least in part, p73 dependent. In addition, Nutlin-3 treatment increases TAp73alpha protein levels with prolongation of p73 half-life. These results provide the first evidence that Nutlin-3 disrupts endogenous p73-HDM2 interaction and enhances the stability and proapoptotic activities of p73 and thus, provides a rationale for the use of Nutlin-3 in the large number of human tumors in which p53 is inactivated.
Collapse
Affiliation(s)
- L M S Lau
- Division of Hematology/Oncology, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
212
|
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
|
213
|
Verdaguer E, Susana GDA, Clemens A, Pallàs M, Camins A. Implication of the transcription factor E2F-1 in the modulation of neuronal apoptosis. Biomed Pharmacother 2007; 61:390-9. [PMID: 17178208 DOI: 10.1016/j.biopha.2006.11.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Accepted: 11/07/2006] [Indexed: 02/05/2023] Open
Abstract
Neurodegenerative diseases as Alzheimer's disease, Parkinson's disease and other neurological disorders remain major problem worldwide since is currently no effective treatment. Thus, studying the mechanisms involved in neuronal apoptotic pathways is imperative if drugs that might stop or delay these disease processes are to be synthesized. In recent years it has become evident that mitochondria are key component of the neuronal apoptotic route. In addition to mitochondria, other intracellular components have been implicated in this process. Thus, DNA damage and re-entry into the cell cycle may constitute a common pathway in apoptosis in neurological diseases. The implication of cell cycle in neurodegenerative disorders is supported by data on the brain of patients who showed an increase in cell cycle protein expression. Indeed, studies performed in neuronal cell preparations indicate that re-entry into the cell cycle and, more specifically, an increase in the expression of E2F-1 transcription role of DNA damage/repair as a potential mechanism in cell cycle re-entry. In this context, ataxia telangiectasia mutated protein could be the enzyme responsible for neuronal apoptosis activation. Furthermore, the potential routes involved in E2F-1 induced apoptosis, p53-dependent and p53-independent, are similarly reviewed. Under this hypothesis, multiple pathways have been suggested, including the route of caspases. Finally, given the increasing experimental data on the neuroprotective and antiapoptotic effects of cyclin dependent kinase CDK inhibitory drugs, including flavopiridol, their application for the treatment of neurological disorders is proposed.
Collapse
Affiliation(s)
- Ester Verdaguer
- Departament de Farmacologia i Toxicologia, IIBB-CSIC, IDIBAPS, Rossello 161, Planta 6, 08036 Barcelona, Spain
| | | | | | | | | |
Collapse
|
214
|
Talos F, Nemajerova A, Flores ER, Petrenko O, Moll UM. p73 Suppresses Polyploidy and Aneuploidy in the Absence of Functional p53. Mol Cell 2007; 27:647-59. [PMID: 17707235 DOI: 10.1016/j.molcel.2007.06.036] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 02/19/2007] [Accepted: 06/25/2007] [Indexed: 11/25/2022]
Abstract
Previous studies showed that p53 plays a central role in G1 and DNA damage checkpoints, thus contributing to genomic stability. We show here that p73 also plays a role in genomic integrity but this mechanism is manifest only when p53 is lost. Isolated p73 loss in primary cells does not induce genomic instability. Instead, it results in impaired proliferation and premature senescence due to compensatory activation of p53. Combined loss of p73 and p53 rescues these defects, but at the expense of exacerbated genomic instability. This leads to rapid increase in polyploidy and aneuploidy, markedly exceeding that of p53 loss alone. Constitutive deregulation of cyclin-Cdk activities and excess failure of the G2/M DNA damage checkpoint appear to fuel increased ploidy abnormalities upon p53/p73 loss, while primary mitotic defects do not play a causal role. These data indicate that p73 is essential for suppressing polyploidy and aneuploidy when p53 is inactivated.
Collapse
Affiliation(s)
- Flaminia Talos
- Department of Pathology, Health Science Center, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | | | | | | | | |
Collapse
|
215
|
Abstract
Defects in apoptotic programs contribute to a number of human diseases, ranging from neurodegenerative disorders to malignancy, and treatment failure. The genetic basis for apoptosis implies that cell death can be disrupted by mutations, raising the intriguing possibility that cell numbers can be regulated by factors that influence cell survival. It is well documented that the E2F1 transcription factor is a key regulator of apoptotic programs. E2F1-induced cell death occurs via multiple pathways, some of which involve the tumour suppressor p53, and autonomous of p53. This has led to the opinion that E2F1 functions as a tumour surveillance factor, detecting aberrant proliferation and engaging apoptotic pathways to protect the organism from developing tumours. Frequently, novel players are discovered that expand the interpretation of apoptosis control by E2F1. This information will help to produce new strategies to exploit E2F1-induced apoptosis for therapeutic benefit.
Collapse
Affiliation(s)
- B M Pützer
- Department of Vectorology and Experimental Gene Therapy, University of Rostock, Biomedical Research Center, Schillingallee 69, D-18055 Rostock, Germany.
| |
Collapse
|
216
|
Abstract
Although chemotherapy can induce complete responses in patients with chronic lymphocytic leukemia (CLL), it is not considered curative. Treated patients generally develop recurrent disease requiring additional therapy, which can cause worsening immune dysfunction, myelosuppression, and selection for chemotherapy-resistant leukemia-cell subclones. Cellular immune therapy promises to mitigate these complications and potentially provide for curative treatment. Most experience with this is in the use of allogeneic hematopoietic stem-cell transplantation (allo-HSCT), in which graft-versus-leukemia (GVL) effects can be observed and shown responsible for long-term disease-free survival. However, use of allo-HSCT for CLL is limited because of the lack of suitable donors and the treatment-related morbidity/mortality for elderly patients, who constitute the majority at risk for developing this disease. The GVL effect, however, suggests there are specific CLL-associated antigens that could be targeted in autologous cellular immune therapy. Effective strategies for this will have to overcome the disease-related acquired immune deficiency and the capacity of the leukemia-cell to induce T-cell tolerance, thereby compromising the activity of even conventional vaccines in patients with this disease. We will discuss the different strategies being developed to overcome these limitations that might provide for effective cellular immune therapy of CLL.
Collapse
Affiliation(s)
- Arnon P Kater
- Department of Hematology, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | |
Collapse
|
217
|
Niikura Y, Dixit A, Scott R, Perkins G, Kitagawa K. BUB1 mediation of caspase-independent mitotic death determines cell fate. ACTA ACUST UNITED AC 2007; 178:283-96. [PMID: 17620410 PMCID: PMC2064447 DOI: 10.1083/jcb.200702134] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The spindle checkpoint that monitors kinetochore–microtubule attachment has been implicated in tumorigenesis; however, the relation between the spindle checkpoint and cell death remains obscure. In BUB1-deficient (but not MAD2-deficient) cells, conditions that activate the spindle checkpoint (i.e., cold shock or treatment with nocodazole, paclitaxel, or 17-AAG) induced DNA fragmentation during early mitosis. This mitotic cell death was independent of caspase activation; therefore, we named it caspase-independent mitotic death (CIMD). CIMD depends on p73, a homologue of p53, but not on p53. CIMD also depends on apoptosis-inducing factor and endonuclease G, which are effectors of caspase-independent cell death. Treatment with nocodazole, paclitaxel, or 17-AAG induced CIMD in cell lines derived from colon tumors with chromosome instability, but not in cells from colon tumors with microsatellite instability. This result was due to low BUB1 expression in the former cell lines. When BUB1 is completely depleted, aneuploidy rather than CIMD occurs. These results suggest that cells prone to substantial chromosome missegregation might be eliminated via CIMD.
Collapse
Affiliation(s)
- Yohei Niikura
- Department of Molecular Pharmacology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | | | | | |
Collapse
|
218
|
Amin ARMR, Paul RK, Thakur VS, Agarwal ML. A Novel Role for p73 in the Regulation of Akt-Foxo1a-Bim Signaling and Apoptosis Induced by the Plant Lectin, Concanavalin A. Cancer Res 2007; 67:5617-21. [PMID: 17575126 DOI: 10.1158/0008-5472.can-07-0655] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Virtually all human cancers encounter disruption of the "p53 network." From a therapeutic point of view, it is important to devise strategies that eliminate cancer cells, which are often defective in functional p53 and protect p53-expressing normal cells. By comparing the response of a pair of isogenic cell lines, we identify a plant-derived compound, Concanavalin A (Con A), which differentially kills p53-null cells. Further, we find that p53 family member, p73, plays a critical role that is unmasked in the absence of p53. Con A treatment leads to induction of p73 and several others that are important mediators of apoptosis and act downstream, such as p21, Bax, Foxo1a, and Bim. Inactivation of p73 reverses the expression of these proteins and apoptosis. Inhibition of Akt activation sensitizes otherwise resistant cells. These observations thus reveal a novel role for p73 in the regulation of Akt-Foxo1a-Bim signaling and apoptosis especially when p53 is absent.
Collapse
Affiliation(s)
- A R M Ruhul Amin
- Department of Genetics and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
219
|
Wang J, Shen WH, Jin YJ, Brandt-Rauf PW, Yin Y. A Molecular Link between E2F-1 and the MAPK Cascade. J Biol Chem 2007; 282:18521-18531. [PMID: 17452331 DOI: 10.1074/jbc.m610538200] [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] [Indexed: 11/06/2022] Open
Abstract
Transcription factor E2F-1 mediates apoptosis and suppresses tumorigenesis. The mechanisms by which E2F-1 functions in these processes are largely unclear. We report here that E2F-1 acts as a transcriptional regulator of MKP-2 (MAPK phosphatase-2), a dual specificity protein phosphatase (DUSP4) with stringent substrate specificity for MAPKs. We show that E2F-1 is required for the cellular apoptotic response to oxidative damage. MKP-2 is greatly increased following oxidative stress, and E2F-1 is necessary for that induction. We found that E2F-1 is physically associated with the MKP-2 promoter and can transactivate the promoter of the MKP-2 gene. Specifically, E2F-1 binds to a perfect palindromic motif in the MKP-2 promoter. Finally, we show that this E2F-1/MKP-2 pathway mediates apoptosis under oxidative stress and that MKP-2 suppresses tumor formation in nude mice. Our findings demonstrate that E2F-1 is a transcriptional activator of MKP-2 and that MKP-2 is an essential cell death mediator in the E2F-1 pathway. Characterization of MKP-2 as a cell death mediator may lead to the development of new strategies for cancer treatment.
Collapse
Affiliation(s)
- Jianli Wang
- Department of Radiation Oncology, College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Wen Hong Shen
- Department of Radiation Oncology, College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Yan J Jin
- Department of Radiation Oncology, College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Paul W Brandt-Rauf
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032
| | - Yuxin Yin
- Department of Radiation Oncology, College of Physicians and Surgeons, Columbia University, New York, New York 10032; Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032.
| |
Collapse
|
220
|
Ma Y, Cress WD. Transcriptional upregulation of p57 (Kip2) by the cyclin-dependent kinase inhibitor BMS-387032 is E2F dependent and serves as a negative feedback loop limiting cytotoxicity. Oncogene 2007; 26:3532-40. [PMID: 17173074 PMCID: PMC2128050 DOI: 10.1038/sj.onc.1210143] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Revised: 09/06/2006] [Accepted: 10/14/2006] [Indexed: 11/08/2022]
Abstract
In spite of the fact that cyclin-dependent kinase (cdk) inhibiting drugs are potent transcriptional repressors, we discover that p57 (Kip2, CDKN1C) transcription is significantly upregulated by three small molecule cdk inhibitors, including BMS-387032. Treatment of MDA-MB-231 breast cancer cells with BMS-387032 led to a stabilization of the E2F1 protein that was accompanied by significant increases in the p57 mRNA and protein. This increase did not occur in an E2F1-deficient cell line. An E2F1-estrogen receptor fusion protein activated the endogenous p57 promoter in response to hydroxytamoxifen treatment in the presence of cycloheximide. Luciferase constructs driven by the p57 promoter verified that upregulation of p57 mRNA by BMS-387032 is transcriptional and dependent on E2F-binding sites in the promoter. Expression of exogenous p57 significantly decreased the fraction of cells in S phase. Furthermore, p57-deficient MDA-MB-231 cell lines were significantly more sensitive to BMS-387032-induced apoptosis than controls. The results presented in this manuscript demonstrate that small molecule cdk inhibitors transcriptionally activate p57 dependent upon E2F1 and that this activation in turn serves to limit E2F1's death-inducing activity.
Collapse
Affiliation(s)
- Y Ma
- Molecular Oncology Program, H Lee Moffitt Cancer Center and Research Institute, University of South Florida, College of Medicine, Tampa, FL 33612-9497, USA
| | | |
Collapse
|
221
|
Wu J, Jin YJ, Calaf GM, Huang WL, Yin Y. PAC1 is a direct transcription target of E2F-1 in apoptotic signaling. Oncogene 2007; 26:6526-35. [PMID: 17471234 DOI: 10.1038/sj.onc.1210484] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
E2F-1 controls multiple cellular activities through transcriptional regulation of its target genes. As a mediator of cell death, E2F-1 can eliminate latent neoplastic cells through apoptosis. However, the mechanism by which E2F-1 mediates cancer cell killing is largely unknown. In this paper, we report that phosphatase of activated cells 1 (PAC1) phosphatase is a direct transcription target of E2F-1 in signaling apoptosis. We show that ectopic E2F-1 increases expression of PAC1 at both transcriptional and translational levels in breast cancer cells. E2F-1 physically interacts with the promoter of PAC1, binds to its consensus sequence in the promoter and transactivates the PAC1 promoter. E2F-1 suppresses extracellular signal-regulated kinase (ERK) phosphorylation through PAC1 and causes cancer cell death by apoptosis following treatment with a chemotherapeutic agent N-4-hydroxyphenylretinamide (4-HPR). Furthermore, ectopic PAC1 inhibits ERK phosphorylation and mediates cell killing. Moreover, endogenous E2F-1 upregulates PAC1 and suppresses ERK activity, leading to cell death in response to 4-HPR. These results reveal a crucial role of PAC1 in E2F-1-directed apoptosis. Our study demonstrates that E2F-1 mediates apoptosis through transcriptional regulation of PAC1 and subsequent suppression of the ERK signaling. Our findings establish a functional link between E2F-1 and mitogen-activated protein kinases. The E2F-1-PAC1 cascade in cancer cell killing may provide a molecular basis for cancer therapeutic intervention.
Collapse
Affiliation(s)
- J Wu
- Department of Radiation Oncology, Center for Radiological Research, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
222
|
Chen G, Tai AK, Lin M, Chang F, Terhorst C, Huber BT. Increased proliferation of CD8+ T cells in SAP-deficient mice is associated with impaired activation-induced cell death. Eur J Immunol 2007; 37:663-74. [PMID: 17266174 DOI: 10.1002/eji.200636417] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Defective signaling lymphocyte activation molecule (SLAM)-associated protein (SAP) is responsible for the human X-linked lymphoproliferative syndrome. Defects in T helper 2, natural killer, natural killer T and B cells have been demonstrated in SAP-deficient humans and mice, and increased proliferation of CD8+ T cells has been observed. In the current study, we investigated the properties of CD8+ T cell proliferation and activation-induced cell death (AICD), using OT-I T cell receptor (TCR)-transgenic mice on either wild-type (WT) or SAP-/- background. Interestingly, we found that ovalbumin peptide-activated SAP-/- CD8+ T cells have lower AICD compared to their WT counterparts. Furthermore, the induction of p73, a key mediator of TCR-induced apoptosis through the mitochondrial apoptotic pathway, was significantly reduced at both the mRNA and protein levels in the activated mutant cells. Meanwhile, a reduced level of activated caspase 9 was observed in the mutant cells. We conclude that reduced AICD in activated SAP-/- CD8+ T cells is associated with impaired p73 induction, indicating that the initiation of the mitochondrial apoptotic pathway might be impaired. Our data demonstrate an intrinsic defect in SAP-/- CD8+ T cells and shed light on the increased responsiveness of CD8+ T cells in SAP-/- mice.
Collapse
Affiliation(s)
- Gang Chen
- Department of Pathology, Tufts University School of Medicine, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
223
|
Marabese M, Vikhanskaya F, Broggini M. p73: a chiaroscuro gene in cancer. Eur J Cancer 2007; 43:1361-72. [PMID: 17428654 DOI: 10.1016/j.ejca.2007.01.042] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Revised: 01/19/2007] [Accepted: 01/25/2007] [Indexed: 11/24/2022]
Abstract
p73 is a member of the p53 family which is gaining increasing importance in the field of cancer. Its structural homology with p53 led to the assumption that it could act as a new tumour suppressor gene. Increasing knowledge of its function, however, has cast doubts on this role. A particularly interesting characteristic of p73 is that the cell contains different isoforms with distinct and sometimes opposite functions. Evidence in the last few years clearly indicates that p73 does share some activities with p53 but also that it has some distinct functions. This review focuses on p73's role in the development and progression of cancer, analysing the gene structure and regulation and discussing similarities with p53 and differences. Recent results obtained with specific detection methods on the levels and functions of the different isoforms in tumours are also discussed.
Collapse
Affiliation(s)
- Mirko Marabese
- Laboratory of Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri, Via Eritrea 62, 20157 Milan, Italy.
| | | | | |
Collapse
|
224
|
Boominathan L. Some facts and thoughts: p73 as a tumor suppressor gene in the network of tumor suppressors. Mol Cancer 2007; 6:27. [PMID: 17407586 PMCID: PMC1853109 DOI: 10.1186/1476-4598-6-27] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2007] [Accepted: 04/03/2007] [Indexed: 12/30/2022] Open
Abstract
The question of whether p73 is a tumor suppressor gene, is not yet answered with full confidence. The lack of spontaneous tumor formation in p73 null mice and infrequent p73 mutations seen in a variety of cancers analyzed would straightaway negate its role as a primary tumor suppressor gene. However, accumulating evidence suggest that p73 gene and its target genes are hypermethylated in the cancer of lymphoid origin. Here I discuss some facts and thoughts that support the idea that p73 could still be a tumor suppressor gene. The tumor suppressor network in which p73 appears to be a participant involves E2F1, JunB, INK4a/p16, ARF/p19, p57kip2 and BRCA1. Knock out of each gene in E2F-1-p73-JunB-p16INK4a network of tumor suppressor proteins result in lymphoma/leukemia formation. Further, I tried to explain why lymphomas are not seen in p73 null mice and why p73 gene is not prone to frequent mutation.
Collapse
|
225
|
Amin ARMR, Thakur VS, Paul RK, Feng GS, Qu CK, Mukhtar H, Agarwal ML. SHP-2 tyrosine phosphatase inhibits p73-dependent apoptosis and expression of a subset of p53 target genes induced by EGCG. Proc Natl Acad Sci U S A 2007; 104:5419-24. [PMID: 17369354 PMCID: PMC1838507 DOI: 10.1073/pnas.0700642104] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Green tea polyphenol, epigallocatechin-3-gallate (EGCG) differentially regulates the cellular growth of cancer cells in a p53-dependent manner through apoptosis and/or cell cycle arrest. In an effort to further elucidate the mechanism of differential growth regulation by EGCG, we have investigated the role of the tyrosine phosphatase, SHP-2. Comparing the responses of mouse embryonic fibroblasts (MEFs), expressing either WT or functionally inactive/truncated SHP-2, we find that inactivation of SHP-2 remarkably sensitizes cells to EGCG-mediated killing. MEFs lacking functional SHP-2 undergo massive apoptosis upon treatment with EGCG. By comparing gene expression profiles, we have identified a set of transcriptional targets of p53 that are differentially modulated in cells undergoing apoptosis. Western blot and real-time PCR analyses of a select group of genes further confirm that the expression is SHP-2-dependent. Similar observations were made in MEFs lacking p53, confirming that the expression of these "p53 target genes" is p53-independent. In addition, EGCG treatment induced the expression of p73 mRNA and protein in both cell types, but not p63. Inactivation of p73 in cells expressing nonfunctional SHP-2 markedly inhibited apoptosis and p53 target gene expression. Although phosphorylation of JNK is differentially regulated by SHP2, it was found to be dispensable for EGCG-induced apoptosis and p53 target gene expression. Our results have identified SHP-2 as a negative regulator of EGCG-induced-apoptosis and have identified a subset of p53 target genes whose expression is paradoxically not mediated by p53 but by one of its family members, p73.
Collapse
Affiliation(s)
| | | | | | | | - Cheng-Kui Qu
- Hematology and Oncology, Case Western Reserve University, Cleveland, OH 44106
| | | | - Munna L. Agarwal
- Departments of *Genetics and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
226
|
Abstract
DNA double-strand breaks (DSBs) produce a number of cellular responses, some mutually exclusive. Depending on where on the chromosome it occurs, a DSB may become preserved inside a telomere or eliminated by repair. A cell may arrest division via checkpoint activation to fix DSBs or commit suicide by apoptosis. What determines the outcome: to bury, fix, or succumb to DNA DSBs? With this question in mind, we review recent data on cellular responses to DSBs.
Collapse
Affiliation(s)
- Tin Tin Su
- Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309-0347, USA.
| |
Collapse
|
227
|
Abstract
The p53-related genes p63 and p73 exhibit significant structural homology to p53; however, they do not function as classical tumor suppressors and are rarely mutated in human cancers. Both p63 and p73 exhibit tissue-specific roles in normal development and a complex contribution to tumorigenesis that is due to their expression as multiple protein isoforms. The predominant p63/p73 isoforms expressed both in normal development and in many tumors lack the conserved transactivation (TA) domain; these isoforms instead exhibit a truncated N-terminus (DeltaN) and function at least in part as transcriptional repressors. p63 and p73 isoforms are regulated through both transcriptional and post-translational mechanisms, and they in turn regulate diverse cellular functions including proliferation, survival and differentiation. The net effect of p63/p73 expression in a given context depends on the ratio of TA/DeltaN isoforms expressed, on physical interaction between p63 and p73 isoforms, and on functional interactions with p53 at the promoters of specific downstream target genes. These multifaceted interactions occur in diverse ways in tumor-specific contexts, demonstrating a functional 'p53 family network' in human tumorigenesis. Understanding the regulation and mechanistic contributions of p63 and p73 in human cancers may ultimately provide new therapeutic opportunities for a variety of these diseases.
Collapse
Affiliation(s)
- M P Deyoung
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| | | |
Collapse
|
228
|
Vikhanskaya F, Lee MK, Mazzoletti M, Broggini M, Sabapathy K. Cancer-derived p53 mutants suppress p53-target gene expression--potential mechanism for gain of function of mutant p53. Nucleic Acids Res 2007; 35:2093-104. [PMID: 17344317 PMCID: PMC1874625 DOI: 10.1093/nar/gkm099] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Tumour-derived p53 mutants are thought to have acquired 'gain-of-function' properties that contribute to oncogenicity. We have tested the hypothesis that p53 mutants suppress p53-target gene expression, leading to enhanced cellular growth. Silencing of mutant p53 expression in several human cell lines was found to lead to the upregulation of wild-type p53-target genes such as p21, gadd45, PERP and PTEN. The expression of these genes was also suppressed in H1299-based isogenic cell lines expressing various hot-spot p53 mutants, and silencing of mutant p53, but not TAp73, abrogated the suppression. Consistently, these hot-spot p53 mutants were able to suppress a variety of p53-target gene promoters. Analysis using the proto-type p21 promoter construct indicated that the p53-binding sites are dispensable for mutant p53-mediated suppression. However, treatment with the histone deacetylase inhibitor trichostatin-A resulted in relief of mutant p53-mediated suppression, suggesting that mutant p53 may induce hypo-acetylation of target gene promoters leading to the suppressive effects. Finally, we show that stable down-regulation of mutant p53 expression resulted in reduced cellular colony growth in human cancer cells, which was found to be due to the induction of apoptosis. Together, the results demonstrate another mechanism through which p53 mutants could promote cellular growth.
Collapse
Affiliation(s)
- Faina Vikhanskaya
- Laboratory of Molecular Carcinogenesis, National Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore, Department of Biochemistry, National University of Singapore, 10, Kent Ridge Crescent, Singapore 119260 and Department of Oncology, Mario Negri Institute for Pharmacological Research, Via Eritrea No 62, 20154 Milan, Italy
| | - Ming Kei Lee
- Laboratory of Molecular Carcinogenesis, National Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore, Department of Biochemistry, National University of Singapore, 10, Kent Ridge Crescent, Singapore 119260 and Department of Oncology, Mario Negri Institute for Pharmacological Research, Via Eritrea No 62, 20154 Milan, Italy
| | - Marco Mazzoletti
- Laboratory of Molecular Carcinogenesis, National Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore, Department of Biochemistry, National University of Singapore, 10, Kent Ridge Crescent, Singapore 119260 and Department of Oncology, Mario Negri Institute for Pharmacological Research, Via Eritrea No 62, 20154 Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Carcinogenesis, National Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore, Department of Biochemistry, National University of Singapore, 10, Kent Ridge Crescent, Singapore 119260 and Department of Oncology, Mario Negri Institute for Pharmacological Research, Via Eritrea No 62, 20154 Milan, Italy
| | - Kanaga Sabapathy
- Laboratory of Molecular Carcinogenesis, National Cancer Centre, 11 Hospital Drive, Singapore 169610, Singapore, Department of Biochemistry, National University of Singapore, 10, Kent Ridge Crescent, Singapore 119260 and Department of Oncology, Mario Negri Institute for Pharmacological Research, Via Eritrea No 62, 20154 Milan, Italy
- *To whom correspondence should be addressed. +65 6436 8349+65 6226 5694
| |
Collapse
|
229
|
Dong YB, Phelps AM, Yang HL, Jamshidi-Parsian A, Chen C, Hao H, Gomez-Gutierrez JG, Zhou HS, McMasters KM. Induction of apoptosis signal-regulating Kinase 1 by E2F-1 may not be essential for E2F-1-mediated apoptosis in melanoma cells. Tumour Biol 2007; 28:111-22. [PMID: 17287612 DOI: 10.1159/000099370] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2006] [Accepted: 08/18/2006] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES In the present study, we investigate the role of apoptosis signal-regulating kinase 1 (ASK1) mitogen-activated protein (MAP) kinase signal pathways in E2F-1-mediated apoptosis. METHODS A gene expression profile in response to E2F-1 overexpression was performed by cDNA microarray analysis and confirmed by real-time reverse-transcription polymerase chain reaction. Kinase activities were assayed by Western blot analysis or kinase assay. Apoptosis was assessed by morphologic inspection and flow-cytometric analysis. Cytotoxicity was monitored by MTT assay. RESULTS E2F-1 upregulated the expression of ASK1 8-fold compared to the Ad-LacZ-infected control in SK-MEL-2 melanoma cells, which was confirmed by reverse-transcription polymerase chain reaction. Sequence analysis showed that there are 2 putative E2F-1 DNA binding sites in the ASK1 promoter region. Truncated E2F-1 protein, which lacks the transactivation domain, failed to upregulate ASK1, suggesting that ASK1 was regulated at the transcriptional level by E2F-1. E2F-1 overexpression resulted in the transient activation of c-Jun N-terminal kinase (JNK); however, dominant negative mutant ASK1 had no effect on E2F-1 cytotoxicity and JNK activation. p38 was not activated by E2F-1, and inhibition of p38 had no effect on E2F-1-mediated cell death. The ASK1 kinase assay showed that ASK1 activity was not upregulated in response to E2F1 overexpression. The inhibition of ASK1 upstream kinase-AKT can enhance E2F-1-mediated cell death. Moreover, an adenovirus expressing truncated E2F-1 keeps the ability of inducing apoptosis in melanoma cells. CONCLUSIONS ASK1 expression is upregulated by E2F-1 at the transcription level, but the upregulation of ASK1 expression by E2F-1 was not coordinated with an increased ASK1 activity. The ASK1-JNK/p38 pathway does not appear to play a crucial role in E2F-1-induced apoptosis.
Collapse
Affiliation(s)
- Yan Bin Dong
- Department of Surgery, University of Louisville School of Medicine, Louisville, Ky., USA
| | | | | | | | | | | | | | | | | |
Collapse
|
230
|
E2F-1 induces melanoma cell apoptosis via PUMA up-regulation and Bax translocation. BMC Cancer 2007; 7:24. [PMID: 17263886 PMCID: PMC1797184 DOI: 10.1186/1471-2407-7-24] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2006] [Accepted: 01/30/2007] [Indexed: 01/22/2023] Open
Abstract
Background PUMA is a pro-apoptotic Bcl-2 family member that has been shown to be involved in apoptosis in many cell types. We sought to ascertain whether induction of PUMA plays a crucial role in E2F-1-induced apoptosis in melanoma cells. Methods PUMA gene and protein expression levels were detected by real-time PCR and Western blot in SK-MEL-2 and HCT116 cell lines after Ad-E2F-1 infection. Activation of the PUMA promoter by E2F-1 overexpression was detected by dual luciferase reporter assay. E2F-1-induced Bax translocation was shown by immunocytochemistry. The induction of caspase-9 activity was measured by caspase-9 colorimetric assay kit. Results Up-regulation of the PUMA gene and protein by E2F-1 overexpression was detected by real-time PCR and Western blot analysis in the SK-MEL-2 melanoma cell line. In support of this finding, we found six putative E2F-1 binding sites within the PUMA promoter. Subsequent dual luciferase reporter assay showed that E2F-1 expression could increase the PUMA gene promoter activity 9.3 fold in SK-MEL-2 cells. The role of PUMA in E2F-1-induced apoptosis was further investigated in a PUMA knockout cell line. Cell viability assay showed that the HCT116 PUMA-/- cell line was more resistant to Ad-E2F-1-mediated cell death than the HCT116 PUMA+/+ cell line. Moreover, a 2.2-fold induction of the PUMA promoter was also noted in the HCT116 PUMA+/+ colon cancer cell line after Ad-E2F-1 infection. Overexpression of a truncated E2F-1 protein that lacks the transactivation domain failed to up-regulate PUMA promoter, suggesting that PUMA may be a transcriptional target of E2F-1. E2F-1-induced cancer cell apoptosis was accompanied by Bax translocation from the cytosol to mitochondria and the induction of caspase-9 activity, suggesting that E2F-1-induced apoptosis is mediated by PUMA through the cytochrome C/Apaf-1-dependent pathway. Conclusion Our studies strongly demonstrated that E2F-1 induces melanoma cell apoptosis via PUMA up-regulation and Bax translocation. The signaling pathways provided here will further enhance insights on the mechanisms of E2F-1-induced cancer cell apoptosis as a strategy for cancer therapy.
Collapse
|
231
|
Hess-Wilson JK, Daly HK, Zagorski WA, Montville CP, Knudsen KE. Mitogenic action of the androgen receptor sensitizes prostate cancer cells to taxane-based cytotoxic insult. Cancer Res 2007; 66:11998-2008. [PMID: 17178899 PMCID: PMC4443485 DOI: 10.1158/0008-5472.can-06-2249] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Prostate cancer cells are dependent on androgen for growth and survival; as such, inhibition of androgen receptor (AR) activity is the first line of intervention for disseminated disease. Recently, specific cytotoxic agents have been shown to extend survival times in patients with advanced disease. Given the established ability of androgen to modify cell survival in prostate cancer cells, it is imperative to determine the effect of the hormonal environment on cytotoxic response. Here, we show that the response of prostate cancer cells to taxane-induced cell death is significantly enhanced by androgen stimulation in AR-positive, androgen-dependent prostate cancer cells. Similar results were observed on androgen-independent AR activation. By contrast, AR-positive yet androgen-independent or AR-negative cells were refractory to androgen influence on taxane function. The ability of androgen to potentiate taxane activity was dependent on its mitogenic capacity and was separable from overall AR activity, as coadministration of AR antagonists, G(1) cyclin-dependent kinase inhibitors, or high-dose (growth inhibitory) androgen nullified the proapoptotic function of androgen. Observed induction of cell death was attributed to caspase-dependent apoptosis and correlated with p53 activation. Combined, these data indicate that the cytotoxic effects of taxanes are substantially influenced by the hormonal environment and/or status of AR activity in prostate cancer cells and provide the foundation for refinement and optimization of cytotoxic intervention in prostate cancer.
Collapse
Affiliation(s)
- Janet K. Hess-Wilson
- Department of Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Hannah K. Daly
- Department of Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - William A. Zagorski
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Christopher P. Montville
- Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Karen E. Knudsen
- Department of Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- University of Cincinnati Cancer Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
232
|
Abstract
Although mutations in the TP73 gene are extremely rare in human tumours, altered expression is common. In some tumours, most notably leukaemias and lymphomas, expression of TP73 is reduced, suggesting a tumour suppressor role. In contrast, TP73 is over-expressed in many other tumour types, implying that it has oncogenic functions in human tumourigenesis. These conflicting scenarios can be reconciled by the observations that the TP73 gene produces p53-like isoforms (TAp73) and anti-p53 isoforms (DeltaTAp73). Thus, loss of TAp73 or over-expression of DeltaTAp73 should each promote oncogenic transformation, and the balance of expression of the opposing isoforms is the crucial factor. The mechanisms that regulate expression of TP73 isoforms are therefore of great interest. Recent data provide evidence for interacting roles of ZEB1, p300, and a polymorphic 73 bp deletion in intron 1 of the human TP73 gene in this process. Importantly, alterations to the proposed regulatory pathway for controlling TP73 isoform expression in colorectal cancer are associated with adverse clinico-pathological characteristics. Because p73 is also associated with tumour chemosensitivity, these new findings should provide prognostic information and have the potential to guide future therapeutic decisions.
Collapse
Affiliation(s)
- P J Coates
- Pathology and Neurosciences, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK.
| |
Collapse
|
233
|
Jiang H, Hou C, Zhang S, Xie H, Zhou W, Jin Q, Cheng X, Qian R, Zhang X. Matrine upregulates the cell cycle protein E2F-1 and triggers apoptosis via the mitochondrial pathway in K562 cells. Eur J Pharmacol 2007; 559:98-108. [PMID: 17291488 DOI: 10.1016/j.ejphar.2006.12.017] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 12/11/2006] [Accepted: 12/14/2006] [Indexed: 12/28/2022]
Abstract
Matrine is a major component of Sophora Flavescens and has been reported to stimulate differentiation of erythroleukemia cells. Here we show that matrine inhibits cell proliferation or induces apoptosis in a cell type-specific manner. The latter effect was investigated in more detail in the p53 deficient erythroleukemia cell line, K562. Matrine exposure induced apoptosis in a time- and dose-dependent manner in these cells. Interestingly, co-treatment with etoposide potentiated apoptosis. Further analysis of matrine-induced apoptotic changes revealed that E2F-1 and Apaf-1 were upregulated, whereas Rb was downregulated after 24 h of exposure. This was followed by Bax translocation, cytochrome c release, and caspase-9 and -3 activation. These results demonstrate that matrine triggers apoptosis of K562 cells primarily through the mitochondrial pathway and that matrine is a potential anti-tumor drug.
Collapse
Affiliation(s)
- Hua Jiang
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 YueYang Road, Shanghai 200031, China
| | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Clément V, Dunand-Sauthier I, Wiznerowicz M, Clarkson SG. UV-induced apoptosis in XPG-deficient fibroblasts involves activation of CD95 and caspases but not p53. DNA Repair (Amst) 2007; 6:602-14. [PMID: 17208056 DOI: 10.1016/j.dnarep.2006.11.010] [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: 08/22/2006] [Revised: 11/28/2006] [Accepted: 11/29/2006] [Indexed: 12/22/2022]
Abstract
Mildly affected individuals from xeroderma pigmentosum complementation group G (XP-G) possess single amino acid substitutions in the XPG protein that adversely affects its 3' endonuclease function in nucleotide excision repair. More serious mutations in the XPG gene generate truncated or unstable XPG proteins and result in a particularly early and severe form of the combined XP/CS complex. Following UV irradiation, cells from such XP-G/CS patients enter apoptosis more readily than other DNA repair-deficient cells. Here, we explore the mechanisms by which UV triggers the apoptotic cell death program in XP-G and XP-G/CS primary fibroblasts. Activation of the CD95 signalling pathway occurs within minutes and it is the earliest detectable post-UV event in such cells. This is rapidly followed by activation of caspase-8 then caspase-3. Several hours later caspase-9 becomes activated and the mitochondrial membrane potential drops, but without any obvious prior release of cytochrome c. Although p53 accumulates in XPG-deficient cells after UV irradiation, use of RNA interference demonstrates that p53 is not required for their UV-induced apoptotic response. p53 ablation of wild-type fibroblasts reduces MDM2 mRNA levels, inhibits accumulation of the 90kDa/92kDa Mdm2 isoforms, and prevents the nuclear relocalisation of Mdm2 after UV treatment. The same post-UV effects occur in XPG-deficient cells that express normal p53 levels. These results emphasise the importance of the extrinsic apoptotic pathway and aberrant Mdm2 events for the severe UV-induced apoptosis of XPG-deficient primary fibroblasts. XP-G/CS cells constitutively overexpress the pro-apoptotic Bax protein and a long isoform of the E2F1 transcription factor that controls S phase entry, which may prime them to enter apoptosis very readily after UV irradiation.
Collapse
Affiliation(s)
- Virginie Clément
- Department of Microbiology and Molecular Medicine, University Medical Centre (CMU), 1 rue Michel-Servet, 1211 Geneva 4, Switzerland.
| | | | | | | |
Collapse
|
235
|
Li Z, Kreutzer M, Mikkat S, Mise N, Glocker MO, Pützer BM. Proteomic analysis of the E2F1 response in p53-negative cancer cells: new aspects in the regulation of cell survival and death. Proteomics 2007; 6:5735-45. [PMID: 17001603 DOI: 10.1002/pmic.200600290] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
E2F1 is an essential transcription factor that regulates cell-cycle progression and apoptosis. Overexpression of E2F1 sensitizes neoplastic cells to apoptosis and leads to tumor growth suppression, making it an interesting target for anticancer therapy. Use of E2F1 as a therapeutic, however, requires a detailed knowledge of the mechanisms by which it controls cellular proliferation and apoptosis, and of other potential E2F1 activities. In this study, a differential proteome analysis was performed to identify proteins associated with E2F1 activity in inducible p53-deficient Saos-2ERE2F1 osteosarcoma cells. 2-DE revealed a distinct protein profile at 32 h after E2F1 activation. Thirty-three proteins were reproducibly identified as either up-regulated or down-regulated. Proteins were identified by MALDI-MS. They included hitherto unknown E2F1 target proteins of cytoskeletal origin, chaperones, enzymes, proteasomal proteins, and several heterogeneous nuclear ribonucleoproteins, suggesting its role in the ER-stress response, protein degradation, and modulation of pre-mRNA splicing. Protein analysis-derived results were verified by Western blot using representative protein candidates. Thirteen identified proteins were the products of genes known to be cancer related. Thus, proteome analysis provides new information about the complexity of E2F1 activities in human cancer cells that may be considered when using E2F1 as a drug.
Collapse
Affiliation(s)
- Zhenpeng Li
- Department of Vectorology & Experimental Gene Therapy, University of Rostock, Germany
| | | | | | | | | | | |
Collapse
|
236
|
Novel Approaches for Chemosensitization of Breast Cancer Cells: The E1A Story. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 608:144-69. [PMID: 17993239 DOI: 10.1007/978-0-387-74039-3_11] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
237
|
Morris EJ, Michaud WA, Ji JY, Moon NS, Rocco JW, Dyson NJ. Functional identification of Api5 as a suppressor of E2F-dependent apoptosis in vivo. PLoS Genet 2006; 2:e196. [PMID: 17112319 PMCID: PMC1636698 DOI: 10.1371/journal.pgen.0020196] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Accepted: 10/03/2006] [Indexed: 11/24/2022] Open
Abstract
Retinoblastoma protein and E2-promoter binding factor (E2F) family members are important regulators of G1-S phase progression. Deregulated E2F also sensitizes cells to apoptosis, but this aspect of E2F function is poorly understood. Studies of E2F-induced apoptosis have mostly been carried out in tissue culture cells, and the analysis of the factors that are important for this process has been restricted to the testing of a few candidate genes. Using Drosophila as a model system, we have generated tools that allow genetic modifiers of E2F-dependent apoptosis to be identified in vivo and developed assays that allow effects on E2F-induced apoptosis to be studied in cultured cells. Genetic interactions show that dE2F1-dependent apoptosis in vivo involves dArk/Apaf1 apoptosome-dependent activation of both initiator and effector caspases and is sensitive to levels of Drosophila inhibitor of apoptosis-1 (dIAP1). Using these approaches, we report the surprising finding that apoptosis inhibitor-5/antiapoptosis clone-11 (Api5/Aac11) is a critical determinant of dE2F1-induced apoptosis in vivo and in vitro. This functional interaction occurs in multiple tissues, is specific to E2F-induced apoptosis, and is conserved from flies to humans. Interestingly, Api5/Aac11 acts downstream of E2F and suppresses E2F-dependent apoptosis without generally blocking E2F-dependent transcription. Api5/Aac11 expression is often upregulated in tumor cells, particularly in metastatic cells. We find that depletion of Api5 is tumor cell lethal. The strong genetic interaction between E2F and Api5/Aac11 suggests that elevated levels of Api5 may be selected during tumorigenesis to allow cells with deregulated E2F activity to survive under suboptimal conditions. Therefore, inhibition of Api5 function might offer a possible mechanism for antitumor exploitation. The retinoblastoma protein (pRB) was the first human tumor suppressor to be described, and it works by limiting the activity of the E2F transcription factor. The pRB pathway is inactivated in most forms of cancer, and, accordingly, most tumor cells have deregulated E2F. Uncontrolled E2F drives cell proliferation, but it also sensitizes cells to die (apoptosis). E2F-induced apoptosis is not well understood, but it affects the development of cancer and, potentially, could be exploited for cancer treatment. To date, however, there have been very few studies of E2F-induced apoptosis in animal models. The authors describe a series of genetic tools that allow systematic studies of E2F-induced apoptosis in Drosophila. As validation, this approach identified some known regulators of E2F-dependent apoptosis and also identified Api5, a little-studied gene that had not previously been linked to E2F, as a potent suppressor of E2F-induced cell death. The effects of Api5 on E2F occur in several different tissues and are conserved from flies to humans. This last point is significant since Api5 is upregulated in cancer cells. The discovery of the E2F–Api5 interaction demonstrates that important modulators of E2F-induced apoptosis are waiting to be discovered and that they can be found using Drosophila.
Collapse
Affiliation(s)
- Erick J Morris
- Massachusetts General Hospital Cancer Center, Laboratory of Molecular Oncology, Charlestown, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - William A Michaud
- Massachusetts General Hospital Cancer Center, Laboratory of Molecular Oncology, Charlestown, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Jun-Yuan Ji
- Massachusetts General Hospital Cancer Center, Laboratory of Molecular Oncology, Charlestown, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nam-Sung Moon
- Massachusetts General Hospital Cancer Center, Laboratory of Molecular Oncology, Charlestown, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - James W Rocco
- Massachusetts General Hospital Cancer Center, Laboratory of Molecular Oncology, Charlestown, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Nicholas J Dyson
- Massachusetts General Hospital Cancer Center, Laboratory of Molecular Oncology, Charlestown, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
238
|
Ambrosini G, Sambol EB, Carvajal D, Vassilev LT, Singer S, Schwartz GK. Mouse double minute antagonist Nutlin-3a enhances chemotherapy-induced apoptosis in cancer cells with mutant p53 by activating E2F1. Oncogene 2006; 26:3473-81. [PMID: 17146434 DOI: 10.1038/sj.onc.1210136] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
MDM2 is a critical negative regulator of the p53 tumor suppressor protein. Recently, small-molecule antagonists of MDM2, the Nutlins, have been developed to inhibit the p53-MDM2 interaction and activate p53 signaling. However, half of human cancers have mutated p53 and they are resistant to Nutlin treatment. Here, we report that treatment of the p53-mutant malignant peripheral nerve sheath (MPNST) and p53-null HCT116 cells with cisplatin (Cis) and Nutlin-3a induced a degree of apoptosis that was significantly greater than either drug alone. Nutlin-3a also increased the cytotoxicity of both carboplatin and doxorubicin in a series of p53-mutant human tumor cell lines. In the human dedifferentiated liposarcoma cell line (LS141) and the p53 wild-type HCT116 cells, Nutlin-3a induced downregulation of E2F1 and this effect appeared to be proteasome dependent. In contrast, in MPNST and HCTp53-/- cells, Nutlin-3a inhibited the binding of E2F1 to MDM2 and induced transcriptional activation of free E2F1 in the presence of Cis-induced DNA damage. Downregulation of E2F1 by small interfering RNA significantly decreased the level of apoptosis induced by Cis and Nutlin-3a treatment. Moreover, expression of a dominant-negative form of E2F1 rescued cells from apoptosis, whereas cells overexpressing wild-type E2F1 showed an increase in cell death. This correlated with the induction of the proapoptotic proteins p73alpha and Noxa, which are both regulated by E2F1. These results indicate that antagonism of MDM2 by Nutlin-3a in cells with mutant p53 enhances chemosensitivity in an E2F1-dependent manner. Nutlin-3a therefore may provide a therapeutic benefit in tumors with mutant p53 provided it is combined with chemotherapy.
Collapse
Affiliation(s)
- G Ambrosini
- Laboratory of New Drug Development, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
239
|
Domínguez G, Peña C, Silva J, García JM, García V, Rodríguez R, Cantos B, Citores MJ, España P, Bonilla F. The presence of an intronic deletion in p73 and high levels of ZEB1 alter the TAp73/DeltaTAp73 ratio in colorectal carcinomas. J Pathol 2006; 210:390-397. [PMID: 17029218 DOI: 10.1002/path.2066] [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: 01/27/2006] [Accepted: 08/16/2006] [Indexed: 02/05/2023]
Abstract
TAp73 variants largely mimic p53 suppressor activities, while DeltaTAp73 forms act as oncogenes through the inactivation of p53 and TAp73. The present study analysed how TAp73 and DeltaTAp73 levels might be affected by the presence of a 73 bp deletion in a regulatory region of p73. The clinical relevance of this deletion was also examined. ZEB1 can bind to the region repressing p73 transcription in vitro. The relationship between ZEB1 and p73 variant expression levels was studied in the context of this deletion and the levels of the ZEB1 cofactors p300 and CtBP. Tumour and normal tissue from 81 colorectal cancer patients was analysed to evaluate firstly the levels of TAp73, DeltaTAp73 (DeltaEx2p73, DeltaEx2/3p73, and DeltaNp73), ZEB1, p300, and CtBP by quantitative real-time RT-PCR, and secondly the presence of the 73 bp deletion. Tumour characteristics were examined in each patient. Suppressor and oncogenic isoforms of p73 were co-up-regulated in tumour tissues. Overexpression of p73 variants was associated with adverse tumour features. The 73 bp deletion was present in 40% of the patients and was associated with adverse pathological parameters of the tumours and also with TAp73 down-regulation. In those cases harbouring the deletion, the levels of ZEB1 and those of DeltaEx2p73, DeltaEx2/3p73, and DeltaNp73 correlated directly. Variations in the concentration of p300 affected the observed correlations between ZEB1 and the different p73 variants. In conclusion, in colorectal cancer, the 73 bp deletion in the first intron of the p73 gene and different expression levels of ZEB1 and p300 may act in concert to affect the ratio of TAp73/DeltaTAp73 forms, favouring p73 oncogenic variants. In addition, up-regulation of p73 oncogenic isoforms predicts a poor prognosis based on its relationship with advanced tumour stage.
Collapse
Affiliation(s)
- G Domínguez
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Cam H, Griesmann H, Beitzinger M, Hofmann L, Beinoraviciute-Kellner R, Sauer M, Hüttinger-Kirchhof N, Oswald C, Friedl P, Gattenlöhner S, Burek C, Rosenwald A, Stiewe T. p53 family members in myogenic differentiation and rhabdomyosarcoma development. Cancer Cell 2006; 10:281-93. [PMID: 17045206 DOI: 10.1016/j.ccr.2006.08.024] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Revised: 06/05/2006] [Accepted: 08/10/2006] [Indexed: 12/20/2022]
Abstract
The p53 family comprises the tumor suppressor p53 and the structural homologs p63 and p73. How the three family members cooperate in tumor suppression remains unclear. Here, we report different but complementary functions of the individual members for regulating retinoblastoma protein (RB) function during myogenic differentiation. Whereas p53 transactivates the retinoblastoma gene, p63 and p73 induce the cyclin-dependent kinase inhibitor p57 to maintain RB in an active, hypophosphorylated state. DeltaNp73 inhibits these functions of the p53 family in differentiation control, prevents myogenic differentiation, and enables cooperating oncogenes to transform myoblasts to tumorigenicity. DeltaNp73 is frequently overexpressed in rhabdomyosarcoma and essential for tumor progression in vivo. These findings establish differentiation control as a key tumor suppressor activity of the p53 family.
Collapse
Affiliation(s)
- Hakan Cam
- Molecular Tumor Biology Group, Rudolf-Virchow-Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, 97078 Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Russo AJ, Magro PG, Hu Z, Li WW, Peters R, Mandola J, Banerjee D, Bertino JR. E2F-1 overexpression in U2OS cells increases cyclin B1 levels and cdc2 kinase activity and sensitizes cells to antimitotic agents. Cancer Res 2006; 66:7253-60. [PMID: 16849574 DOI: 10.1158/0008-5472.can-05-3725] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The E2F transcription factors play a critical role in coordinating transcription of specific genes essential for G1-S transition. In early G1, the retinoblastoma protein (pRB) becomes phosphorylated by cyclin-dependent kinases, disrupting pRB binding to E2F-1-3, allowing "free" E2F to regulate genes involved in proliferation. In the present study, we used a tetracycline E2F-1 inducible U2OS osteosarcoma cell line to investigate the effect of increasing levels of E2F-1 on the cytotoxicity of various chemotherapeutic drugs. Upon overexpression of E2F-1, there was no detectable change in cytotoxicity to doxorubicin, cisplatin, 5-fluorouracil, or etoposide. In contrast, overexpression of E2F-1 resulted in a marked increase in sensitivity to vinblastine and paclitaxel, drugs that are known to be more effective against cells in M phase. Therefore, we investigated the effect of E2F-1 overexpression on proteins regulating the G2-M transition and M phase, in particular cyclin B1 and cdc2 kinase. Cyclin B1 mRNA and protein levels increased within 24 hours of E2F1 induction together with an increase in associated cdc2 kinase activity. Overexpression of cyclin B1 also resulted in a specific increase in sensitivity to paclitaxel and an increase in the cellular growth rate. Knockdown of cyclin B1 using an RNA interference oligo resulted in a slower cellular growth rate and an increase in resistance to paclitaxel. These studies add support to recent reports that show E2F regulates genes involved in mitotic entry and exit and allow the suggestion that mitotic inhibitors may have selective effects in tumors that overexpress E2F-1.
Collapse
Affiliation(s)
- Angelo J Russo
- The Cancer Institute of New Jersey, Robert Wood Johnson School of Medicine, University of Medicine and Dentistry of New Jersey, New Brunswick, New Jersey 08903, USA
| | | | | | | | | | | | | | | |
Collapse
|
242
|
García-Alvarez G, Ventura V, Ros O, Aligué R, Gil J, Tauler A. Glycogen synthase kinase-3beta binds to E2F1 and regulates its transcriptional activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1773:375-82. [PMID: 17050006 DOI: 10.1016/j.bbamcr.2006.09.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Revised: 09/04/2006] [Accepted: 09/05/2006] [Indexed: 01/02/2023]
Abstract
GSK3beta and E2F1 play an important role in the control of proliferation and apoptosis. Previous work has demonstrated that GSK3beta indirectly regulates E2F activity through modulation of cyclin D1 levels. In this work we show that GSK3beta phosphorylates human E2F1 in vitro at serine 403 and threonine 433, both residues localized at its transactivation domain. This phosphorylation was not detected in vivo. However, co-immunoprecipitation experiments do reveal in vivo binding of these proteins. Moreover, uninhibitable and catalitycally inactive GSK3beta forms inhibit the transcriptional activity of a fusion protein containing E2F1 transactivation domain. Both forms of GSK3beta inhibit E2F1 with similar efficiency. Interestingly the effect was independent of the mutation of serine 403 and threonine 433 to alanine. This suggests that this transcriptional modulation is independent of GSK3beta kinase activity and phosphorylation state of serine 403 and threonine 433. The re-targeting of these GSK3beta forms to the nucleus results in a higher capacity to regulate E2F1 transcriptional activity. Depletion of the levels of GSK3beta protein using siRNA activates E2F1 transcriptional activity. The data presented in this study offer a new mechanism of regulation of E2F1 by direct binding of GSK3beta to its transactivation domain.
Collapse
Affiliation(s)
- Gisela García-Alvarez
- Departament de Bioquímica i Biologia Molecular, Facultat de Farmàcia, Universitat de Barcelona. Av. Diagonal 643, E-08028 Barcelona, Catalunya, Spain
| | | | | | | | | | | |
Collapse
|
243
|
Pluta A, Nyman U, Joseph B, Robak T, Zhivotovsky B, Smolewski P. The role of p73 in hematological malignancies. Leukemia 2006; 20:757-66. [PMID: 16541141 DOI: 10.1038/sj.leu.2404166] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The P73 gene is a homologue of the P53 tumor suppressor. Owing to its structural similarity with p53, p73 was originally considered to have tumor suppressor function. However, the discovery of N-terminal truncated isoforms with oncogenic properties showed a 'two in one' structure of its product, p73 protein. The full-length variants are strong inducers of apoptosis, whereas the truncated isoforms inhibit proapoptotic activity of p53 and the full-length p73. Thus, p73 is involved in the regulation of cell cycle, cell death and development. Moreover, it plays a role in carcinogenesis and controls tumor sensitivity to treatment. p73 is commonly expressed in tumor cells in hematological malignancies. Overexpression of p73 protein and aberrant expression of its particular isoforms, with very low frequency of P73 hypermethylation or mutations, were found in malignant myeloproliferations, including acute myeloblastic leukemia. In contrast, hypermethylation and subsequent inactivation of the P73 gene are the most common findings in malignant lymphoproliferative disorders, especially acute lymphoblastic leukemia (ALL) and non-Hodgkin's lymphomas. Assessment of P73 methylation may provide important prognostic information, as was confirmed in patients with ALL. This review summarizes some aspects of p73 biology with particular reference to its possible pathogenetic role and prognostic significance in hematological malignancies.
Collapse
Affiliation(s)
- A Pluta
- Department of Hematology, Medical University of Lodz and Copernicus Memorial Hospital, Lodz, Poland
| | | | | | | | | | | |
Collapse
|
244
|
Wichmann A, Jaklevic B, Su TT. Ionizing radiation induces caspase-dependent but Chk2- and p53-independent cell death in Drosophila melanogaster. Proc Natl Acad Sci U S A 2006; 103:9952-7. [PMID: 16785441 PMCID: PMC1502560 DOI: 10.1073/pnas.0510528103] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ionizing radiation (IR) can induce apoptosis via p53, which is the most commonly mutated gene in human cancers. Loss of p53, however, can render cancer cells refractory to therapeutic effects of IR. Alternate p53-independent pathways exist but are not as well understood as p53-dependent apoptosis. Studies of how IR induces p53-independent cell death could benefit from the existence of a genetically tractable model. In Drosophila melanogaster, IR induces apoptosis in the imaginal discs of larvae, typically assayed at 4-6 hr after exposure to a LD(50) dose. In mutants of Drosophila Chk2 or p53 homologs, apoptosis is severely diminished in these assays, leading to the widely held belief that IR-induced apoptosis depends on these genes in Drosophila. In this article, we show that IR-induced apoptosis still occurs in the imaginal discs of chk2 and p53 mutant larvae, albeit with a delay. We demonstrate that this phenomenon is a true apoptotic response because it requires caspase activity and the chromosomal locus that encodes the pro-apoptotic genes reaper, hid, and grim. We also show that Chk2- and p53-independent apoptosis is IR dose-dependent and is therefore probably triggered by a DNA damage signal. We conclude that Drosophila has Chk2- and p53-independent pathways to activate caspases and induce apoptosis in response to IR. This work establishes Drosophila as a model for p53-independent apoptosis, which is of potential therapeutic importance for inducing cell death in p53-deficient cancer cells.
Collapse
Affiliation(s)
- Anita Wichmann
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309-0347
| | - Burnley Jaklevic
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309-0347
| | - Tin Tin Su
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309-0347
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
245
|
Belloni L, Moretti F, Merlo P, Damalas A, Costanzo A, Blandino G, Levrero M. DNp73alpha protects myogenic cells from apoptosis. Oncogene 2006; 25:3606-12. [PMID: 16652159 DOI: 10.1038/sj.onc.1209321] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2005] [Revised: 10/24/2005] [Accepted: 10/24/2005] [Indexed: 11/09/2022]
Abstract
The P73 gene is transcribed from two promoters, P1 and P2, that direct the expression of multiple transactivation competent (TA) and dominant negative (DN) isoforms. TAp73 transcription factors mediate cell cycle arrest and/or apoptosis in response to DNA damage and are involved in developmental processes. P73 mRNA levels increase and the P1p73 promoter is upregulated during myogenic differentiation of C2C12 skeletal muscle satellite cells. The DNp73 proteins act as trans-repressors of p53- and p73-dependent transcription, and possess both antiapoptotic and pro-proliferative potential. Here, we show that DNp73alpha is expressed in proliferating C2C12 myoblasts, rapidly accumulates in differentiating myocytes and remains elevated in C2C12 myotubes. By combining transactivation assays and chromatin immunoprecipitation analysis, we could show that the upregulation of the P2p73 promoter during myogenic differentiation is mediated by the coordinated recruitment and activity of MyoD and p53/p73. Abrogation of DNp73 expression by specific siRNA led to a strong potentiation of the spontaneous apoptosis of C2C12 myoblasts induced to differentiate. Finally, unlike TAp73 that contributes to DNA damage-induced apoptosis of myotubes, endogenous DNp73 mediates the relative resistance of differentiated myotubes to DNA damage. Altogether, our findings identify DNp73alpha as an important target in designing strategies aimed at the potentiation of the regenerative potential of skeletal satellite cells.
Collapse
Affiliation(s)
- L Belloni
- Fondazione A Cesalpino and Department of Internal Medicine, University of Rome La Sapienza, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
246
|
Joy J, Nalabothula N, Ghosh M, Popp O, Jochum M, Machleidt W, Gil-Parrado S, Holak TA. Identification of calpain cleavage sites in the G1 cyclin-dependent kinase inhibitor p19(INK4d). Biol Chem 2006; 387:329-35. [PMID: 16542156 DOI: 10.1515/bc.2006.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Calpains are a large family of Ca2+-dependent cysteine proteases that are ubiquitously distributed across most cell types and vertebrate species. Calpains play a role in cell differentiation, apoptosis, cytoskeletal remodeling, signal transduction and the cell cycle. The cell cycle proteins cyclin D1 and p21(KIP1), for example, have been shown to be affected by calpains. However, the rules that govern calpain cleavage specificity are poorly understood. We report here studies on the pattern of mu-calpain proteolysis of the p19(INK4d) protein, a cyclin-dependent kinase 4/6 inhibitor that negatively regulates the mammalian cell cycle. Our data show new characteristics of calpain action: mu-calpain cleaves p19(INK4d) immediately after the first and second ankyrin repeats that are structurally less stable compared to the other repeats. This is in contrast to features observed so far in the specificity of calpains for their substrates. These results imply that calpain may be involved in the cell cycle by regulating the cell cycle regulatory protein turnover through CDK inhibitors and cyclins.
Collapse
Affiliation(s)
- Joma Joy
- Max-Planck-Institut für Biochemie, D-82152 Martinsried, Germany
| | | | | | | | | | | | | | | |
Collapse
|
247
|
Harbour JW. Eye cancer: unique insights into oncogenesis: the Cogan Lecture. Invest Ophthalmol Vis Sci 2006; 47:1736-45. [PMID: 16638975 PMCID: PMC1769553 DOI: 10.1167/iovs.05-1291] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- J William Harbour
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| |
Collapse
|
248
|
Yamasaki L. Modeling cell cycle control and cancer with pRB tumor suppressor. Results Probl Cell Differ 2006; 42:227-56. [PMID: 16903213 DOI: 10.1007/b136682] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cancer is a complex syndrome of diseases characterized by the increased abundance of cells that disrupts the normal tissue architecture within an organism. Defining one universal mechanism underlying cancer with the hope of designing a magic bullet against cancer is impossible, largely because there is so much variation between various types of cancer and different individuals. However, we have learned much in past decades about different journeys that a normal cell takes to become cancerous, and that the delicate balance between oncogenes and tumor suppressor is upset, favoring growth and survival of the tumor cell. One of the most important cellular barriers to cancer development is the retinoblastoma tumor suppressor (pRB) pathway, which is inactivated in a wide range of human tumors and controls cell cycle progression via repression of the E2F/DP transcription factor family. Much of the clarity with which we view tumor suppression via pRB is due to our belief in the universality of the cell cycle and our attempts to model tumor pathways in vivo, nowhere so evident as in the multitude of data emerging from mutant mouse models that have been engineered to understand how cell cycle regulators limit growth in vivo and how deregulation of these regulators facilitates cancer development. In spite of this clarity, we have witnessed with incredulity several stunning results in the last 2 years that have challenged the very foundations of the cell cycle paradigm and made us question seriously how important these cell cycle regulators actually are.
Collapse
Affiliation(s)
- Lili Yamasaki
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| |
Collapse
|
249
|
Shapiro GS, Van Peursem C, Ornelles DA, Schaack J, DeGregori J. Recombinant adenoviral vectors can induce expression of p73 via the E4-orf6/7 protein. J Virol 2006; 80:5349-60. [PMID: 16699015 PMCID: PMC1472169 DOI: 10.1128/jvi.02016-05] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Accepted: 03/06/2006] [Indexed: 11/20/2022] Open
Abstract
Despite the utility of recombinant adenoviral vectors in basic research, their therapeutic promise remains unfulfilled. Most engineered adenoviral vectors use a heterologous promoter to transcribe a foreign gene. We show that adenoviruses containing the cytomegalovirus immediate-early promoter induce the expression of the proapoptotic cellular protein TAp73 via the cyclin-dependent kinase-retinoblastoma protein-E2F pathway in murine embryonic fibroblasts. Cells transduced with these vectors also expressed high levels of the adenoviral E4-orf6/7 and E2A proteins. By contrast, adenoviruses containing the ubiquitin C promoter failed to elicit these effects. E4-orf6/7 is necessary and sufficient for increased TAp73 expression, as shown by using retrovirus-mediated E4-orf6/7 expression and adenovirus with the E4-orf6/7 gene deleted. Activation of TAp73 likely occurs via E4-orf6/7-induced dimerization of E2F and subsequent binding to the inverted E2F-responsive elements within the TAp73 promoter. In addition, adenoviral vectors containing the cytomegalovirus immediate-early promoter, but not the ubiquitin C promoter, cooperated with chemotherapeutic agents to decrease cellularity in vitro. In contrast to murine embryonic fibroblasts, adenoviruses containing the ubiquitin C promoter, but not the cytomegalovirus immediate-early promoter, induced both E4-orf6/7 and TAp73 in human foreskin fibroblasts, emphasizing the importance of cellular context for promoter-dependent effects. Because TAp73 is important for the efficacy of chemotherapy, adenoviruses that increase TAp73 expression may enhance cancer therapies by promoting apoptosis. However, such adenoviruses may impair the long-term survival of transduced cells during gene replacement therapies. Our findings reveal previously unknown effects of foreign promoters in recombinant adenoviral vectors and suggest means to improve the utility of engineered adenoviruses by better controlling their impact on viral and cellular gene expression.
Collapse
Affiliation(s)
- Gary S Shapiro
- Department of Biochemistry and Molecular Genetics, Mail Stop 8101, Aurora, CO 80045, USA
| | | | | | | | | |
Collapse
|
250
|
Bell LA, O'Prey J, Ryan KM. DNA-binding independent cell death from a minimal proapoptotic region of E2F-1. Oncogene 2006; 25:5656-63. [PMID: 16652153 DOI: 10.1038/sj.onc.1209580] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The ability to induce cell cycle progression while evading cell death is a defining characteristic of cancer. Deregulation of E2F is a common event in most human cancers. Paradoxically, this can lead to both cell cycle progression and apoptosis. Although the way in which E2F causes cell cycle progression is well characterized, the pathways by which E2F induces cell death are less well defined. Many of the known mechanisms through which E2F induces apoptosis occur through regulation of E2F target genes. However, mutants of E2F-1 that lack the transactivation domain are still able to induce cell death. To further investigate this activity, we refined a transactivation independent mutant to identify a minimal apoptotic domain. This revealed that only 75 amino acids from within the DNA-binding domain of E2F-1 is sufficient for cell death and that this activity is also present in the DNA-binding domains of E2F-2 and E2F-3. However, analysis of this domain from E2F-1 revealed it does not bind DNA and is consequently unable to transactivate, repress or de-repress E2F target genes. This provocative observation therefore defines a potential new mechanism of death from E2F and opens up new opportunities for inducing cell death in tumours for therapeutic gain.
Collapse
Affiliation(s)
- L A Bell
- Tumour Cell Death Laboratory, Beatson Institute for Cancer Research, Cancer Research UK Beatson Laboratories, Garscube Estate, Glasgow, UK
| | | | | |
Collapse
|