151
|
Patenaude A, Fortin JS, Deschenes R, Côté MF, Lacroix J, C.-Gaudreault R, Petitclerc É. Chloroethyl urea derivatives block tumour growth and thioredoxin-1 nuclear translocation. Can J Physiol Pharmacol 2010; 88:1102-14. [DOI: 10.1139/y10-084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aryl chloroethyl ureas (CEUs) are new protein alkylating agents exhibiting anticancer activity both in vitro and in vivo. We report herein that 14C-labeled CEU derivatives, designated CEU-025 and CEU-027, covalently bind to thioredoxin-1 (TRX1). Covalent binding of these molecules slightly decreases the disulfide-reducing activity of recombinant TRX1, when compared with the effect of strong thioalkylating agents such as N-ethylmaleimide. Moreover, site-directed mutagenesis and diamide competition assays demonstrated that TRX1 cysteinyl residues are not the prime targets of CEUs. CEU-025 abrogates the nuclear translocation of TRX1 in human cancer cells. In addition, we show that CEU-025 can block TRX1 nuclear translocation induced by cisplatin. Unexpectedly, pretreatment with sublethal CEU-025 concentrations that block TRX1 nuclear translocation protected the cells against cisplatin cytotoxicity. Overexpression of TRX1 in HT1080 fibrosarcoma cells attenuated CEU-025 cytotoxicity, while its suppression using TRX1-specific siRNA increased the effects of CEU-025, suggesting that loss of function of TRX1 is involved, at least in part, in the cytotoxic activity of CEU-025. These results suggest that CEU-025 and CEU-027 exhibit anticancer activity through a novel, unique mechanism of action. The importance of TRX1 and the dependence of the cytotoxicity of CEU-025 and CEU-027 on TRX1 intracellular localization are also discussed.
Collapse
Affiliation(s)
- Alexandre Patenaude
- Unité de Biotechnologie et de Bioingénierie, CHUQ, Hôpital Saint-François d’Assise, Université Laval, Québec, QC, Canada
- Héma-Québec, Ingénierie cellulaire, Québec, QC, Canada
| | - Jessica S. Fortin
- Unité de Biotechnologie et de Bioingénierie, CHUQ, Hôpital Saint-François d’Assise, Université Laval, Québec, QC, Canada
- Héma-Québec, Ingénierie cellulaire, Québec, QC, Canada
| | - Réna Deschenes
- Unité de Biotechnologie et de Bioingénierie, CHUQ, Hôpital Saint-François d’Assise, Université Laval, Québec, QC, Canada
- Héma-Québec, Ingénierie cellulaire, Québec, QC, Canada
| | - Marie-France Côté
- Unité de Biotechnologie et de Bioingénierie, CHUQ, Hôpital Saint-François d’Assise, Université Laval, Québec, QC, Canada
- Héma-Québec, Ingénierie cellulaire, Québec, QC, Canada
| | - Jacques Lacroix
- Unité de Biotechnologie et de Bioingénierie, CHUQ, Hôpital Saint-François d’Assise, Université Laval, Québec, QC, Canada
- Héma-Québec, Ingénierie cellulaire, Québec, QC, Canada
| | - René C.-Gaudreault
- Unité de Biotechnologie et de Bioingénierie, CHUQ, Hôpital Saint-François d’Assise, Université Laval, Québec, QC, Canada
- Héma-Québec, Ingénierie cellulaire, Québec, QC, Canada
| | - Éric Petitclerc
- Unité de Biotechnologie et de Bioingénierie, CHUQ, Hôpital Saint-François d’Assise, Université Laval, Québec, QC, Canada
- Héma-Québec, Ingénierie cellulaire, Québec, QC, Canada
| |
Collapse
|
152
|
Schlereth K, Charles JP, Bretz AC, Stiewe T. Life or death: p53-induced apoptosis requires DNA binding cooperativity. Cell Cycle 2010; 9:4068-76. [PMID: 20948308 DOI: 10.4161/cc.9.20.13595] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The tumor suppressor p53 provides exquisite protection from cancer by balancing cell survival and death in response to stress. Sustained stress or irreparable damage trigger p53's killer functions to permanently eliminate genetically-altered cells as a potential source of cancer. To prevent the unnecessary loss of cells that could cause premature aging as a result of stem cell attrition, the killer functions of p53 are tightly regulated and balanced against protector functions that promote damage repair and support survival in response to low stress or mild damage. In molecular terms these p53-based cell fate decisions involve protein interactions with cofactors and modifying enzymes, which modulate the activation of distinct sets of p53 target genes. In addition, we demonstrate that part of this regulation occurs at the level of DNA binding. We show that the killer function of p53 requires the four DNA binding domains within the p53 tetramer to interact with one another. These intermolecular interactions enable cooperative binding of p53 to less perfect response elements in the genome, which are present in many target genes essential for apoptosis. Modulating p53 interactions within the tetramer could therefore present a novel promising strategy to fine-tune p53-based cell fate decisions.
Collapse
|
153
|
Abstract
The majority of human cancers acquire mutations that abrogate the p53 tumor suppressor network and, as a consequence, p53 is one of the most extensively studied proteins in cancer research. Because of its potent tumor suppressive activity, it is widely assumed that a molecular understanding of p53 action will produce fundamental insights into natural processes that limit tumorigenesis and may identify key molecular targets for therapeutic intervention. p53 functions largely as a transcription factor, and can trigger a variety of antiproliferative programs by activating or repressing key effector genes. Despite a significant body of literature detailing the biochemical and biological functions of p53, much remains to be elucidated. Indeed, the p53 network is as complex and enigmatic as it is relevant. It is the goal of this article, written 30 years after the discovery of p53, to present a concise review of the tumor suppressor role of the p53 network and to highlight the context-dependent nature of p53 target-gene functions.
Collapse
|
154
|
Asiri YA. Probucol attenuates cyclophosphamide-induced oxidative apoptosis, p53 and Bax signal expression in rat cardiac tissues. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2010; 3:308-16. [PMID: 21150336 PMCID: PMC3154034 DOI: 10.4161/oxim.3.5.13107] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2010] [Revised: 07/20/2010] [Accepted: 07/21/2010] [Indexed: 11/19/2022]
Abstract
Cyclophosphamide (CP) is a widely used in cancer chemotherapy and immunosuppression, which could cause toxicity of the normal cells due to its toxic metabolites. Probucol, cholesterol-lowering drug, acts as potential inhibitor of DNA damage and shows to protect against doxorubicin-induced cardiomyopathy by enhancing the endogenous antioxidant system including glutathione peroxidase, catalase and superoxide dismutase. This study examined the possible protective effects of probucol, a lipid-lowering compound with strong antioxidant properties, against CP-induced cardiotoxicity. This objective could be achieved through studying the gene expression-based on the possible protective effects of probucol against CP-induced cardiac failure in rats. Adult male Wistar albino rats were assigned into 4 treatment groups: Animals in the first (control) and second (probucol) groups were injected intraperitoneally with corn oil and probucol (61 mg/kg/day), respectively, for two weeks. Animals in the third (CP) and fourth (probucol plus CP) groups were injected with the same doses of corn oil and probucol (61 mg/kg/day), respectively, for one week before and one week after a single dose of CP (200 mg/kg, I.P.). The p53, Bax, Bcl2 and oxidative genes signal expression were measured by real time PCR. CP-induced cardiotoxicity was clearly observed by a significant increase in serum creatine phosphokinase isoenzyme (CK-MB) (117%), lactate dehydrogenase (LDH) (64%), free (69%) and esterified cholesterol (42%) and triglyceride (69%) compared to control group. In cardiac tissues, CP significantly increases the mRNA expression levels of apoptotic genes, p53 with 2 folds and Bax with 1.6 fold, and decreases the anti-apoptotic gene Bcl2 with 0.5 fold. Moreover, CP caused down-regulation of antioxidant genes, glutathione peroxidase, catalase, and superoxide dismutase and increased the lipid peroxidation and decreased adenosine triphosphate (ATP) (40%) and ATP/ADP (44%) in cardiac tissues. Probucol pretreatment not only counteracted significantly the CP-induced increase in cardiac enzymes and apoptosis but also it induced a significant increase in mRNA expression of antioxidant enzymes and improved ATP, ATP/ADP, glutathione (GSH) in cardiac tissues. In conclusion, data from the present study suggest that probucol prevents the development of CP-induced cardiotoxicity by a mechanism related, at least in part, to its ability to increase mRNA expression of antioxidant genes and to decrease apoptosis in cardiac tissues with the consequent improvement in mitochondrial oxidative phosphorylation and energy production.
Collapse
Affiliation(s)
- Yosef A Asiri
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| |
Collapse
|
155
|
Soria C, Estermann FE, Espantman KC, O’Shea CC. Heterochromatin silencing of p53 target genes by a small viral protein. Nature 2010; 466:1076-81. [PMID: 20740008 PMCID: PMC2929938 DOI: 10.1038/nature09307] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 06/23/2010] [Indexed: 01/26/2023]
Abstract
The transcription factor p53 (also known as TP53) guards against tumour and virus replication and is inactivated in almost all cancers. p53-activated transcription of target genes is thought to be synonymous with the stabilization of p53 in response to oncogenes and DNA damage. During adenovirus replication, the degradation of p53 by E1B-55k is considered essential for p53 inactivation, and is the basis for p53-selective viral cancer therapies. Here we reveal a dominant epigenetic mechanism that silences p53-activated transcription, irrespective of p53 phosphorylation and stabilization. We show that another adenoviral protein, E4-ORF3, inactivates p53 independently of E1B-55k by forming a nuclear structure that induces de novo H3K9me3 heterochromatin formation at p53 target promoters, preventing p53-DNA binding. This suppressive nuclear web is highly selective in silencing p53 promoters and operates in the backdrop of global transcriptional changes that drive oncogenic replication. These findings are important for understanding how high levels of wild-type p53 might also be inactivated in cancer as well as the mechanisms that induce aberrant epigenetic silencing of tumour-suppressor loci. Our study changes the longstanding definition of how p53 is inactivated in adenovirus infection and provides key insights that could enable the development of true p53-selective oncolytic viral therapies.
Collapse
Affiliation(s)
- Conrado Soria
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037-1099
| | - Fanny E. Estermann
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037-1099
| | - Kristen C. Espantman
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037-1099
| | - Clodagh C. O’Shea
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037-1099
| |
Collapse
|
156
|
ChIP (chromatin immunoprecipitation) analysis demonstrates co-ordinated binding of two transcription factors to the promoter of thep53tumour-suppressor gene. Cell Biol Int 2010; 34:883-91. [DOI: 10.1042/cbi20090401] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
157
|
Hunziker A, Jensen MH, Krishna S. Stress-specific response of the p53-Mdm2 feedback loop. BMC SYSTEMS BIOLOGY 2010; 4:94. [PMID: 20624280 PMCID: PMC2913930 DOI: 10.1186/1752-0509-4-94] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Accepted: 07/12/2010] [Indexed: 11/25/2022]
Abstract
Background The p53 signalling pathway has hundreds of inputs and outputs. It can trigger cellular senescence, cell-cycle arrest and apoptosis in response to diverse stress conditions, including DNA damage, hypoxia and nutrient deprivation. Signals from all these inputs are channeled through a single node, the transcription factor p53. Yet, the pathway is flexible enough to produce different downstream gene expression patterns in response to different stresses. Results We construct a mathematical model of the negative feedback loop involving p53 and its inhibitor, Mdm2, at the core of this pathway, and use it to examine the effect of different stresses that trigger p53. In response to DNA damage, hypoxia, etc., the model exhibits a wide variety of specific output behaviour - steady states with low or high levels of p53 and Mdm2, as well as spiky oscillations with low or high average p53 levels. Conclusions We show that even a simple negative feedback loop is capable of exhibiting the kind of flexible stress-specific response observed in the p53 system. Further, our model provides a framework for predicting the differences in p53 response to different stresses and single nucleotide polymorphisms.
Collapse
|
158
|
Mouchet N, Adamski H, Bouvet R, Corre S, Courbebaisse Y, Watier E, Mosser J, Chesné C, Galibert MD. In vivo identification of solar radiation-responsive gene network: role of the p38 stress-dependent kinase. PLoS One 2010; 5:e10776. [PMID: 20505830 PMCID: PMC2874014 DOI: 10.1371/journal.pone.0010776] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 05/04/2010] [Indexed: 02/03/2023] Open
Abstract
Solar radiation is one of the most common threats to the skin, with exposure eliciting a specific protective cellular response. To decrypt the underlying mechanism, we used whole genome microarrays (Agilent 44K) to study epidermis gene expression in vivo in skin exposed to simulated solar radiation (SSR). We procured epidermis samples from healthy Caucasian patients, with phototypes II or III, and used two different SSR doses (2 and 4 J/cm(2)), the lower of which corresponded to the minimal erythemal dose. Analyses were carried out five hours after irradiation to identify early gene expression events in the photoprotective response. About 1.5% of genes from the human genome showed significant changes in gene expression. The annotations of these affected genes were assessed. They indicated a strengthening of the inflammation process and up-regulation of the JAK-STAT pathway and other pathways. Parallel to the p53 pathway, the p38 stress-responsive pathway was affected, supporting and mediating p53 function. We used an ex vivo assay with a specific inhibitor of p38 (SB203580) to investigate genes the expression of which was associated with active p38 kinase. We identified new direct p38 target genes and further characterized the role of p38. Our findings provide further insight into the physiological response to UV, including cell-cell interactions and cross-talk effects.
Collapse
Affiliation(s)
- Nicolas Mouchet
- CNRS UMR 6061 Institut de Génétique et Développement de Rennes, Equipe RTO, Rennes, France
- Université de Rennes 1, IFR140 GFAS, Faculté de Médecine, Rennes, France
- PROCLAIM, Saint Grégoire, France
| | | | - Régis Bouvet
- CHU Rennes, Laboratoire de Génomique Médicale, Plateforme Transcriptomique GenOuest, Rennes, France
| | - Sébastien Corre
- CNRS UMR 6061 Institut de Génétique et Développement de Rennes, Equipe RTO, Rennes, France
- Université de Rennes 1, IFR140 GFAS, Faculté de Médecine, Rennes, France
| | | | - Eric Watier
- CHU Rennes, Service de Chirurgie Plastique, Rennes, France
| | - Jean Mosser
- CHU Rennes, Laboratoire de Génomique Médicale, Plateforme Transcriptomique GenOuest, Rennes, France
| | | | - Marie-Dominique Galibert
- CNRS UMR 6061 Institut de Génétique et Développement de Rennes, Equipe RTO, Rennes, France
- Université de Rennes 1, IFR140 GFAS, Faculté de Médecine, Rennes, France
- CHU Rennes, Laboratoire de Génomique Médicale, Plateforme Transcriptomique GenOuest, Rennes, France
- * E-mail:
| |
Collapse
|
159
|
Zhao CY, Szekely L, Bao W, Selivanova G. Rescue of p53 function by small-molecule RITA in cervical carcinoma by blocking E6-mediated degradation. Cancer Res 2010; 70:3372-81. [PMID: 20395210 DOI: 10.1158/0008-5472.can-09-2787] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Proteasomal degradation of p53 by human papilloma virus (HPV) E6 oncoprotein plays a pivotal role in the survival of cervical carcinoma cells. Abrogation of HPV-E6-dependent p53 destruction can therefore be a good strategy to combat cervical carcinomas. Here, we show that a small-molecule reactivation of p53 and induction of tumor cell apoptosis (RITA) is able to induce the accumulation of p53 and rescue its tumor suppressor function in cells containing high-risk HPV16 and HPV18 by inhibiting HPV-E6-mediated proteasomal degradation. RITA blocks p53 ubiquitination by preventing p53 interaction with E6-associated protein, required for HPV-E6-mediated degradation. RITA activates the transcription of proapoptotic p53 targets Noxa, PUMA, and BAX, and repressed the expression of pro-proliferative factors CyclinB1, CDC2, and CDC25C, resulting in p53-dependent apoptosis and cell cycle arrest. Importantly, RITA showed substantial suppression of cervical carcinoma xenografts in vivo. These results provide a proof of principle for the treatment of cervical cancer in a p53-dependent manner by using small molecules that target p53.
Collapse
Affiliation(s)
- Carolyn Ying Zhao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | |
Collapse
|
160
|
Yan W, Chen X. Characterization of functional domains necessary for mutant p53 gain of function. J Biol Chem 2010; 285:14229-38. [PMID: 20212049 DOI: 10.1074/jbc.m109.097253] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Tumor cells, including SW480 carcinoma cells that carry a mutant p53, are addicted to the mutant for their survival and resistance to growth suppression by chemotherapeutic agents. Here, we investigated whether various classes of p53 mutants share a common property and functional domains necessary for mutant p53 gain of function. To test this, we generated SW480 cell lines in which endogenous mutant R273H/P309S can be inducibly or stably knocked down, whereas a small interfering RNA-resistant mutant p53 along with a mutated functional domain can be inducibly or stably expressed. We found that both contact-site (R248W and R273H) and conformation (G245S and R249S) mutants are able to maintain the transformed phenotypes of SW480 cells conferred by endogenous mutant p53. We also found that activation domains 1-2 and the proline-rich domain are required for mutant p53 gain of function. Interestingly, we showed that the C-terminal basic domain, which is required for wild-type p53 activity, is an inhibitory domain for mutant p53. Furthermore, we showed that deletion of the basic domain enhances, whereas a mutation in activation domains 1-2 and deletion of the proline-rich domain abolish mutant p53 to regulate Gro1 and Id2, both of which are regulated by and mediate endogenous mutant p53 gain of function. These results indicate that both conformation and contact-site mutants share a property for cell transformation, and the domains critical for wild-type p53 tumor suppression are also required for mutant p53 tumor promotion. Thus, the inhibitory basic domain and the common property for p53 mutants can be explored for targeting tumors with mutant p53.
Collapse
Affiliation(s)
- Wensheng Yan
- Comparative Cancer Center, University of California, Davis, CA 95616, USA
| | | |
Collapse
|
161
|
Kon N, Kobayashi Y, Li M, Brooks CL, Ludwig T, Gu W. Inactivation of HAUSP in vivo modulates p53 function. Oncogene 2009; 29:1270-9. [PMID: 19946331 DOI: 10.1038/onc.2009.427] [Citation(s) in RCA: 151] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hausp is a deubiquitinase that has been shown to regulate the p53-Mdm2 pathway. Cotransfection of p53 and Hausp stabilizes p53 through the removal of ubiquitin moieties from polyubiquitinated p53. Interestingly, knockout or RNA interference-mediated knockdown of Hausp in human cells also resulted in the stabilization of p53 due to the destabilization of Mdm2, suggesting a dynamic role of Hausp in p53 activation. To understand the physiological functions of Hausp, we generated hausp knockout mice. Hausp knockout mice die during early embryonic development between embryonic days E6.5 and E7.5. The hausp knockout embryos showed p53 activation, but no apparent increase in apoptosis. Embryonic lethality was caused by a dramatic reduction in proliferation and termination in development, in part due to p53 activation and/or abrogation of p53-independent functions. Although deletion of p53 did not completely rescue the embryonic lethality of the hausp knockout, embryonic development was extended in both hausp and p53 double knockout embryos. These data show that Hausp has a critical role in regulating the p53-Mdm2 pathway.
Collapse
Affiliation(s)
- N Kon
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
162
|
Rajagopalan S, Sade RS, Townsley FM, Fersht AR. Mechanistic differences in the transcriptional activation of p53 by 14-3-3 isoforms. Nucleic Acids Res 2009; 38:893-906. [PMID: 19933256 PMCID: PMC2817464 DOI: 10.1093/nar/gkp1041] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
p53 maintains genome integrity by initiating the transcription of genes involved in cell-cycle arrest, senescence, apoptosis and DNA repair. The activity of p53 is regulated by both post-translational modifications and protein–protein interactions. p53 that has been phosphorylated at S366, S378 and T387 binds 14-3-3 proteins in vitro. Here, we show that these sites are potential 14-3-3 binding sites in vivo. Epsilon (ε) and gamma (γ) isoforms required phosphorylation at either of these sites for efficient interaction with p53, while for sigma (σ) and tau (τ) these sites are dispensable. Further, σ and τ bound more weakly to p53 C-terminal phosphopeptides than did ε and γ. However, the four isoforms bound tightly to di-phosphorylated p53 C-terminal peptides than did the mono-phosphorylated counterparts. Interestingly, all the isoforms studied transcriptionally activated wild-type p53. σ and τ stabilized p53 levels in cells, while ε and γ stimulated p53-DNA binding activity in vitro. Overall, the results suggest that structurally and functionally similar 14-3-3 isoforms may exert their regulatory potential on p53 through different mechanisms. We discuss the isoform-specific roles of 14-3-3 in p53 stabilization and activation of specific-DNA binding.
Collapse
|
163
|
Sahu G, Wang D, Chen CB, Zhurkin VB, Harrington RE, Appella E, Hager GL, Nagaich AK. p53 binding to nucleosomal DNA depends on the rotational positioning of DNA response element. J Biol Chem 2009; 285:1321-32. [PMID: 19887449 DOI: 10.1074/jbc.m109.081182] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The sequence-specific binding to DNA is crucial for the p53 tumor suppressor function. To investigate the constraints imposed on p53-DNA recognition by nucleosomal organization, we studied binding of the p53 DNA binding domain (p53DBD) and full-length wild-type p53 protein to a single p53 response element (p53RE) placed near the nucleosomal dyad in six rotational settings. We demonstrate that the strongest p53 binding occurs when the p53RE in the nucleosome is bent in the same direction as observed for the p53-DNA complexes in solution and in co-crystals. The p53RE becomes inaccessible, however, if its orientation in the core particle is changed by approximately 180 degrees. Our observations indicate that the orientation of the binding sites on a nucleosome may play a significant role in the initial p53-DNA recognition and subsequent cofactor recruitment.
Collapse
Affiliation(s)
- Geetaram Sahu
- Laboratory of Chemistry, Division of Therapeutic Proteins, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
164
|
Bukreeva EI, Aksenov ND, Bardin AA, Pospelov VA, Pospelova TV. Effect of histone deacetylase inhibitor sodium butyrate (NaB) on transformants E1a+cHa-Ras expressing wild type p53 with suppressed transactivation function. CELL AND TISSUE BIOLOGY 2009; 3:445-453. [DOI: 10.1134/s1990519x09050071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
165
|
Barsotti AM, Prives C. Pro-proliferative FoxM1 is a target of p53-mediated repression. Oncogene 2009; 28:4295-305. [PMID: 19749794 DOI: 10.1038/onc.2009.282] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The p53 tumor suppressor protein acts as a transcription factor to modulate cellular responses to a wide variety of stresses. In this study we show that p53 is required for the downregulation of FoxM1, an essential transcription factor that regulates many G2/M-specific genes and is overexpressed in a multitude of solid tumors. After DNA damage, p53 facilitates the repression of FoxM1 mRNA, which is accompanied by a decrease in FoxM1 protein levels. In cells with reduced p53 expression, FoxM1 is upregulated after DNA damage. Nutlin, a small-molecule activator of p53, suppresses FoxM1 levels in two cell lines in which DNA damage facilitates only mild repression. Mechanistically, p53-mediated inhibition of FoxM1 is partially p21 and retinoblastoma (Rb) family dependent, although in some cases p21-independent repression of FoxM1 was also observed. The importance of FoxM1 to cell fate was indicated by the observation that G2/M arrest follows FoxM1 ablation. Finally, our results indicate a potential contribution of p53-mediated repression of FoxM1 for maintenance of a stable G2 arrest.
Collapse
Affiliation(s)
- A M Barsotti
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | | |
Collapse
|
166
|
Scoumanne A, Zhang J, Chen X. PRMT5 is required for cell-cycle progression and p53 tumor suppressor function. Nucleic Acids Res 2009; 37:4965-76. [PMID: 19528079 PMCID: PMC2731901 DOI: 10.1093/nar/gkp516] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Protein arginine methyltransferases (PRMTs) mediate the transfer of methyl groups to arginines in proteins involved in signal transduction, transcriptional regulation and RNA processing. Tumor suppressor p53 coordinates crucial cellular processes, including cell-cycle arrest and DNA repair, in response to stress signals. Post-translational modifications and interactions with co-factors are important to regulate p53 transcriptional activity. To explore whether PRMTs modulate p53 function, we generated multiple cell lines in which PRMT1, CARM1 and PRMT5 are inducibly knocked down. Here, we showed that PRMT5, but not PRMT1 or CARM1, is essential for cell proliferation and PRMT5 deficiency triggers cell-cycle arrest in G1. In addition, PRMT5 is required for p53 expression and induction of p53 targets MDM2 and p21 upon DNA damage. Importantly, we established that PRMT5 knockdown prevents p53 protein synthesis. Furthermore, we found that PRMT5 regulates the expression of translation initiation factor eIF4E and growth suppression mediated upon PRMT5 knockdown is independent of p53 but is dependent on eIF4E. Taken together, we uncovered that arginine methyltransferase PRMT5 is a major pro-survival factor regulating eIF4E expression and p53 translation.
Collapse
Affiliation(s)
- A Scoumanne
- Center for Comparative Oncology, University of California at Davis, CA 95616, USA
| | | | | |
Collapse
|
167
|
Dhar SK, St Clair DK. Nucleophosmin blocks mitochondrial localization of p53 and apoptosis. J Biol Chem 2009; 284:16409-16418. [PMID: 19366707 PMCID: PMC2713525 DOI: 10.1074/jbc.m109.005736] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 04/08/2009] [Indexed: 01/26/2023] Open
Abstract
Activation of p53 is an important mechanism in apoptosis. However, whether the presence of p53 in mitochondria plays an important role in p53-mediated apoptosis is unclear. Here, we demonstrate that overexpression of NPM (nucleophosmin) significantly suppresses 12-O-tetradecanoylphorbol 13-acetate (TPA)-mediated apoptosis, in part, by blocking the mitochondrial localization of p53. Within 1 h following TPA treatment of skin epithelial (JB6) cells, p53 accumulated in mitochondria. Expression of NPM enhances p53 levels in the nucleus but reduces p53 levels in mitochondria, as detected by immunocytochemistry and Western blot analysis. The suppressive effect of NPM on p53 mitochondrial localization is also observed in TPA-treated primary epithelial cells and in JB6 cells treated with doxorubicin. NPM enhances the expression of p53 target gene p21 and bax. However, the increase in Bax level in the absence of p53 in mitochondria did not lead to an increase in TPA-induced apoptosis, suggesting that the presence of p53 in mitochondria is important. Suppression of NPM by NPM small interfering RNA leads to an increase of p53 levels in mitochondria and apoptosis. Furthermore, suppression of NPM in tumor cells with a high constitutive level of NPM results in p53 translocation to mitochondria and enhances TPA-mediated apoptosis. The results demonstrate the effect of NPM on p53 localization in mitochondria and apoptosis. Together, the data indicate that the presence of p53 in mitochondria plays an important role in stress-induced apoptosis and suggest that NPM may protect cells from apoptosis by reducing the mitochondrial level of p53.
Collapse
Affiliation(s)
- Sanjit Kumar Dhar
- From the Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky 40536
| | - Daret K St Clair
- From the Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky 40536.
| |
Collapse
|
168
|
Ahn J, Poyurovsky MV, Baptiste N, Beckerman R, Cain C, Mattia M, McKinney K, Zhou J, Zupnick A, Gottifredi V, Prives C. Dissection of the sequence-specific DNA binding and exonuclease activities reveals a superactive yet apoptotically impaired mutant p53 protein. Cell Cycle 2009; 8:1603-15. [PMID: 19462533 PMCID: PMC2898518 DOI: 10.4161/cc.8.10.8548] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Both sequence-specific DNA binding and exonuclease activities have been mapped to the central conserved core domain of p53. To gain more information about these two activities a series of mutants were generated that changed core domain histidine residues. Of these mutants, only one, H115N p53, showed markedly reduced exonuclease activity (ca. 15% of wild-type). Surprisingly, purified H115N p53 protein was found to be significantly more potent than wild-type p53 in binding to DNA by several criteria including gel mobility shift assay, filter binding and DNase I footprinting. Interestingly as well, non-specific DNA binding by the core domain of H115N p53 is superior to that of wild-type p53. To study H115N p53 in vivo, clones of H1299 cells expressing tetracycline regulated wild-type or H115N p53 were generated. H115N was both more potent than wild-type p53 in inducing p53 target genes such as p21 and PIG3 and was also more effective in arresting cells in G1. Unexpectedly, in contrast to wild-type p53, H115N p53 was markedly impaired in causing apoptosis when cells were subjected to DNA damage. Our results indicate that the exonuclease activity and transcriptional activation functions of p53 can be separated. They also extend previous findings showing that cell cycle arrest and apoptosis are separable functions of p53. Finally, these experiments confirm that DNA binding and xonuclease activities are distinct features of the p53 core domain.
Collapse
Affiliation(s)
- Jinwoo Ahn
- Department of Structural Biology; University of Pittsburgh School of Medicine; Pittsburgh, PA USA
| | | | - Nicole Baptiste
- Department of Biological Sciences; Columbia University; New York, NY USA
| | - Rachel Beckerman
- Department of Biological Sciences; Columbia University; New York, NY USA
| | - Christine Cain
- Department of Biological Sciences; Columbia University; New York, NY USA
| | - Melissa Mattia
- Department of Oncological Sciences; Mount Sinai School of Medicine; New York, NY USA
| | - Kristine McKinney
- Dana-Farber Cancer Institute; Harvard Medical School; Boston, MA USA
| | - Jianmin Zhou
- Department of Biological Sciences; Columbia University; New York, NY USA
| | - Andrew Zupnick
- Department of Biological Sciences; Columbia University; New York, NY USA
| | - Vanesa Gottifredi
- Cell Cycle and Genomic Stability Laboratory; Fundación Instituto Leloir-CONICET; Universidad de Buenos Aires; Buenos Aires, Argentina
| | - Carol Prives
- Department of Biological Sciences; Columbia University; New York, NY USA
| |
Collapse
|
169
|
Yoshizawa J, Takizawa A, Takeuchi O, Hiraku O, Sasaki K, Morimoto Y, Atsuda K, Inoue G, Suzuki Y, Asanuma F, Yamada Y. Experimental study of combination therapy with S-1 against pancreatic cancer. Cancer Chemother Pharmacol 2009; 64:1211-9. [DOI: 10.1007/s00280-009-0990-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2008] [Accepted: 03/12/2009] [Indexed: 11/30/2022]
|
170
|
Hua G, Wang S, Zhong C, Xue P, Fan Z. Ignition of p53 bomb sensitizes tumor cells to granzyme K-mediated cytolysis. THE JOURNAL OF IMMUNOLOGY 2009; 182:2152-9. [PMID: 19201868 DOI: 10.4049/jimmunol.0802307] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Inactivation of tumor suppressor p53 results in loss of the apoptosis-regulating function of the p53 protein in tumor cells. Restoration of wild-type p53 expression in p53 mutant tumor cells increases tumor susceptibility to CTL-mediated cytolysis. However, the direct role of p53 in regulating tumor sensitivity to NK cell-mediated lysis and the functional relationship between p53 and granzymes in the control of tumor killing are still poorly documented. In this study, we found that p53 can sensitize tumor-killing susceptibility to NK and granzyme K-mediated cytolysis. Granzyme K is constitutively expressed in high levels in NK cells and induces rapid caspase-independent cell death. Granzyme K may exert a critical role in NK cell-mediated tumor clearance. p53 associates with granzyme K and is a physiological substrate of granzyme K. p53 was processed to three cleavage products of p40, p35, and p13 fragments at Lys(24) and Lys(305). These three cleavage products harbor strong proapoptotic activities that amplify the proapoptotic action of p53 to potentiate tumor-killing sensitivity. Therefore, p53 is as a cytotoxic bomb that can be triggered by granzyme K, leading to potentiating killing efficacy.
Collapse
Affiliation(s)
- Guoqiang Hua
- National Laboratory of Biomacromolecules and Center for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | |
Collapse
|
171
|
Nozyński J, Zakliczyński M, Zembala-Nozyńska E, Konecka-Mrówká D, Przybylski R, Nikiel B, Mlynarczyk-Liszka J, Lange D, Mrówka A, Przybylski J, Maruszewski M, Zembala M. Cardiocyte nuclear chromatin density correlates with transplanted heart left ventricular mass. Transplant Proc 2009; 41:281-4. [PMID: 19249535 DOI: 10.1016/j.transproceed.2008.10.067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 09/24/2008] [Accepted: 10/15/2008] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Cardiocyte hypertrophy is accompanied by polyploidy, seen as a decrease in chromatin density in the enlarged nucleus. Repeated biopsies of a transplanted heart offer the possibility of a dynamic evaluation of these phenomena. The aim of this work was an evaluation of cardiocyte nuclear chromatin density in transplanted hearts during long-term follow-up. MATERIALS AND METHODS The material encompassed myocardial biopsy specimens taken during the first week, first month, and then on an annual basis up to 10 years after surgery. Only biopsy specimens with no rejection were considered (grade "0" International Society for Heart and Lung Transplantation [ISHLT] 122 biopsy specimens). The control group consisted of 7 donor heart specimens. We evaluated the optical density-mean gray level-of cardiomyocyte nuclear chromatin. We determined correlations of this index with the nuclear area, and with left ventricle ultrasound measurements, using correlation analysis. RESULTS The chromatin mean gray level decreased with time, correlating positively with interventricular septum thickness, left ventricle posterior wall diameter, and left ventricular mass. Analysis of individual periods showed a significant positive correlation of the mean grey level with the cardiocyte nuclear surface in year 3, 4, and 9 after transplantation, thereby suggesting the occurrence of polyploidy at those times. The significant negative correlation of these values (1 week and 1 year) indicated normalization of early cardiocyte hypertrophy. CONCLUSIONS With the passage of time chromatin condenses, leading to pyknosis. The activity of cardiocyte chromatin correlated with left ventricular hypertrophy. Compensatory cardiomyocyte polyploidy is a periodical phenomenon.
Collapse
Affiliation(s)
- J Nozyński
- Department of Cardiac Surgery & Transplantation, Silesian Center for Heart Disease, Zabrze, Poland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Yu X, Riley T, Levine AJ. The regulation of the endosomal compartment by p53 the tumor suppressor gene. FEBS J 2009; 276:2201-12. [PMID: 19302216 DOI: 10.1111/j.1742-4658.2009.06949.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The endosomal compartment of the cell is involved in a number of functions including: (a) internalizing membrane proteins to multivesicular bodies and lysosomes; (b) producing vesicles that are secreted from the cell (exosomes); and (c) generating autophagic vesicles that, especially in times of nutrient deprivation, supply cytoplasmic components to the lysosome for degradation and recycling of nutrients. The p53 protein responds to various stress signals by initiating a transcriptional program that restores cellular homeostasis and prevents the accumulation of errors in a cell. As part of this process, p53 regulates the transcription of a set of genes encoding proteins that populate the endosomal compartment and impact upon each of these endosomal functions. Here, we demonstrate that p53 regulates transcription of the genes TSAP6 and CHMP4C, which enhance exosome production, and CAV1 and CHMP4C, which produce a more rapid endosomal clearance of the epidermal growth factor receptor from the plasma membrane. Each of these p53-regulated endosomal functions results in the slowing of cell growth and division, the utilization of catabolic resources and cell-to-cell communication by exosomes after a stress signal is detected by the p53 protein. These processes avoid errors during stress and restore homeostasis once the stress is resolved.
Collapse
Affiliation(s)
- Xin Yu
- The Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ, USA
| | | | | |
Collapse
|
173
|
Nathan SS, Pereira BP, Zhou YF, Gupta A, Dombrowski C, Soong R, Pho RW, Stein GS, Salto-Tellez M, Cool SM, van Wijnen AJ. Elevated expression of Runx2 as a key parameter in the etiology of osteosarcoma. Mol Biol Rep 2009; 36:153-8. [PMID: 18931939 PMCID: PMC2981062 DOI: 10.1007/s11033-008-9378-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Accepted: 09/30/2008] [Indexed: 11/28/2022]
Abstract
To understand the molecular etiology of osteosarcoma, we isolated and characterized a human osteosarcoma cell line (OS1). OS1 cells have high osteogenic potential in differentiation induction media. Molecular analysis reveals OS1 cells express the pocket protein pRB and the runt-related transcription factor Runx2. Strikingly, Runx2 is expressed at higher levels in OS1 cells than in human fetal osteoblasts. Both pRB and Runx2 have growth suppressive potential in osteoblasts and are key factors controlling competency for osteoblast differentiation. The high levels of Runx2 clearly suggest osteosarcomas may form from committed osteoblasts that have bypassed growth restrictions normally imposed by Runx2. Interestingly, OS1 cells do not exhibit p53 expression and thus lack a functional p53/p21 DNA damage response pathway as has been observed for other osteosarcoma cell types. Absence of this pathway predicts genomic instability and/or vulnerability to secondary mutations that may counteract the anti-proliferative activity of Runx2 that is normally observed in osteoblasts. We conclude OS1 cells provide a valuable cell culture model to examine molecular events that are responsible for the pathologic conversion of phenotypically normal osteoblast precursors into osteosarcoma cells.
Collapse
Affiliation(s)
- S. Suresh Nathan
- Musculoskeletal Research Laboratories, Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 10 Lower Kent Ridge Road, S119074, SINGAPORE
| | - Barry P. Pereira
- Musculoskeletal Research Laboratories, Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 10 Lower Kent Ridge Road, S119074, SINGAPORE
| | - Ye-fang Zhou
- Oncology Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 28 Medical Drive, Singapore 117456, SINGAPORE
| | - Anurag Gupta
- Oncology Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 28 Medical Drive, Singapore 117456, SINGAPORE
| | - Christian Dombrowski
- Laboratory of Stem Cells and Tissue Repair, Institute of Medical Biology, Singapore, 61 Biopolis Place, Proteos, Singapore 138673, SINGAPORE
| | - Ritchie Soong
- Oncology Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 28 Medical Drive, Singapore 117456, SINGAPORE
| | - Robert W.H. Pho
- Musculoskeletal Research Laboratories, Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 10 Lower Kent Ridge Road, S119074, SINGAPORE
| | - Gary S. Stein
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Manuel Salto-Tellez
- Oncology Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 28 Medical Drive, Singapore 117456, SINGAPORE
| | - Simon M. Cool
- Laboratory of Stem Cells and Tissue Repair, Institute of Medical Biology, Singapore, 61 Biopolis Place, Proteos, Singapore 138673, SINGAPORE
| | - Andre J. van Wijnen
- Musculoskeletal Research Laboratories, Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 10 Lower Kent Ridge Road, S119074, SINGAPORE
- Oncology Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 28 Medical Drive, Singapore 117456, SINGAPORE
- Department of Cell Biology and Cancer Center, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
174
|
Karni-Schmidt O, Zupnick A, Castillo M, Ahmed A, Matos T, Bouvet P, Cordon-Cardo C, Prives C. p53 is localized to a sub-nucleolar compartment after proteasomal inhibition in an energy-dependent manner. J Cell Sci 2008; 121:4098-105. [PMID: 19033390 DOI: 10.1242/jcs.030098] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The tumor suppressor p53 is activated in response to many forms of cellular stress leading to cell cycle arrest, senescence or apoptosis. Appropriate sub-cellular localization is essential for modulating p53 function. We recently showed that p53 localizes to the nucleolus after proteasome inhibition with MG132 and this localization requires sequences within its carboxyl terminus. In the present study, we found that after treatment with MG132, p53 associates with a discrete sub-nucleolar component, the fibrillar center (FC), a region mainly enriched with RNA polymerase I. Moreover, we now demonstrate that this localization is an energy-dependent process as reduction of ATP levels prevents nucleolar localization. In addition, p53 sub-nucleolar accumulation is abolished when cells are subjected to various types of genotoxic stress. Furthermore, we show that monoubiquitination of p53, which causes it to localize to the cytoplasm and nucleoplasm, does not prevent the association of p53 with the nucleolus after MG132 treatment. Importantly, we demonstrate that p53 nucleolar association occurs in lung and bladder carcinomas.
Collapse
Affiliation(s)
- Orit Karni-Schmidt
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | | | | | | | | | | | | |
Collapse
|
175
|
Abstract
The p53 tumor suppressor continues to hold distinction as the most frequently mutated gene in human cancer. The ability of p53 to induce programmed cell death, or apoptosis, of cells exposed to environmental or oncogenic stress constitutes a major pathway whereby p53 exerts its tumor suppressor function. In the past decade, we have discovered that p53 is not alone in its mission to destroy damaged or aberrantly proliferating cells: it has two homologs, p63 and p73, that in various cellular contexts and stresses contribute to this process. In this review, the mechanisms whereby p53, and in some cases p63 and p73, induce apoptosis are discussed. Other reviews have focused more extensively on the contribution of individual p53-regulated genes to apoptosis induction by this protein, whereas in this review, we focus more on those factors that mediate the decision between growth arrest and apoptosis by p53, p63 and p73, and on the post-translational modifications and protein-protein interactions that influence this decision.
Collapse
Affiliation(s)
- E. Christine Pietsch
- Division of Medical Sciences, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia PA, 19111
| | - Stephen M. Sykes
- Brigham and Women's Hospital, 1 Blackfan Circle, Boston, MA 02115
| | - Steven B. McMahon
- Kimmel Cancer Center, Thomas Jefferson Medical College, 233 S. 10th St. Philadelphia, Pennsylvania 19107
| | - Maureen E. Murphy
- Division of Medical Sciences, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia PA, 19111
| |
Collapse
|
176
|
Zhang J, Zhu B, Liu Y, Jiang Z, Wang Y, Li Y, Hua H, Wang Z. High Expression of Circadian Gene mPer2 Diminishes Radiosensitivity of Tumor Cells. Cancer Biother Radiopharm 2008; 23:561-70. [PMID: 18999929 DOI: 10.1089/cbr.2008.0496] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jing Zhang
- Health Ministry Key Laboratory of Chronobiology, West China Medical Center, Sichuan University, Chengdu, People's Republic of China
| | - Bin Zhu
- Health Ministry Key Laboratory of Chronobiology, West China Medical Center, Sichuan University, Chengdu, People's Republic of China
| | - Yanyou Liu
- Health Ministry Key Laboratory of Chronobiology, West China Medical Center, Sichuan University, Chengdu, People's Republic of China
| | - Zhou Jiang
- Health Ministry Key Laboratory of Chronobiology, West China Medical Center, Sichuan University, Chengdu, People's Republic of China
| | - Yuhui Wang
- Health Ministry Key Laboratory of Chronobiology, West China Medical Center, Sichuan University, Chengdu, People's Republic of China
| | - Ying Li
- Health Ministry Key Laboratory of Chronobiology, West China Medical Center, Sichuan University, Chengdu, People's Republic of China
| | - Hui Hua
- Health Ministry Key Laboratory of Chronobiology, West China Medical Center, Sichuan University, Chengdu, People's Republic of China
| | - Zhengrong Wang
- Health Ministry Key Laboratory of Chronobiology, West China Medical Center, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
177
|
Salah Z, Haupt S, Maoz M, Baraz L, Rotter V, Peretz T, Haupt Y, Bar-Shavit R. p53 controls hPar1 function and expression. Oncogene 2008; 27:6866-74. [DOI: 10.1038/onc.2008.324] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
178
|
Saffari M, Dinehkabodi OS, Ghaffari SH, Modarressi MH, Mansouri F, Heidari M. Identification of novel p53 target genes by cDNA AFLP in glioblastoma cells. Cancer Lett 2008; 273:316-22. [PMID: 18814959 DOI: 10.1016/j.canlet.2008.08.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Revised: 05/27/2008] [Accepted: 08/13/2008] [Indexed: 11/28/2022]
Abstract
The p53 plays critical role in cellular functions such as cell cycle arrest and apoptosis. We overexpressed wild-type p53 (wt-p53) in U87 glioblastoma cells via recombinant adenovirus Ad-GFP-P53 which encodes p53 and green fluorescent protein. The transcript profiles were investigated using cDNA amplified fragment length polymorphism approach. Semi-quantitative RT-PCR and DNA sequencing results for the selected genes showed that Cathepsin B and cell cycle associated protein-1 or Caprin-I, genes were suppressed whereas Annexin-II gene overexpressed in response to the overexpression of wt-p53 gene. Our results suggest that these genes could be important mediators of p53-dependent tumor growth suppression in glioblastoma.
Collapse
Affiliation(s)
- Mojtaba Saffari
- Department of Medical Genetics, Tehran University of Medical Sciences, Pour Sina Avenue, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
179
|
Characterization of the p53 response to oncogene-induced senescence. PLoS One 2008; 3:e3230. [PMID: 18800172 PMCID: PMC2535567 DOI: 10.1371/journal.pone.0003230] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Accepted: 08/24/2008] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND P53 activation can trigger various outcomes, among them reversible growth arrest or cellular senescence. It is a live debate whether these outcomes are influenced by quantitative or qualitative mechanisms. Furthermore, the relative contribution of p53 to Ras-induced senescence is also matter of controversy. METHODOLOGY/PRINCIPAL FINDINGS This study compared situations in which different signals drove senescence with increasing levels of p53 activation. The study revealed that the levels of p53 activation do not determine the outcome of the response. This is further confirmed by the clustering of transcriptional patterns into two broad groups: p53-activated or p53-inactivated, i.e., growth and cellular arrest/senescence. Furthermore, while p53-dependent transcription decreases after 24 hrs in the presence of active p53, senescence continues. Maintaining cells in the arrested state for long periods does not switch reversible arrest to cellular senescence. Together, these data suggest that a Ras-dependent, p53-independent, second signal is necessary to induce senescence. This study tested whether PPP1CA (the catalytic subunit of PP1alpha), recently identified as contributing to Ras-induced senescence, might be this second signal. PPP1CA is induced by Ras; its inactivation inhibits Ras-induced senescence, presumably by inhibiting pRb dephosphorylation. Finally, PPP1CA seems to strongly co-localize with pRb only during senescence. CONCLUSIONS The levels of p53 activation do not determine the outcome of the response. Rather, p53 activity seems to act as a necessary but not sufficient condition for senescence to arise. Maintaining cells in the arrested state for long periods does not switch reversible arrest to cellular senescence. PPP1CA is induced by Ras; its inactivation inhibits Ras-induced senescence, presumably by inhibiting pRb dephosphorylation. Finally, PPP1CA seems to strongly co-localize with pRb only during senescence, suggesting that PP1alpha activation during senescence may be the second signal contributing to the irreversibility of the senescent phenotype.
Collapse
|
180
|
A complex barcode underlies the heterogeneous response of p53 to stress. Nat Rev Mol Cell Biol 2008; 9:702-12. [DOI: 10.1038/nrm2451] [Citation(s) in RCA: 314] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
181
|
Ban J, Bennani-Baiti IM, Kauer M, Schaefer KL, Poremba C, Jug G, Schwentner R, Smrzka O, Muehlbacher K, Aryee DN, Kovar H. EWS-FLI1 suppresses NOTCH-activated p53 in Ewing's sarcoma. Cancer Res 2008; 68:7100-9. [PMID: 18757425 PMCID: PMC4964910 DOI: 10.1158/0008-5472.can-07-6145] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Although p53 is the most frequently mutated gene in cancer, half of human tumors retain wild-type p53, whereby it is unknown whether normal p53 function is compromised by other cancer-associated alterations. One example is Ewing's sarcoma family tumors (ESFT), where 90% express wild-type p53. ESFT are characterized by EWS-FLI1 oncogene fusions. Studying 6 ESFT cell lines, silencing of EWS-FLI1 in a wild-type p53 context resulted in increased p53 and p21(WAF1/CIP1) levels, causing cell cycle arrest. Using a candidate gene approach, HEY1 was linked to p53 induction. HEY1 was rarely expressed in 59 primary tumors, but consistently induced upon EWS-FLI1 knockdown in ESFT cell lines. The NOTCH signaling pathway targets HEY1, and we show NOTCH2 and NOTCH3 to be expressed in ESFT primary tumors and cell lines. Upon EWS-FLI1 silencing, NOTCH3 processing accompanied by nuclear translocation of the activated intracellular domain was observed in all but one p53-mutant cell line. In cell lines with the highest HEY1 induction, NOTCH3 activation was the consequence of JAG1 transcriptional induction. JAG1 modulation by specific siRNA, NOTCH-processing inhibition by either GSI or ectopic NUMB1, and siRNA-mediated HEY1 knockdown all inhibited p53 and p21(WAF1/CIP1) induction. Conversely, forced expression of JAG1, activated NOTCH3, or HEY1 induced p53 and p21(WAF1/CIP1). These results indicate that suppression of EWS-FLI1 reactivates NOTCH signaling in ESFT cells, resulting in p53-dependent cell cycle arrest. Our data link EWS-FLI1 to the NOTCH and p53 pathways and provide a plausible basis both for NOTCH tumor suppressor effects and oncogenesis of cancers that retain wild-type p53.
Collapse
Affiliation(s)
- Jozef Ban
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | | | - Max Kauer
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | | | | | - Gunhild Jug
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Raphaela Schwentner
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Oskar Smrzka
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Karin Muehlbacher
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Dave N.T. Aryee
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Heinrich Kovar
- Children’s Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| |
Collapse
|
182
|
Maran A, Shogren KL, Benedikt M, Sarkar G, Turner RT, Yaszemski MJ. 2-methoxyestradiol-induced cell death in osteosarcoma cells is preceded by cell cycle arrest. J Cell Biochem 2008; 104:1937-45. [PMID: 18384113 PMCID: PMC2821714 DOI: 10.1002/jcb.21758] [Citation(s) in RCA: 21] [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] [Indexed: 12/20/2022]
Abstract
2-Methoxyestradiol (2-ME), a naturally occurring mammalian metabolite of 17beta-Estradiol (E2), induces cell death in osteosarcoma cells. To further understand the molecular mechanisms of action, we have investigated cell cycle progression in 2-ME-treated human osteosarcoma (MG63, SaOS-2 and LM7 [corrected]) cells. At 5 microM, 2-ME induced growth arrest by inducing a block in cell cycle; 2-ME-treatment resulted in 2-fold increases in G1 phase cells and a decrease in S phase cells in MG63 and SaOS-2 osteosarcoma cell lines, compared to the appropriate vehicle controls. 2-ME-treatment induced a threefold increase in the G2 phase in LM7 [corrected] osteosarcoma cells. The results demonstrated steroid specificity, as the tumorigenic metabolite, 16alpha-hydroxyestradiol (16-OHE), did not have any effect on cell cycle progression in osteosarcoma cells. The cell cycle arrest coincided with an increase in expression of the cell cycle markers p21, p27 and p53 proteins in 2-ME-treated osteosarcoma cells. Also, MG63 cells, transiently transfected with cDNA for a 'loss of function mutant' RNA-dependent protein kinase (PKR) protein, were resistant to 2-ME-induced cell cycle arrest. These results suggest that 2-ME works in concert with factors regulating cell cycle progression, and cell cycle arrest precedes cell death in 2-ME-treated osteosarcoma cells.
Collapse
Affiliation(s)
- Avudaiappan Maran
- Department of Orthopedics, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | |
Collapse
|
183
|
Guo XR, Cheng B, Zheng YC, Lin ST, Li PY. Effects of down-regulation of p21 by HBx gene on HepG2 cell proliferation and apoptosis. Shijie Huaren Xiaohua Zazhi 2008; 16:2080-2085. [DOI: 10.11569/wcjd.v16.i19.2080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish gene-transfected cell strain HepG2/HBx and study the effect of HBx on cell cycle, proliferation and apoptosis of HepG2 cells as well as the potential regulative role of p21.
METHODS: HBx was transfected into HepG2 cells and G418 selection was used to obtain the positive clones of HepG2/HBx cells. Then HBx mRNA expression and protein expression were detected using RT-PCR and western blot analysis respectively. MTT assay and flow cytometry were adopted to measure the proliferation, cell cycle and apoptosis of HepG2/HBx, HepG2 and HepG2/pcDNA3.1 (HepG2 cells transfected with pcDNA3.1) cells. Semi-quantified RT-PCR was used to evaluate the expression of p21 and p53 in three groups.
RESULTS: The expression of mRNA and protein of HBx in HepG2/HBx cells was confirmed by RT-PCR and western blot respectively. The proliferation of HepG2/HBx cells was accelerated. The proportion of HepG2/HBx cells decreased significantly in G0/G1 phase (43.34% ± 3.11% vs 57.69 ± 4.28%, P < 0.01), but increased remarkably in S phase (28.69% ± 1.17% vs 22.41% ± 1.99%, P < 0.05) and the apoptosis rate of HepG2/HBx cells was at a significantly lower level (1.19% ± 0.06% vs 5.43% ± 0.42%, P < 0.001). Compared with HepG2 and HepG2/pcDNA3.1 cells, the expression of p21 mRNA in HepG2/HBx was down-regulated (0.16 ± 0.05 vs 0.78 ± 0.15, P < 0.001), while there was no significant difference in the expression of p53 gene.
CONCLUSION: The HBx gene down-regulates the expression of p21 mRNA, which may play an important role in accelerating cell cycle, improving growth and inhibiting apoptosis of HepG2 cells.
Collapse
|
184
|
Millau JF, Bastien N, Drouin R. P53 transcriptional activities: a general overview and some thoughts. Mutat Res 2008; 681:118-133. [PMID: 18639648 DOI: 10.1016/j.mrrev.2008.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2007] [Revised: 05/05/2008] [Accepted: 06/19/2008] [Indexed: 12/20/2022]
Abstract
P53 is a master transcriptional regulator controlling several main cellular pathways. Its role is to adapt gene expression programs in order to maintain cellular homeostasis and genome integrity in response to stresses. P53 is found mutated in about half of human cancers and most mutations are clustered within the DNA-binding domain of the protein resulting in altered p53 transcriptional activity. This illustrates the importance of the gene regulations achieved by p53. The aim of this review is to provide a global overview of the current understanding of p53 transcriptional activities and to discuss some ongoing questions and unresolved points about p53 transcriptional activity.
Collapse
Affiliation(s)
- Jean-François Millau
- Service of Genetics, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke J1H 5N4, QC, Canada
| | - Nathalie Bastien
- Service of Genetics, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke J1H 5N4, QC, Canada
| | - Régen Drouin
- Service of Genetics, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12th Avenue North, Sherbrooke J1H 5N4, QC, Canada.
| |
Collapse
|
185
|
Multiple pathways are involved in drug resistance to doxorubicin in an osteosarcoma cell line. Anticancer Drugs 2008; 19:257-65. [PMID: 18510171 DOI: 10.1097/cad.0b013e3282f435b6] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Drug resistance continues to be a stumbling block in achieving a better cure rate in several cancers, including osteosarcoma. To understand this, we developed a doxorubicin drug-resistant osteosarcoma cell line (143B-DR-DOX). This cell line had an IC50 of 75 micromol/l compared with the parental 143B cell line's IC50 of 0.4 micromol/l. Using a 22000 70-mer oligomicroarray, gene expression studies were performed in four replicates. Data analysis was done using the TIGR Microarray suite. Seventy-four genes were found to be either upregulated (21) or downregulated (53). Real time quantitative-PCR was done on 21 genes, which confirmed the gene expression data for 11 genes. Choosing the significant fold change criteria of greater than 2-fold upregulation or downregulation, four genes including multidrug resistance 1, interleukin-8, Krüppel-like factor 2 and MGC4175 were found to be upregulated and seven genes including epidermal growth factor receptor-coamplified and overexpressed protein, uridine phosphorylase 1, a disintegrin and metalloproteinase domain 19, cytochrome C1, SEC, S-adenosyl homocysteine hydrolase and p53 were found to be downregulated. The data suggest that apart from the known gene alterations in doxorubicin resistance (multidrug resistance 1, topoisomerase IIbeta), others can also contribute to the drug-resistance phenotype. The involvement of interleukin-8 and Krüppel-like factor 2 suggests that the peroxisome proliferator-activated receptors gamma pathway may also be involved in doxorubicin drug resistance in the 143B-DR-DOX cell line.
Collapse
|
186
|
Shangary S, Ding K, Qiu S, Nikolovska-Coleska Z, Bauer JA, Liu M, Wang G, Lu Y, McEachern D, Bernard D, Bradford CR, Carey TE, Wang S. Reactivation of p53 by a specific MDM2 antagonist (MI-43) leads to p21-mediated cell cycle arrest and selective cell death in colon cancer. Mol Cancer Ther 2008; 7:1533-42. [PMID: 18566224 PMCID: PMC2494594 DOI: 10.1158/1535-7163.mct-08-0140] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
MDM2 oncoprotein binds directly to the p53 tumor suppressor and inhibits its function in cancers retaining wild-type p53. Blocking this interaction using small molecules is a promising approach to reactivate p53 function and is being pursued as a new anticancer strategy. The spiro-oxindole MI-43, a small-molecule inhibitor of the MDM2-p53 interaction, was designed and examined for its cellular mechanism of action and therapeutic potential in colon cancer. MI-43 binds to MDM2 protein with a K(i) value of 18 nmol/L and is 300 times more potent than a native p53 peptide. MI-43 blocks the intracellular MDM2-p53 interaction and induces p53 accumulation in both normal and cancer cells, with wild-type p53 without causing p53 phosphorylation. Induction of p53 leads to modulation of the expression of p53 target genes, including up-regulation of p21 and MDM2 in normal primary human cells and in colon cancer cells with wild-type p53. Using HCT-116 isogenic colon cancer cell lines differing only in p53 status or RNA interference to knockdown expression of p53 in the RKO colon cancer cell line, we show that the cell growth inhibition and cell death induction by MI-43 is p53 dependent. Furthermore, induction of cell cycle arrest by MI-43 is dependent on p53 and p21. In normal cells, MI-43 induces cell cycle arrest but not apoptosis. This study suggests that p53 activation by a potent and specific spiro-oxindole MDM2 antagonist may represent a promising therapeutic strategy for the treatment of colon cancer and should be further evaluated in vivo and in the clinic.
Collapse
Affiliation(s)
- Sanjeev Shangary
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Ke Ding
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Su Qiu
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Zaneta Nikolovska-Coleska
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Joshua A. Bauer
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Pharmacology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Meilan Liu
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Guoping Wang
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Yipin Lu
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Medicinal Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Donna McEachern
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Denzil Bernard
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Carol R. Bradford
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Thomas E. Carey
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Pharmacology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Shaomeng Wang
- Comprehensive Cancer Center, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Pharmacology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
- Department of Medicinal Chemistry, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109, USA
| |
Collapse
|
187
|
Buck M. A novel domain of BRCA1 interacts with p53 in breast cancer cells. Cancer Lett 2008; 268:137-45. [PMID: 18501503 DOI: 10.1016/j.canlet.2008.03.061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2007] [Revised: 03/25/2008] [Accepted: 03/26/2008] [Indexed: 02/02/2023]
Abstract
The interactions between BRCA1 and p53 are relevant for understanding hereditary breast and ovarian cancer. Although in vitro studies reported that BRCA1 (amino acids 224-500) and the second BRCT domain of the BRCA1 C-terminus may interact with p53, quantitative biophysical measurements indicate that these regions of BRCA1 do not bind efficiently to p53. Here we show that BRCA1 interacts with p53 in vivo in breast cancer cells, through another BRCA1 domain (amino acids 772-1292). Expression of a truncated BRCA1 (amino acids 772-1292) stimulated p53 DNA-binding and transcription activities and apoptosis, recapitulating some effects of DNA damage. These results suggest that a novel domain of BRCA1 may interact with p53 in breast cancer cells.
Collapse
Affiliation(s)
- Martina Buck
- Department of Medicine, Veterans Healthcare Medical Center, San Diego, CA 92161, USA.
| |
Collapse
|
188
|
Lee DH, Kim C, Zhang L, Lee YJ. Role of p53, PUMA, and Bax in wogonin-induced apoptosis in human cancer cells. Biochem Pharmacol 2008; 75:2020-33. [PMID: 18377871 PMCID: PMC2424318 DOI: 10.1016/j.bcp.2008.02.023] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 02/05/2008] [Accepted: 02/20/2008] [Indexed: 11/27/2022]
Abstract
We observed that treatment of prostate cancer cells for 24 h with wogonin, a naturally occurring monoflavonoid, induced cell death in a dose- and time-dependent manner. Exposure of wogonin to LNCaP cells was associated with increased intracellular levels of p21(Cip-1), p27(Kip-1), p53, and PUMA, oligomerization of Bax, release of cytochrome c from the mitochondria, and activation of caspases. We also confirmed the role of p53 by noting that knock-in in p53 expression by transfecting p53 DNA increased wogonin-induced apoptosis in p53-null PC-3 cells. To study the mechanism of PUMA up-regulation, we determined the activities of PUMA promoter in the wogonin treated and untreated cells. Increase of the intracellular levels of PUMA protein was due to increase in transcriptional activity. Data from chromatin immunoprecipitation (ChIP) analyses revealed that wogonin activated the transcription factor p53 binding activity to the PUMA promoter region. We observed that the up-regulation of PUMA mediated wogonin cytotoxicity. Further characterization of the transcriptional response to wogonin in HCT116 human colon cancer cells demonstrated that PUMA induction was p53-dependent; deficiency in either p53 or PUMA significantly protected HCT116 cells against wogonin-induced apoptosis. Also, wogonin promoted mitochondrial translocation and multimerization of Bax. Interestingly, wogonin (100 microM) treatment did not affect the viability of normal human prostate epithelial cells (PrEC). Taken together, these results indicate that p53-dependent transcriptional induction of PUMA and oligomerization of Bax play important roles in the sensitivity of cancer cells to apoptosis induced by caspase activation through wogonin.
Collapse
Affiliation(s)
- Dae-Hee Lee
- Departments of Surgery and Pharmacology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Clifford Kim
- Departments of Surgery and Pharmacology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Lin Zhang
- University of Pittsburgh Cancer Institute, Hillman Cancer Center, Departments of Pathology and Pharmacology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Yong J. Lee
- Departments of Surgery and Pharmacology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
189
|
Helton ES, Zhang J, Chen X. The proline-rich domain in p63 is necessary for the transcriptional and apoptosis-inducing activities of TAp63. Oncogene 2008; 27:2843-50. [PMID: 18037962 PMCID: PMC2662334 DOI: 10.1038/sj.onc.1210948] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Revised: 10/22/2007] [Accepted: 10/24/2007] [Indexed: 11/09/2022]
Abstract
p63 shares considerable sequence identity with p53, especially in its DNA-binding, activation and tetramerization domains. When the upstream promoter is used for p63 expression, three major transactivation p63 (TAp63) isoforms (alpha, beta and gamma) are produced. p63 is also expressed from an alternate promoter located in intron 3, producing three major DeltaNp63 isoforms. Recent studies demonstrated that p63 has the potential to function as a tumor suppressor or an oncoprotein. To further address this, we generated cell lines that inducibly express each TAp63 isoform. We showed that TAp63 isoforms are capable of inducing p53-responsive genes, inhibiting cell proliferation and promoting apoptosis. Interestingly, we discovered that both the activation domain (residues 1-59) and the proline-rich domain (residues 67-127) are required for TAp63 transcriptional activity. Likewise, TAp63beta(DeltaPRD), deleted of residues 60-133, possessed a greatly attenuated ability to induce endogenous target genes and promote apoptosis, but retained the ability to inhibit cell proliferation when expressed in stable, inducible cell lines. TAp63beta(DeltaPRD) also functioned as a dominant negative to wild-type p63beta in a dose-dependent manner. Furthermore, the loss of function seen with deletion of the proline-rich domain was not due to a DNA-binding defect, as TAp63beta(DeltaPRD) was found to strongly bind endogenous promoters using chromatin immunoprecipitation assay. Finally, mutational analysis revealed that a PXXP motif at residues 124-127 contributes to the transcriptional activity of TAp63. Altogether, our findings suggest that TAp63 transcriptional activity can be regulated by modification(s) of, or protein interactions with, the p63 proline-rich domain.
Collapse
Affiliation(s)
- E S Helton
- Department of Surgical and Radiological Sciences, Center for Comparative Oncology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | | | | |
Collapse
|
190
|
Sugihara T, Murano H, Tanaka K, Oghiso Y. Inverse dose-rate-effects on the expressions of extra-cellular matrix-related genes in low-dose-rate gamma-ray irradiated murine cells. JOURNAL OF RADIATION RESEARCH 2008; 49:231-40. [PMID: 18285661 DOI: 10.1269/jrr.07074] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Based on the results of previous microarray analyses of murine NIH3T3/PG13Luc cells irradiated with continuous low-dose-rate (LDR) gamma-ray or end-high-dose-rate-irradiations (end-HDR) at the end of the LDR-irradiation period, the inverse dose-rate-effects on gene expression levels were observed. To compare differences of the effects between LDR-irradiation and HDR-irradiation, HDR-irradiations at 2 different times, one (ini-HDR) at the same time at the start of LDR-irradiation and the other (end-HDR), were performed. The up-regulated genes were classified into two types, in which one was up-regulated in LDR-, ini-HDR-, and end-HDR irradiation such as Cdkn1a and Ccng1, which were reported as p53-dependent genes, and the other was up-regulated in LDR- and ini-HDR irradiations such as pro-collagen TypeIa2/Col1a2, TenascinC/Tnc, and Fibulin5/Fbln5, which were reported as extra-cellular matrix-related (ECM) genes. The time dependent gene expression patterns in LDR-irradiation were also classified into two types, in which one was an early response such as in Cdkn1a and Ccng1 and the other was a delayed response such as the ECM genes which have no linearity to total dose. The protein expression pattern of Cdkn1a increased dose dependently in LDR- and end-HDR-irradiations, but those of p53Ser15/18 and MDM2 in LDR-irradiations were different from end-HDR-irradiations. Furthermore, the gene expression levels of the ECM genes in embryonic fibroblasts from p53-deficient mice were not increased by LDR- and end-HDR-irradiation, so the delayed expressions of the ECM genes seem to be regulated by p53. Consequently, the inverse dose-rate-effects on the expression levels of the ECM genes in LDR- and end-HDR-irradiations may be explained from different time responses by p53 status.
Collapse
Affiliation(s)
- Takashi Sugihara
- Department of Radiobiology, Institute for Environmental Sciences, Kamikita, Aomori, Japan.
| | | | | | | |
Collapse
|
191
|
Abstract
Cell growth arrest and apoptosis are two best-known biological functions of tumor-suppressor p53. However, genetic evidence indicates that not only is p21 the major mediator of G(1) arrest, but also it can prevent apoptosis with an unknown mechanism. Here, we report the discovery of a p53 target gene dubbed killin, which lies in close proximity to pten on human chromosome 10 and encodes a 20-kDa nuclear protein. We show that Killin is not only necessary but also sufficient for p53-induced apoptosis. Genetic and biochemical analysis demonstrates that Killin is a high-affinity DNA-binding protein, which potently inhibits eukaryotic DNA synthesis in vitro and appears to trigger S phase arrest before apoptosis in vivo. The DNA-binding domain essential for DNA synthesis inhibition was mapped to within 42 amino acid residues near the N terminus of Killin. These results support Killin as a missing link between p53 activation and S phase checkpoint control designed to eliminate replicating precancerous cells, should they escape G(1) blockade mediated by p21.
Collapse
|
192
|
The contribution of transactivation subdomains 1 and 2 to p53-induced gene expression is heterogeneous but not subdomain-specific. Neoplasia 2008; 9:1057-65. [PMID: 18084613 DOI: 10.1593/neo.07688] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 09/22/2007] [Accepted: 09/25/2007] [Indexed: 12/14/2022] Open
Abstract
Two adjacent regions within the transactivation domain of p53 are sufficient to support sequence-specific transactivation when fused to a heterologous DNA binding domain. It has been hypothesized that these two subdomains of p53 may contribute to the expression of distinct p53-responsive genes. Here we have used oligonucleotide microarrays to identify transcripts induced by variants of p53 with point mutations within subdomains 1, 2, or 1 and 2 (QS1, QS2, and QS1/QS2, respectively). The expression of 254 transcripts was increased in response to wild-type p53 expression but most of these transcripts were poorly induced by these variants of p53. Strikingly, a number of known p53-regulated transcripts including TNFRSF10B, BAX, BTG2, and POLH were increased to wild-type levels by p53(QS1) and p53(QS2) but not p53(QS1/QS2), indicating that either subdomain 1 or 2 is sufficient for p53-dependent expression of a small subset of p53-responsive genes. Unexpectedly, there was no evidence for p53(QS1)- or p53(QS2)-specific gene expression. Taken together, we found heterogeneity in the requirement for transactivation subdomains 1 and 2 of p53 without any subdomain-specific contribution to p53-induced gene expression.
Collapse
|
193
|
Hu W, Feng Z, Teresky AK, Levine AJ. p53 regulates maternal reproduction through LIF. Nature 2008; 450:721-4. [PMID: 18046411 DOI: 10.1038/nature05993] [Citation(s) in RCA: 348] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Accepted: 10/10/2007] [Indexed: 12/11/2022]
Abstract
Extensive studies have shown that p53 is important in tumour prevention. However, little is known about its normal physiological function. Here we show that p53 is important in reproduction, in a gender-specific manner. Significant decreases in embryonic implantation, pregnancy rate and litter size were observed in matings with p53-/- female mice but not with p53-/- male mice. The gene encoding leukaemia inhibitory factor (LIF), a cytokine critical for implantation, was identified as a p53-regulated gene that functions as the downstream mediator of this effect. p53 can regulate both basal and inducible transcription of LIF. Loss of p53 decreased both the level and function of LIF in uteri. Lower LIF levels were observed in the uteri of p53-/- mice than in those of p53+/+ mice, particularly at day 4 of pregnancy, when transiently induced high levels of LIF were crucial for embryonic implantation. This observation probably accounts for the impaired implantation of embryos in p53-/- female mice. Administration of LIF to pregnant p53-/- mice restored maternal reproduction by improving implantation. These results demonstrate a function for p53 in maternal reproduction through the regulation of LIF. Evidence is accumulating that p53 may have a similar function in humans.
Collapse
Affiliation(s)
- Wenwei Hu
- Cancer Institute of New Jersey, University of Medicine and Dentistry of New Jersey, New Brunswick, New Jersey 08903, USA
| | | | | | | |
Collapse
|
194
|
Leu JIJ, George DL. Hepatic IGFBP1 is a prosurvival factor that binds to BAK, protects the liver from apoptosis, and antagonizes the proapoptotic actions of p53 at mitochondria. Genes Dev 2008; 21:3095-109. [PMID: 18056423 DOI: 10.1101/gad.1567107] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Liver is generally refractory to apoptosis induced by the p53 tumor suppressor protein, but the molecular basis remains poorly understood. Here we show that p53 transcriptional activation leads to enhanced expression of hepatic IGFBP1 (insulin-like growth factor-binding protein-1). Exhibiting a previously unanticipated role, a portion of intracellular IGFBP1 protein localizes to mitochondria where it binds to the proapoptotic protein BAK and hinders BAK activation and apoptosis induction. Interestingly, in many cells and tissues p53 also has a direct apoptotic function at mitochondria that includes BAK binding and activation. When IGFBP1 is in a complex with BAK, formation of a proapoptotic p53/BAK complex and apoptosis induction are impaired, both in cultured cells and in liver. In contrast, livers of IGFBP1-deficient mice exhibit spontaneous apoptosis that is accompanied by p53 mitochondrial accumulation and evidence of BAK oligomerization. These data support the importance of BAK as a mediator of p53's mitochondrial function. The results also identify IGFBP1 as a negative regulator of the BAK-dependent pathway of apoptosis, whose expression integrates the transcriptional and mitochondrial functions of the p53 tumor suppressor protein.
Collapse
Affiliation(s)
- J I-Ju Leu
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
195
|
Adaptor Protein LAPF Recruits Phosphorylated p53 to Lysosomes and Triggers Lysosomal Destabilization in Apoptosis. Cancer Res 2007; 67:11176-85. [DOI: 10.1158/0008-5472.can-07-2333] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
196
|
Tanaka T, Ohkubo S, Tatsuno I, Prives C. hCAS/CSE1L associates with chromatin and regulates expression of select p53 target genes. Cell 2007; 130:638-50. [PMID: 17719542 DOI: 10.1016/j.cell.2007.08.001] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2006] [Revised: 03/23/2007] [Accepted: 08/01/2007] [Indexed: 12/21/2022]
Abstract
The p53 tumor suppressor protein regulates many genes that can determine different cellular outcomes such as growth arrest or cell death. Promoter-selective transactivation by p53, although critical for the different cellular outcomes, is not well understood. We report here that the human cellular apoptosis susceptibility protein (hCAS/CSE1L) associates with a subset of p53 target promoters, including PIG3, in a p53-autonomous manner. Downregulation of hCAS/CSE1L decreases transcription from those p53 target promoters to which it preferentially binds and reduces apoptosis. In addition, hCAS/CSE1L silencing leads to increased methylation of histone H3 lysine 27 within the PIG3 gene. hCAS/CSE1L was previously shown to function as a nucleo-cytoplasmic transport factor, as does its closely related yeast homologue Cse1, which can also associate with chromatin and serve as a barrier protein that prevents spreading of heterochromatin. Thus, human CAS/CSE1L can bind select genes with significant functional consequences for p53-mediated transcription and determine cellular outcome.
Collapse
Affiliation(s)
- Tomoaki Tanaka
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | | | | |
Collapse
|
197
|
Lo Nigro C, Arnolfo E, Taricco E, Fruttero A, Russi EG, Lucio F, Ribero S, Comino A, Merlano M, Ungari S. The cisplatin-irradiation combination suggests that apoptosis is not a major determinant of clonogenic death. Anticancer Drugs 2007; 18:659-67. [PMID: 17762394 DOI: 10.1097/cad.0b013e328087388f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
It is commonly believed that tumor cells treated with anticancer agents, chemotherapy and/or radiation, die by apoptosis and that tumors which do not undergo apoptosis are resistant to treatment. In this study, we investigated the molecular basis underlying cisplatin cytotoxicity in the murine teratocarcinoma F9 cell line to see whether irradiation enhances cisplatin-induced cytotoxicity. We compared the apoptosis induced by chemo and/or radiotherapy with other cellular effects such as cell survival, clonogenic capability, cell cycle perturbation, expression of p53 and p53-related mRNAs, and necrosis. When combined with radiation, a clear additive cytotoxic effect of cisplatin was demonstrated. We found that both cisplatin and radiation induced cell death, but the level of induced apoptosis was low and there was no correlation with the results of the clonogenic assays: we noted a difference between cytotoxic effects in the clonogenic assay and the extent of apoptosis by fluorescence-activated cell sorter analysis, suggesting that cell killing reflected not only apoptosis but also cell cycle arrest, and that apoptosis, cell kinetics and clonogenicity suppression were independent processes.
Collapse
Affiliation(s)
- Cristiana Lo Nigro
- Molecular Biology Unit, Azienda Ospedaliera S. Croce e Carle, Cuneo, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Nakano H, Yonekawa H, Shinohara K. Threshold Level of p53 Required for the Induction of Apoptosis in X-Irradiated MOLT-4 Cells. Int J Radiat Oncol Biol Phys 2007; 68:883-91. [PMID: 17544001 DOI: 10.1016/j.ijrobp.2007.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 03/06/2007] [Accepted: 03/07/2007] [Indexed: 12/20/2022]
Abstract
PURPOSE To determine the threshold level for the initiation of apoptosis by studying the quantitative aspect of p53 response to DNA damage in individual cells, to better understand the process in X-ray-induced p53-dependent apoptosis. METHODS AND MATERIALS Time-sequential changes in p53 protein level were obtained for X-irradiated MOLT-4 cells using flow cytometry and analyzed. RESULTS The changes in the cellular frequency distribution pattern of p53 content could be divided into two parts at a certain p53 level. The p53 vs. side-scatter in flow cytometry showed the sequential changes of p53 increase followed by an increase in cell death. On the basis of these results we determined a threshold level of p53 for the initiation of apoptosis. The level was estimated to be (1.08 +/- 0.05) x 10(5) molecules per cell, which was approximately threefold higher than the mean content of control cells. The minimum times for p53 level to reach this threshold level were independent of X-ray dose and 1.4-1.6 h. The times for the signal transduction from the p53 accumulation to disruption of the mitochondrial membrane potential, caspase-3 activation, and cell death were 1.6, 2.1, and 2.8 h, respectively. CONCLUSIONS The threshold level of p53 for the initiation of apoptosis and the time sequence in the course of apoptotic events were determined in X-irradiated MOLT-4 cells.
Collapse
Affiliation(s)
- Hisako Nakano
- Department of Laboratory Animal Science, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | | | | |
Collapse
|
199
|
Benítez JA, Arregui L, Vergara P, Segovia J. Targeted-simultaneous expression of Gas1 and p53 using a bicistronic adenoviral vector in gliomas. Cancer Gene Ther 2007; 14:836-46. [PMID: 17599090 DOI: 10.1038/sj.cgt.7701076] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The targeted expression of transgenes is one of the principal goals of gene therapy, and it is particularly relevant for the treatment of brain tumors. In this study, we examined the effect of the overexpression of human gas1 (growth arrest specific 1) and human p53 cDNAs, both under the transcriptional control of a promoter of the human glial fibrillary acidic protein (gfa2), employing adenoviral expression vectors, in glioma cells. We showed that the targeted overexpression of gas1 and p53 (AdSGas1 and AdSp53, respectively) in rat glioma cells (C6) reduced the number of viable cells and induced apoptosis. Moreover, the adenovirally targeted expression of these genes also reduced tumor growth in vivo. Unexpectedly, there was no additive effect when both gas1 and p53 were simultaneously expressed in the same cells using a bicistronic adenoviral vector. We suggest that Gas1 does not act in combination with p53 in the C6 and U373 glioma cell lines, inducing apoptosis and cell cycle arrest. Our results indicate that the targeted expression of tumor suppressor genes (gas1 and p53) regulated by the gfa2 promoter, together with adenoviral vectors may provide an interesting approach for adjuvant selective glioma gene therapy.
Collapse
Affiliation(s)
- J A Benítez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, México DF, México
| | | | | | | |
Collapse
|
200
|
Pagano A, Métrailler-Ruchonnet I, Aurrand-Lions M, Lucattelli M, Donati Y, Argiroffo CB. Poly(ADP-ribose) polymerase-1 (PARP-1) controls lung cell proliferation and repair after hyperoxia-induced lung damage. Am J Physiol Lung Cell Mol Physiol 2007; 293:L619-29. [PMID: 17575013 DOI: 10.1152/ajplung.00037.2007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Oxygen-based therapies expose lung to elevated levels of ROS and induce lung cell damage and inflammation. Injured cells are replaced through increased proliferation and differentiation of epithelial cells and fibroblasts. Failure to modulate these processes leads to excessive cell proliferation, collagen deposition, fibrosis, and chronic lung disease. Poly(ADP-ribose) polymerase-1 (PARP-1) is activated in response to DNA damage and participates in DNA repair, genomic integrity, and cell death. In this study, we evaluated the role of PARP-1 in lung repair during recovery after acute hyperoxia exposure. We exposed PARP-1 -/- and wild-type mice for 64 h to 100% hyperoxia and let them recover in air for 5-21 days. PARP-1-deficient mice exhibited significantly higher lung cell hyperplasia and proliferation than PARP-1 +/+ animals after 5 and 10 days of recovery. This was accompanied by an increased inflammatory response in PARP-1 -/- compared with wild-type animals, characterized by neutrophil infiltration and increased IL-6 levels in bronchoalveolar lavages. These lesions were reversible, since the extent of the hyperplastic regions was reduced after 21 days of recovery and did not result in fibrosis. In vitro, lung primary fibroblasts derived from PARP-1 -/- mice showed a higher proliferative response than PARP-1 +/+ cells during air recovery after hyperoxia-induced growth arrest. Altogether, these results reveal an essential role of PARP-1 in the control of cell repair and tissue remodeling after hyperoxia-induced lung injury.
Collapse
Affiliation(s)
- Alessandra Pagano
- Department of Pathology-Immunology, Medical School, University of Geneva, Switzerland.
| | | | | | | | | | | |
Collapse
|