301
|
Abstract
Mutations that drive uncontrolled cell-cycle progression are requisite events in tumorigenesis. But evolution has installed in the proliferative programmes of mammalian cells a variety of innate tumour-suppressive mechanisms that trigger apoptosis or senescence, should proliferation become aberrant. These contingent processes rely on a series of sensors and transducers that act in a coordinated network to target the machinery responsible for apoptosis and cell-cycle arrest at different points. Although oncogenic mutations that disable such networks can have profound and varied effects on tumour evolution, they may leave intact latent tumour-suppressive potential that can be harnessed therapeutically.
Collapse
Affiliation(s)
- Scott W Lowe
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, New York 11724, USA.
| | | | | |
Collapse
|
302
|
Scian MJ, Stagliano KER, Ellis MA, Hassan S, Bowman M, Miles MF, Deb SP, Deb S. Modulation of gene expression by tumor-derived p53 mutants. Cancer Res 2004; 64:7447-54. [PMID: 15492269 DOI: 10.1158/0008-5472.can-04-1568] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
p53 mutants with a single amino acid substitution are overexpressed in a majority of human cancers containing a p53 mutation. Overexpression of the mutant protein suggests that there is a selection pressure on the cell indicative of an active functional role for mutant p53. Indeed, H1299 cells expressing mutant p53-R175H, p53-R273H or p53-D281G grow at a faster rate compared with a control cell line. Using p53-specific small interfering RNA, we show that the growth rate of mutant p53-expressing cells decreases as mutant p53 level decreases, demonstrating that the increased cellular growth is dependent on p53 expression. Increased growth rate is not observed for H1299 cell clones expressing mutant p53-D281G (L22Q/W23S), which has been shown to be defective in transactivation in transient transcriptional assays. This shows that the increased growth rate imparted by mutant p53 in H1299 cells requires the transactivation function of mutant p53. By performing microarray hybridization analyses, we show that constitutive expression of three common p53 mutants (p53-R175H, p53-R273H, and p53-D281G) in H1299 human lung carcinoma cells evokes regulation of a common set of genes, a significant number of which are involved in cell growth regulation. Predictably, H1299 cells expressing p53-D281G (L22Q/W23S) are defective in up-regulating a number of these genes. The differences in expression profiles induced by individual p53 mutants in the cells may be representative of the p53 mutants and how they can affect gene expression resulting in the observed "gain of function" phenotypes (i.e., increased growth rate, decreased sensitivity to chemotherapeutic agents, and so forth).
Collapse
Affiliation(s)
- Mariano J Scian
- Department of Biochemistry, Massey Cancer Center, and Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | | | | | | | | | | | | |
Collapse
|
303
|
Bortoluzzi MC, Yurgel LS, Dekker NP, Jordan RCK, Regezi JA. Assessment of p63 expression in oral squamous cell carcinomas and dysplasias. ACTA ACUST UNITED AC 2004; 98:698-704. [PMID: 15583543 DOI: 10.1016/j.tripleo.2004.04.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES p63, a p53 homologue, may be associated with tumorigenesis in epithelial tissues through its inhibition of p53 transactivation functions. We sought to determine the pattern and levels of p63 expression in oral dysplasias and carcinomas using standard immunohistochemical staining. We also assessed and compared expression of p53 and a cell proliferation marker in these lesions. STUDY DESIGN This retrospective cross-sectional survey (n=67) included hyperkeratosis (10), mild dysplasia (9), moderate dysplasia (11), severe dysplasia/in situ carcinoma (10), squamous cell carcinoma (SCC) (22 [9 well differentiated, 7 moderately differentiated, 6 poor differentiated]), and normal mucosa (5). Serial sections were stained immunohistochemically with antibodies to p63 (4A4 recognizing all p63 isotypes), p53 (DO-7), and Ki-67 (MIB-1) proteins. In preinvasive lesions, both the percentage of positive cells and staining patterns (negative, basal, suprabasal) were assessed. In oral SCCs, the percentage of positive cells was assessed. Statistical analysis was done using the Tukey-Kramer multiple comparisons test. RESULTS A suprabasal p63 staining pattern was evident in keratinocyte nuclei in the entire range of noninvasive lesions studied, including normal mucosa. Most nuclei in invasive SCCs stained positive. When all grades of dysplasia were combined, the percent of p63 positive cells was significantly greater than hyperkeratosis (P < .01), and well-differentiated SCC (P < .001). Moderately differentiated SCC had statistically significant more positive cells than well-differentiated SSC (P < .01). Comparison of serial sections showed different p63 staining patterns compared to p53 or Ki-67 staining patterns. CONCLUSIONS We conclude that p63 is expressed in oral carcinomas and dysplasias, as determined by immunohistochemical staining with a primary antibody to all isotypes. Neither staining pattern nor percentage of stained cells could be used to differentiate the lesions studied. The statistically significant differences found between some groups are not likely to be of diagnostic value. p63 is not coexpressed with p53 expression or Ki-67 suggesting functional independence. When antibodies to the p63 isotypes become available, oral dysplasias and carcinomas should be reassessed.
Collapse
|
304
|
Abstract
Survivin regulates cell division and inhibits apoptosis by blocking caspase activation. The tumor suppressor p53 inhibits cell cycle progression and induces apoptosis. Since Survivin overexpression and loss of wild-type p53 expression/function occur in most cancers, we investigated whether Survivin regulates p53. Stable overexpression of Survivin protects BaF3 cells from Adriamycin-induced apoptosis, while dominant-negative (T34A) and antisense (AS) Survivin accelerate apoptosis. In BaF3 cells and transiently transfected MCF7 breast cancer cells, elevation of total and phospho-Ser15-p53 in response to Adriamycin is blocked by Survivin and enhanced by Survivin disruption. Furthermore, in Adriamycin-treated MCF7 cells, ectopic Survivin decreased p53 mRNA and increased mRNA and protein of the p53 homologues DeltaNp63 and TAp73 and mRNA for DeltaNp73, suggesting that Survivin may differentially regulate p53 family transcription. Concomitant with decreasing p53 mRNA, Survivin decreased Mdm2 mRNA. Survivin disruption by T34A or AS Survivin resulted in reduced Mdm2 protein. The caspase inhibitor, Z-VAD-FMK, blocked the decrease in Mdm2 as well as the increase in p53 resulting from Survivin disruption, indicating that Survivin regulates Mdm2 at the post-translational level. Proteosome inhibition confirmed that reduced p53 protein observed in cells overexpressing Survivin is due to enhanced p53 degradation resulting from Survivin-mediated inhibition of Mdm2 cleavage by caspases. In summary, our results identify regulatory interactions between Survivin and p53 at the mRNA and protein levels, and suggest that the p53 homologues DeltaNp63, TAp73 and DeltaNp73 may also be regulated by Survivin.
Collapse
Affiliation(s)
- Zhanxiang Wang
- Department of Microbiology and Immunology and The Walther Oncology Center, Indiana University School of Medicine, 950 West Walnut Street, Indianapolis, IN 46202, USA
| | | | | |
Collapse
|
305
|
Chen CF, Lohnes D. Dominant-negative retinoic acid receptors elicit epidermal defects through a non-canonical pathway. J Biol Chem 2004; 280:3012-21. [PMID: 15528198 DOI: 10.1074/jbc.m411522200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Previous work has shown that a dominant-negative retinoic acid receptor alpha (dnRARalpha), expressed under the K14 promoter, causes severe epidermal defects. Similar defects are, however, not seen in RARalphagamma double null mutant mice, which lack the entire complement of RARs expressed in the epidermis. To investigate the mechanism of action of these dominant-negative receptors, dnRARalpha or a DNA binding-deficient variant, dnRARalpha(DBD), were targeted to the basal epidermis. Expression of either receptor type led to similar epidermal phenotypes suggesting that both RAR mutants acted through a common mechanism. The epidermal phenotype was reminiscent of defects seen in p63(-/-) mice. Consistent with this, reduced p63 expression was observed in transgenic offspring expressing either RAR mutant, suggesting that down-regulation of p63 might underlie the effects of these receptors on epidermal development. By contrast, expression of p63 in the epidermis of RARalphagamma(-/-) offspring was unaffected, indicating that RARs were not essential for p63 expression. These findings suggest that dnRARs may impact on epidermal development through one or more non-canonical pathways, which are independent of receptor-DNA interaction.
Collapse
Affiliation(s)
- Chang Feng Chen
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal K1H 8M5, Canada
| | | |
Collapse
|
306
|
Dupont S, Zacchigna L, Adorno M, Soligo S, Volpin D, Piccolo S, Cordenonsi M. Convergence of p53 and TGF-beta signaling networks. Cancer Lett 2004; 213:129-38. [PMID: 15327827 DOI: 10.1016/j.canlet.2004.06.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2004] [Accepted: 06/03/2004] [Indexed: 11/21/2022]
Abstract
p53 is a protein with many talents. One of the most fundamental is the ability to act as essential growth checkpoint that protects cells against cellular transformation. p53 does so through the induction of genes leading to growth arrest or apoptosis. Most of the studies focusing on the mechanisms of p53 activity have been performed in cultured cells upon treatment with well-established p53-activating inputs, such as high doses of radiations, DNA-damaging drugs and activated oncogenes. However, how the tumor suppressive functions of p53 become concerted with the extracellular cues arriving at the cell surface during tissue homeostasis, remains largely unknown. Intriguingly, two recent papers have shed new light into this unexplored field, indicating that p53 plays a key role in TGF-beta-induced growth arrest and, unexpectedly, in the developmental effects of TGF-beta in early embryos. Here we review and comment on these findings and on their implications for cancer biology.
Collapse
Affiliation(s)
- Sirio Dupont
- Department of Histology Microbiology and Medical iotechnologies, Section of Histology and Embryology, University of Padua, viale Colombo 3, 35121, Italy
| | | | | | | | | | | | | |
Collapse
|
307
|
Calabrò V, Mansueto G, Santoro R, Gentilella A, Pollice A, Ghioni P, Guerrini L, La Mantia G. Inhibition of p63 transcriptional activity by p14ARF: functional and physical link between human ARF tumor suppressor and a member of the p53 family. Mol Cell Biol 2004; 24:8529-40. [PMID: 15367673 PMCID: PMC516740 DOI: 10.1128/mcb.24.19.8529-8540.2004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The ARF/MDM2/p53 pathway is a principal defense mechanism to protect the organism from uncontrolled effects of deregulated oncogenes. Oncogenes activate ARF, which interacts with and inhibits the ubiquitin ligase MDM2, resulting in p53 stabilization and activation. Once stabilized and activated, p53 can either induce or repress a wide array of different gene targets, which in turn can regulate cell cycle, DNA repair, and a number of apoptosis-related genes. Here we show that, unlike p53, p63, a member of the p53 family, directly interacts with p14(ARF). Through this interaction ARF inhibits p63-mediated transactivation and transrepression. In p63-transfected cells, ARF, which normally localizes into nucleoli, accumulates in the nucleoplasm. Based on these observations, we suggest that stimuli inducing p14(ARF) expression can, at the same time, activate p53 and impair p63 transcriptional activity, altering the pattern of p53 target gene expression. Here we show, for the first time, a physical and functional link between the p14(ARF) tumor suppressor protein and p63, a member of the p53 family.
Collapse
Affiliation(s)
- Viola Calabrò
- Department of Genetics, General and Molecular Biology, University of Naples Federico II, Via Mezzocannone, 8, 80134 Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
308
|
Kurata SI, Okuyama T, Osada M, Watanabe T, Tomimori Y, Sato S, Iwai A, Tsuji T, Ikawa Y, Katoh I. p51/p63 Controls Subunit α3 of the Major Epidermis Integrin Anchoring the Stem Cells to the Niche. J Biol Chem 2004; 279:50069-77. [PMID: 15361520 DOI: 10.1074/jbc.m406322200] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
p51/p63, a member of the tumor suppressor p53 gene family, is crucial for skin development. We describe here identification of ITGA3 encoding integrin alpha(3) as a target of its trans-activating function, proposing that p51/p63 allows epidermal stem cells to express laminin receptor alpha(3)beta(1) for anchorage to the basement membrane. When activated by genotoxic stress or overexpressed ectopically in non-adherent cells, p51/p63 transduced a phenotype to attach to extracellular matrices, which was accompanied by expression of ITGA3. Motifs matching the p53-binding consensus sequence were located in a scattered form in intron 1 of human ITGA3, and served as p51/p63-responsive elements in reporter assays. In addition to the trans-activating ability of the TA isoform, we detected a positive effect of the DeltaN isoform on ITGA3. The high level alpha(3) production in human keratinocyte stem cells diminished upon elimination of p51/p63 by small interfering RNA or by Ca(2+)-induced differentiation. Furthermore, a chromatin immunoprecipitation experiment indicated a physical interaction of p51/p63 with intron 1 of ITGA3. This study provides a molecular basis for the standing hypothesis that p51/p63 is essential for epidermal-mesenchymal interactions.
Collapse
Affiliation(s)
- Shun-Ichi Kurata
- Department of Biochemical Genetics, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510 Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
309
|
Alvarez AR, Sandoval PC, Leal NR, Castro PU, Kosik KS. Activation of the neuronal c-Abl tyrosine kinase by amyloid-β-peptide and reactive oxygen species. Neurobiol Dis 2004; 17:326-36. [PMID: 15474370 DOI: 10.1016/j.nbd.2004.06.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2004] [Revised: 06/16/2004] [Accepted: 06/22/2004] [Indexed: 11/24/2022] Open
Abstract
The deposition and accumulation of amyloid-beta-peptide (Abeta) in the brain are considered a sine qua non for Alzheimer's disease. The experimental delivery of fibrilized Abeta serves as a cellular model for several facets of the disease including the induction of synaptic dysfunction and apoptosis. c-Abl kinase is involved in the regulation of apoptosis and its pro-apoptotic function is in part mediated by its interaction with p73, a p53 homologue. We found that c-Abl activation is involved in cell signals that regulate neuronal death response to Abeta fibrils. Abeta peptide fibrils induced an increase of the c-Abl activity in rat hippocampal neurons as well as an increase in nuclear p73 protein levels and the p73-c-Abl complex. The neuronal cell death induced by Abeta fibrils was prevented by the inhibition of c-Abl with imatinib mesylate (Gleevec or STI571) and by the inhibition c-Abl expression by RNAi. These results directly point to a therapeutic strategy for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Alejandra R Alvarez
- FONDAP Center for Cell and Molecular Biology Joaquin V. Luco, P. Universidad Católica de Chile, Santiago, 114-D, Chile.
| | | | | | | | | |
Collapse
|
310
|
Yamamoto Y, Oelgeschläger M. Regulation of bone morphogenetic proteins in early embryonic development. Naturwissenschaften 2004; 91:519-34. [PMID: 15517134 DOI: 10.1007/s00114-004-0575-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Bone morphogenetic proteins (BMPs), a large subgroup of the TGF-beta family of secreted growth factors, control fundamental events in early embryonic development, organogenesis and adult tissue homeostasis. The plethora of dose-dependent cellular processes regulated by BMP signalling demand a tight regulation of BMP activity. Over the last decade, a number of proteins have been identified that bind BMPs in the extracellular space and regulate the interaction of BMPs with their cognate receptors, including the secreted BMP antagonist Chordin. In the early vertebrate embryo, the localized secretion of BMP antagonists from the dorsal blastopore lip establishes a functional BMP signalling gradient that is required for the determination of the dorsoventral - or back to belly - body axis. In particular, inhibition of BMP activity is essential for the formation of neural tissue in the development of vertebrate and invertebrate embryos. Here we review recent studies that have provided new insight into the regulation of BMP signalling in the extracellular space. In particular, we discuss the recently identified Twisted gastrulation protein that modulates, in concert with metalloproteinases of the Tolloid family, the interaction of Chordin with BMP and a family of proteins that share structural similarities with Chordin in the respective BMP binding domains. In addition, genetic and functional studies in zebrafish and frog provide compelling evidence that the secreted protein Sizzled functionally interacts with the Chd-BMP pathway, despite being expressed ventrally in the early gastrula-stage embryo. These intriguing discoveries may have important implications, not only for our current concept of early embryonic patterning, but also for the regulation of BMP activity at later developmental stages and tissue homeostasis in the adult.
Collapse
Affiliation(s)
- Yukiyo Yamamoto
- Department of Developmental Biology, Max-Planck Institute of Immunobiology, Stübeweg 51, 79108, Freiburg, Germany
| | | |
Collapse
|
311
|
Voeltzel T, Morel AP, Rostan MC, Ji J, Chiodino C, Ponchel F, Vigouroux J, Caron de Fromentel C, Soussi T, Ozturk M. A New Set of Monoclonal Antibodies Directed to Proline-Rich and Central Regions of p53. ACTA ACUST UNITED AC 2004; 23:287-92. [PMID: 15672606 DOI: 10.1089/hyb.2004.23.287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The p53 protein can adopt several conformations in cells--"latent," "active," or mutant--depending on cellular stress or mutations of the TP53 gene. Today, only a few antibodies discriminating these conformations are available. We produced three new anti-p53 monoclonal antibodies (MAbs) directed against epitopes of human p53. The H53C1 MAb recognizes an epitope located at the N-terminal part of the central region of p53 and can discriminate mutant from wild-type conformation. The H53C2 and H53C3 MAbs are against different epitopes within the proline-rich region of p53. Moreover, the H53C2 epitope is located in the second negative regulatory domain of p53 between residues 80 and 93. These MAbs can be used as new tools to study and modulate the cellular functions of p53.
Collapse
Affiliation(s)
- T Voeltzel
- INSERM U590, Centre Léon Bérard, Lyon, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
312
|
Abstract
Expression of p63, a transcription factor that is transcribed into six isoforms, is required for proper development of stratified epithelia, such as the epidermis. In the absence of p63, epithelia remain single-layered. The molecular role of p63 in development and differentiation of stratified epithelia, however, remains controversial. Based on recent studies, we now believe that p63 has a dual role and is essential for development as well as maintenance of the epidermis. During embryogenesis, p63 may be the molecular switch required for initiation of epithelial stratification. This is based on our recent data demonstrating that ectopic expression of a p63 isoform in single-layered epithelia results in the induction of a stratification program. Furthermore, in the mature epidermis, p63 may maintain the proliferative potential of basal keratinocytes. This is suggested by the observation that p63 is primarily expressed in the basal compartment of the epidermis, that p63 expression induces hyperproliferation, and that its expression needs to be downregulated for terminal differentiation to take place. In this review, we discuss recent evidence supporting this dual role for p63 and place it in the context of our increasing knowledge of epidermal development and differentiation.
Collapse
Affiliation(s)
- Maranke I Koster
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
313
|
Honeycutt KA, Koster MI, Roop DR. Genes involved in stem cell fate decisions and commitment to differentiation play a role in skin disease. J Investig Dermatol Symp Proc 2004; 9:261-8. [PMID: 15369222 DOI: 10.1111/j.1087-0024.2004.09312.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
Multipotent stem cells residing in the bulge region of the hair follicle give rise to cells of different fates including those forming hair follicles, interfollicular epidermis, and associated glands. Stem cell fate determination is regulated by genes involved in both proliferation and differentiation, which are tightly regulated processes. Understanding the molecular mechanisms by which proliferation and differentiation are regulated will provide useful insight into treating human diseases caused by the deregulation of these processes. Two genes involved in regulating proliferation and differentiation are c-Myc and p63, both of which have been found to be deregulated/mutated in several human diseases. Accelerating proliferation leads to neoplastic human diseases and deregulated c-Myc has been implicated in a variety of cancers. Evidence indicates that c-Myc also diverts stem cells to an epidermal and sebaceous gland fate at the expense of the hair follicle fate. Therefore, deregulation of c-Myc has the potential to not only accelerate tumorigenesis, but also influence skin tumor phenotype. In addition, the inhibition of differentiation may also predispose to the development of skin cancer. Recent evidence suggests that the transcription factor p63, is not only responsible for the initiation of an epithelial stratification program during development, but also the maintenance of the proliferative potential of basal keratinocytes in mature epidermis. Mutations in the p63 gene have been shown to cause ectodermal dysplasias and deregulated expression of p63 has been observed in squamous cell carcinomas. In this review, we will discuss recent data implicating a role for both c-Myc and p63 in human skin diseases.
Collapse
|
314
|
Shih IM, Kurman RJ. p63 Expression Is Useful in the Distinction of Epithelioid Trophoblastic and Placental Site Trophoblastic Tumors by Profiling Trophoblastic Subpopulations. Am J Surg Pathol 2004; 28:1177-83. [PMID: 15316317 DOI: 10.1097/01.pas.0000130325.66448.a1] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Human trophoblast is composed of a heterogeneous population of cells, which give rise to a variety of trophoblastic tumors and tumor-like lesions. In this report, we analyzed the expression pattern of the p63 gene, a transcription factor belonging to the p53 family, in different trophoblastic subpopulations and in trophoblastic lesions. p63 has various isoforms that are classified into two groups designated TA and DeltaNp63 isoforms. The TA isoforms have a p53-like suppressor function, whereas the DeltaNp63 isoforms exert an oncogenic effect. Based on immunohistochemistry and RT-PCR, it appears that cytotrophoblast expresses the DeltaNp63 isoform whereas chorionic-type intermediate trophoblast in the fetal membranes, placental site nodules, and epithelioid trophoblastic tumors expresses the TAp63 isoform. Intermediate trophoblast in the implantation site and placental site trophoblastic tumors does not express p63. Based on the expression patterns of p63 and the previously described expression patterns of other trophoblastic markers, including HLA-G, cytokeratin 18, hPL, and Ki-67, we developed an immunohistochemical algorithm to diagnose trophoblastic lesions. A validation set of 22 trophoblastic lesions and 34 nontrophoblastic tumors were classified correctly using this algorithm. In conclusion, the findings in this study demonstrate that different trophoblastic subpopulations and their related trophoblastic lesions are characterized by distinctive patterns of p63 expression. Recognizing these distinctive expression patterns helps to further elucidate the biology of trophoblast and can also provide a useful tool for the differential diagnosis of trophoblastic lesions.
Collapse
Affiliation(s)
- I Ming Shih
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD 21231, USA.
| | | |
Collapse
|
315
|
Bernassola F, Salomoni P, Oberst A, Di Como CJ, Pagano M, Melino G, Pandolfi PP. Ubiquitin-dependent degradation of p73 is inhibited by PML. ACTA ACUST UNITED AC 2004; 199:1545-57. [PMID: 15184504 PMCID: PMC2211783 DOI: 10.1084/jem.20031943] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
p73 has been identified recently as a structural and functional homologue of the tumor suppressor p53. Here, we report that p73 stability is directly regulated by the ubiquitin–proteasome pathway. Furthermore, we show that the promyelocytic leukemia (PML) protein modulates p73 half-life by inhibiting its degradation in a PML–nuclear body (NB)–dependent manner. p38 mitogen-activated protein kinase–mediated phosphorylation of p73 is required for p73 recruitment into the PML-NB and subsequent PML-dependent p73 stabilization. We find that p300-mediated acetylation of p73 protects it against ubiquitinylation and that PML regulates p73 stability by positively modulating its acetylation levels. As a result, PML potentiates p73 transcriptional and proapoptotic activities that are markedly impaired in Pml−/− primary cells. Our findings demonstrate that PML plays a crucial role in modulating p73 function, thus providing further insights on the molecular network for tumor suppression.
Collapse
Affiliation(s)
- Francesca Bernassola
- Molecular Biology Program, Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10021, USA
| | | | | | | | | | | | | |
Collapse
|
316
|
Daniely Y, Liao G, Dixon D, Linnoila RI, Lori A, Randell SH, Oren M, Jetten AM. Critical role of p63 in the development of a normal esophageal and tracheobronchial epithelium. Am J Physiol Cell Physiol 2004; 287:C171-81. [PMID: 15189821 DOI: 10.1152/ajpcell.00226.2003] [Citation(s) in RCA: 221] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The trachea and esophagus originate from the foregut endoderm during early embryonic development. Their epithelia undergo a series of changes involving the differentiation of stem cells into unique cell types and ultimately forming the mature epithelia. In this study, we monitored the expression of p63 in the esophagus and the trachea during development and examined in detail morphogenesis in p63(-/-) mice. At embryonic day 15.5 (E15.5), the esophageal and tracheobronchial epithelia contain two to three layers of cells; however, only the progenitor cells express p63. These progenitor cells differentiate first into ciliated cells (p63(-)/beta-tubulin IV(+)) and after birth into mature basal cells (p63(+)/K14(+)/K5(+)/BS-I-B4(+)). In the adult pseudostratified, columnar tracheal epithelium, K14(+)/K5(+)/BS-I-B4(+) basal cells stain most intensely for p63, whereas ciliated and mucosecretory cells are negative. In stratified squamous esophageal epithelium and during squamous metaplasia in the trachea, cells in the basal layer stain strongest for p63, whereas p63 staining declines progressively in transient amplifying and squamous differentiated cells. Generally, p63 expression is restricted to human squamous cell carcinomas, and adenocarcinomas and Barrett's metaplasia do not stain for p63. Examination of morphogenesis in newborn p63(-/-) mice showed an abnormal persistence of ciliated cells in the esophagus. Significantly, in both tissues, lack of p63 expression results in the development of a highly ordered, columnar ciliated epithelium deficient in basal cells. These observations indicate that p63 plays a critical role in the development of normal esophageal and tracheobronchial epithelia and appears to control the commitment of early stem cells into basal cell progeny and the maintenance of basal cells.
Collapse
Affiliation(s)
- Yaron Daniely
- Cell Biology Section, Division of Intramural Research, National Institute of Environmental Health Sciences, Triangle Park, NC 27709, USA
| | | | | | | | | | | | | | | |
Collapse
|
317
|
Finzer P, Krueger A, Stöhr M, Brenner D, Soto U, Kuntzen C, Krammer PH, Rösl F. HDAC inhibitors trigger apoptosis in HPV-positive cells by inducing the E2F-p73 pathway. Oncogene 2004; 23:4807-17. [PMID: 15077164 DOI: 10.1038/sj.onc.1207620] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Histone deacetylase (HDAC) inhibitors induce an intrinsic type of apoptosis in human papillomavirus (HPV)-positive cells by disrupting the mitochondrial transmembrane potential (deltapsim). Loss of deltapsim was only detected in E7, but not in E6 oncogene-expressing cells. HDAC inhibition led to a time-dependent degradation of the pocket proteins pRb, p107 and p130, releasing 'free' E2F-1 following initial G1 arrest. Inhibition of proteasomal proteolysis, but not of caspase activity rescued pRb from degradation and functionally restored its inhibitory effect on the cyclin E gene, known to be suppressed by pRb-E2F-1 in conjunction with HDAC1. Using siRNA targeted against p53, E2F-1 still triggered apoptosis by inducing the E2F-responsive proapoptotic alpha- and beta-isoforms of p73. These data may determine future therapeutic strategies in which HDAC inhibitors can effectively eliminate HPV-positive cells by an apoptotic route that does not rely on the reactivation of the 'classical' p53 pathway through a preceding shut-off of viral gene expression.
Collapse
Affiliation(s)
- Patrick Finzer
- Deutsches Krebsforschungszentrum, Forschungsschwerpunkt Angewandte Tumorvirologie, Abteilung Virale Transformationsmechanismen, Im Neuenheimer Feld 242, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
318
|
Abstract
While p53 family members have distinct nonoverlapping functions, the involvement of p63 and p73 in p53-mediated apoptosis is controversial. Results of a recent study indicate that at least in thymocytes, p53-dependent apoptosis occurs independently of p63 and p73.
Collapse
Affiliation(s)
- Samuel Benchimol
- Ontario Cancer Institute, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
319
|
Senoo M, Manis JP, Alt FW, McKeon F. p63 and p73 are not required for the development and p53-dependent apoptosis of T cells. Cancer Cell 2004; 6:85-9. [PMID: 15261144 DOI: 10.1016/j.ccr.2004.06.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2004] [Revised: 04/06/2004] [Accepted: 05/14/2004] [Indexed: 11/21/2022]
Abstract
The recent discoveries of p63 and p73, homologs of the tumor suppressor p53, raised the possibility of a network of these family members governing cell cycle arrest and apoptosis in response to stress. However, mice lacking p73 show no tendency for spontaneous tumors, and mutations in p63 or p73 are rare in human tumors, rendering any obligate role of these genes in cell death and tumor suppression unclear. In an effort to reconcile these incongruent data, we examined the genetic interactions between p53, p63, and p73 in well-established paradigms of p53-dependent and -independent T cell death using primary, genetically defined lymphocytes. Our findings challenge the generality of the notion that p63 and p73 are required for p53 function or for apoptosis in T cells.
Collapse
Affiliation(s)
- Makoto Senoo
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
320
|
Scian MJ, Stagliano KER, Deb D, Ellis MA, Carchman EH, Das A, Valerie K, Deb SP, Deb S. Tumor-derived p53 mutants induce oncogenesis by transactivating growth-promoting genes. Oncogene 2004; 23:4430-43. [PMID: 15077194 DOI: 10.1038/sj.onc.1207553] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We have studied the mechanism of mutant p53-mediated oncogenesis using several tumor-derived mutants. Using a colony formation assay, we found that the majority of the mutants increased the number of colonies formed compared to the vector. Expression of tumor-derived p53 mutants increases the rate of cell growth, suggesting that the p53 mutants have 'gain of function' properties. We have studied the gene expression profile of cells expressing tumor-derived p53-D281G to identify genes transactivated by mutant p53. We report the transactivation of two genes, asparagine synthetase and human telomerase reverse transcriptase. Quantitative real-time PCR confirms this upregulation. Transient transfection promoter assays verify that tumor-derived p53 mutants transactivate these promoters significantly. An electrophoretic mobility shift assay shows that tumor-derived p53-mutants cannot bind to the wild-type p53 consensus sequence. The results presented here provide some evidence of a possible mechanism for mutant p53-mediated transactivation.
Collapse
Affiliation(s)
- Mariano J Scian
- Department of Biochemistry and the Massey Cancer Center, Virginia Commonwealth University, PO Box 980614, Richmond, VA 23298, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
321
|
Obad S, Brunnström H, Vallon-Christersson J, Borg A, Drott K, Gullberg U. Staf50 is a novel p53 target gene conferring reduced clonogenic growth of leukemic U-937 cells. Oncogene 2004; 23:4050-9. [PMID: 15064739 DOI: 10.1038/sj.onc.1207524] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The tumor suppressor gene p53 is a transcription factor that mediates both cell cycle arrest and apoptosis. Interestingly, p53 also induces differentiation of a number of tissues, including leukemic cells. However, although p53-mediated differentiation of leukemic U-937 cells depends on the transcriptional activity of p53, a p53 target gene mediating differentiation has hitherto not been identified. To screen for novel p53 target genes in leukemic cells, a cDNA microarray analysis was performed with U-937-4/ptsp53 cells, expressing a temperature-sensitive p53 mutant. We report that transcription of the Staf50 (stimulated transacting factor of 50 kDa) gene is upregulated in response to wild-type p53 in U-937-4, K562 and MCF-7 cells. Staf50 was directly activated by p53, as determined by the independence of de novo protein synthesis. Moreover, while the proximal promoter of Staf50 was found not to be p53 responsive, a functional enhancer-like p53-response element in intron 1 of the Staf50 gene was identified that was also transactivated by the p53-family member p73. Direct binding of p53 to the response element was shown by electrophoretic mobility shift analysis. Ectopic expression of Staf50 in U-937 cells resulted in reduced clonogenic growth. Moreover, levels of endogenous Staf50 mRNA correlated to all-trans retinoic acid-induced differentiation of promyelocytic NB-4 and HL60 cells, suggesting that Staf50 could be involved in proliferation and/or differentiation of leukemic cells.
Collapse
Affiliation(s)
- Susanna Obad
- Department of Hematology, Lund University, BMC; C14, Lund S-221 84, Sweden
| | | | | | | | | | | |
Collapse
|
322
|
Goldschneider D, Blanc E, Raguénez G, Barrois M, Legrand A, Le Roux G, Haddada H, Bénard J, Douc-Rasy S. Differential response of p53 target genes to p73 overexpression in SH-SY5Y neuroblastoma cell line. J Cell Sci 2004; 117:293-301. [PMID: 14676279 DOI: 10.1242/jcs.00834] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
p73, the first p53 gene homologue, encodes an array of p73 proteins including p73 alpha full-length (TAp73 alpha) and amino-truncated isoforms (Delta Np73 alpha), two proteins with opposite biological functions. TAp73 alpha can induce tumor suppressive properties, while Delta Np73 alpha antagonizes p53 as well as TAp73 in a dominant-negative manner. In human malignant neuroblasts, p53 protein is wild-type but known to be excluded from the nucleus, therefore disabling its function as a tumor suppressor. The present study investigates whether there is a functional link between p73 isoforms and p53 in neuroblastoma. Experiments were performed on two neuroblastoma cell lines differing in their p53 status, e.g. wild-type p53 SH-5Y5Y cells and mutated p53 IGR-N-91 cells. Data indicate that (i) both TA- and Delta N-p73 alpha enhance p53 protein level in SH-SY5Y cells, whereas level remains unchanged in IGR-N-91 cells; (ii) only in SH-SY5Y cells does forced TAp73 alpha overexpression markedly induce nuclear accumulation of p53 protein; (iii) p21 protein expression is increased in both cell lines infected with TAp73, suggesting that, in IGR-N-91 cells, p21 is induced by p73 through a p53-independent pathway; (iv) in the SHSY5Y cell line, Btg2 expression is strongly enhanced in cells overexpressing TA, and to a lesser extent in cells overexpressing Delta N. Taken together our results suggest that TAp73 may restore p53 function in NB with wild-type nonfunctional p53, but not in NB with mutated p53.
Collapse
Affiliation(s)
- David Goldschneider
- Centre National de la Recherche Scientifique-Unité Mixte de Recherche 8126, 39 rue Camille Desmoulins, 94805 Villejuif, France
| | | | | | | | | | | | | | | | | |
Collapse
|
323
|
Chan WM, Siu WY, Lau A, Poon RYC. How many mutant p53 molecules are needed to inactivate a tetramer? Mol Cell Biol 2004; 24:3536-51. [PMID: 15060172 PMCID: PMC381690 DOI: 10.1128/mcb.24.8.3536-3551.2004] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The tumor suppressor p53 is transcription factor composed of four identical subunits. The majority of the mutations in p53 are missense mutations that impair DNA binding. On the other hand, the p53-related p63 and p73 genes are rarely mutated, but many cell types express natural variants lacking the N-terminal transactivation domain (NDelta). Compelling evidence indicates that both the DNA binding-defective and NDelta mutants can impair the function of wild-type p53 in a dominant-negative manner. Interestingly, it is uncertain how many mutant subunit(s) a p53 tetramer can tolerate. In this study, we first made theoretical predictions based on the number of mutant p53 monomers needed to inactivate a tetramer and then tested how well the experimental data fit the predicted values. Surprisingly, these experiments reveal that DNA binding-defective p53 mutants (R249S and R273H) are very ineffective in impairing the transcriptional activity of p53: at least three mutants are required to inactivate a tetramer. In marked contrast, p53NDelta is a very potent inhibitor of p53: one NDelta subunit per tetramer is sufficient to abolish the transcriptional activity. DNA binding is not necessary for the NDelta proteins to inactivate p53. Similarly, NDelta variants of p63 and p73 are also powerful inhibitors of members of the p53 family. These results have important implications for our thinking about the mechanism of tumorigenesis involving missense p53 mutants or the N-terminally truncated isoforms.
Collapse
Affiliation(s)
- Wan Mui Chan
- Department of Biochemistry, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | | | | | | |
Collapse
|
324
|
Lo PK, Wang FF. 5'-Heterogeneity of mouse Dda3 transcripts is attributed to differential initiation of transcription and alternative splicing. Arch Biochem Biophys 2004; 425:221-32. [PMID: 15111131 DOI: 10.1016/j.abb.2004.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2004] [Revised: 03/22/2004] [Indexed: 10/26/2022]
Abstract
We have previously shown that mouse Dda3 gene is a p53 and p73 transcriptional target whose expression suppresses tumor cell growth. Here, we report the identification of multiple variants of Dda3 transcripts with diverse 5' sequences through 5'] rapid amplification of cDNA ends (5'-RACE) and RT-PCR. Analysis by primer extension and RNase protection revealed that the 5'-heterogeneity was generated by transcription initiation at multiple sites in exon 1 and intron 1 and by alternative splicing. These transcripts, both coding and non-coding, exhibited distinct expression patterns in various adult tissues and were developmentally regulated. Furthermore, they were induced in a p53-dependent manner by various stress signals. These data demonstrated that differential initiation of transcription and alternative splicing both participate in the regulation of Dda3 gene expression.
Collapse
Affiliation(s)
- Pang-Kuo Lo
- Institute of Biochemistry, National Yang-Ming University, Shih-Pai, Taipei 112, Taiwan
| | | |
Collapse
|
325
|
Affiliation(s)
- Frank McKeon
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
326
|
Assessment of p63 expression in the salivary gland neoplasms adenoid cystic carcinoma, polymorphous low-grade adenocarcinoma, and basal cell and canalicular adenomas. ACTA ACUST UNITED AC 2004. [DOI: 10.1016/j.tripleo.2003.09.010] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
327
|
Yang X, Klein R, Tian X, Cheng HT, Kopan R, Shen J. Notch activation induces apoptosis in neural progenitor cells through a p53-dependent pathway. Dev Biol 2004; 269:81-94. [PMID: 15081359 DOI: 10.1016/j.ydbio.2004.01.014] [Citation(s) in RCA: 237] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2003] [Revised: 01/13/2004] [Accepted: 01/14/2004] [Indexed: 10/26/2022]
Abstract
Notch signaling is involved in a variety of cell-fate decisions during development. Here we investigate the role of Notch signaling in apoptotic cell death of neural progenitors through the generation and analysis of cell type-specific conditional transgenic and knockout mice. We show that conditional expression of a constitutively active form of Notch1 in early neural progenitor cells, but not postmitotic neurons, selectively induces extensive apoptosis, resulting in a markedly reduced progenitor population. Conversely, attenuation of Notch signaling in Notch1 conditional knockout or Presenilin-1-/- mice results in reduced apoptosis of early neural progenitor cells. Furthermore, Notch activation in neural progenitor cells leads to elevated levels of nuclear p53 and transcriptional upregulation of the target genes Bax and Noxa, and the promotion of apoptotic cell death by Notch activation is completely suppressed by p53 deficiency. Together, these complementary gain-of-function and loss-of-function studies reveal a previously unappreciated role of Notch signaling in the regulation of apoptotic cell death during early mammalian neural development.
Collapse
Affiliation(s)
- Xudong Yang
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
328
|
Terrinoni A, Ranalli M, Cadot B, Leta A, Bagetta G, Vousden KH, Melino G. p73-alpha is capable of inducing scotin and ER stress. Oncogene 2004; 23:3721-5. [PMID: 15116103 DOI: 10.1038/sj.onc.1207342] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
p73, like its family member p53, can induce programmed cell death following DNA damage. Here, we report that this mechanism also involves endoplasmic reticulum (ER) stress and the transactivation of scotin, a protein identified recently as a p53 target able to induce ER stress. By using Tet-On inducible cell lines (Saos 2 osteosarcoma cells that lack p53), we observed that TAp73alpha elicits significant alterations in the morphology of the ER system, namely in the fine subcellular localization of calnexin. We found that both TAp73alpha and p53 are strong inducers of scotin. On the other hand, the transcriptionally deficient short isoforms DeltaNp73alpha did not upregulate the steady-state mRNA level of scotin, as evaluated by real-time RT-PCR. Following the induction of scotin, ER staining with calnexin showed evidence of morphological alteration, with variations in the intracellular concentration of free calcium, visualized by fluo-3 staining. The induction of ER stress by p73 was further supported by the transcriptional induction of Gadd 153, a transcription factor induced under ER stress conditions. In conclusion, the data reported indicate the ability of TAp73alpha and p53 (not DeltaNp73alpha) to elicit scotin transactivation and ER stress. This molecular mechanism might contribute to the effector events inducing apoptosis downstream of p73.
Collapse
Affiliation(s)
- Alessandro Terrinoni
- Biochemistry Laboratory, IDI-IRCCS, Department of Experimental Medicine and Biochemical Sciences, University of Rome 'Tor Vergata', 00133 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
329
|
Douc-Rasy S, Goldschneider D, Million K, Bénard J. [Interrelations between p73 and p53: a model, neuroblastoma]. Med Sci (Paris) 2004; 20:317-24. [PMID: 15067577 DOI: 10.1051/medsci/2004203317] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Homologies in sequence and gene organization of p53 and their relatives, p73 and p63, suggest similar biological functions. However differences exist between the p53 family members. Indeed in human tumors p53 is often mutated while p63 and p73 are very rarely mutated. In addition, in contrast to p53 which is transcribed in a unique mRNA species spanning all gene exons, each homologue expresses two types of isoforms: some with transactivation domain (TAD) showing tumor suppressive properties, the others deprived of TAD, with oncogenic properties. If p53 responds to immediate genotoxic stress, its homologues participate to the cell homeostasis of specific tissues along their development and differentiation, neuronal tissue for p73, epithelial for p63. However a collaboration between the three p53 family members has been shown to occur in response to cell genotoxic damages. Neuroblastic tumors characterized by a large spectrum of neuronal differentiation constitute a good model to study relationship between p73 and p53 as well as the regulation of their respective expression.
Collapse
|
330
|
Abstract
The tumor suppressor p53 is tightly controlled at low levels in cells by constant ubiquitination and proteasomal degradation. In response to stresses, ubiquitination of p53 is inhibited through diverse pathways, depending on the nature of the stimulus and cell type. This leads to the accumulation and activation of p53, which induces cell cycle arrest and/or apoptosis to prevent cells from transformation. Many studies have indicated that defects of the p53 system are present in most, if not all, human tumor cells. Meanwhile, significant progress has been made in understanding the molecular mechanisms of p53 ubiquitination and the regulation of the p53 system. Therefore, it is possible now to consider targeting ubiquitination as a means to regulate and reactivate p53 in tumors. Emerging evidence suggests that inhibiting the E3 activity of Mdm2, blocking the interaction of p53 and Mdm2, and restoring the function of mutated p53 are potential effective strategies to kill certain tumor cells selectively. It is conceivable that new chemotherapeutic agents based on these studies will be generated in the not-so-distant future.
Collapse
Affiliation(s)
- Yili Yang
- Regulation of Protein Function Laboratory, Center for Cancer Research, National Cancer Institute-Frederick, 1050 Boyles Street, 560/22-64, Frederick, MD 21702, USA.
| | | | | |
Collapse
|
331
|
Abstract
The p53 family of proteins includes three members, p53, p63, and p73. The levels and stability of p53 are controlled in large part by MDM2, which can bind the p53 N-terminus and promote its degradation. Because the MDM2 gene is transcriptionally activated by p53, it forms part of an autoregulatory feedback loop that directly links the transcriptional activity of p53 with its degradation. In contrast, little is known about the mechanisms that control p63 or p73 stability. In the current study, p73 deletion or point mutants that lacked transactivation activity were stable compared to wild-type p73. A naturally occurring p73 variant (DeltaNp73) was also stable compared to wild-type p73. Finally, fusion of the VP16-transactivation domain to an inactive, stable p73 mutant restored transactivation function and rendered the mutant protein unstable. These results demonstrate that p73 transactivation activity is necessary for rapid p73 turnover, and suggest that one or more transcriptional targets of p73 may promote its degradation.
Collapse
Affiliation(s)
- Liqing Wu
- Department of Radiation and Cellular Oncology, The University of Chicago, 5841 S. Maryland Ave., MC1105, room G-06, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
332
|
Courtois S, Caron de Fromentel C, Hainaut P. p53 protein variants: structural and functional similarities with p63 and p73 isoforms. Oncogene 2004; 23:631-8. [PMID: 14737098 DOI: 10.1038/sj.onc.1206929] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Since its discovery in 1979, many studies have reported that the p53 tumour suppressor protein could be expressed in the form of products smaller than those predicted by the full-length amino-acid sequence. These products differ from full-length p53 in their N- or C-terminal regions, but generally conserve the central, DNA-binding domain. They appear to be expressed at rather low levels and to be restricted to particular cell types and/or physiological circumstances, suggesting that they play very narrow and specific roles. Several mechanisms have been proposed to explain their timely occurrence, including alternative splicing, internal initiation of translation or proteolytic cleavage. A precise assessment of the various 'p53 isoforms' reveals striking similarities with several isoforms of the p53 homologous proteins p63 or p73, suggesting that regulated production of specific, N- or C-terminal variants may be a 'trademark' of all family members. In this review, we summarize the published evidence on the structure, mode of production, expression and function of the p53 isoforms, and discuss their properties in the light of recent data on the structure and function of p63/p73 isoforms.
Collapse
Affiliation(s)
- Stéphanie Courtois
- Unit of Molecular Carcinogenesis, International Agency for Research on Cancer, 150 cours Albert Thomas, 69372 Lyon Cedex 08, France
| | | | | |
Collapse
|
333
|
Abstract
The recent discovery of two genes, termed p63 and p73, encoding transcription factors highly homologous to p53 presents unexpected challenges and opportunities for the understanding and treatment of cancers. The questions raised are many but center on determining whether these new genes possess novel tumor suppressor functions, cooperate with p53, or impart oncogenic effects. At present there is considerable discord in the field concerning these concepts with some favoring a tumor suppressor role for the p53 family members and others an oncogenic influence. In support of a tumor suppressor role is the ability of p73 and p63 isoforms to transactivate p53 target genes and the large body of work linking p73, and to some extent p63, in apoptotic events in response to cellular stresses generally considered the purview of p53. More recently, p73 has been implicated in cell death following T cell activation, the response of cancers to chemotherapy, and finally, along with p63, to the function of p53 itself. Opposing this view is the fact that the p73 and p63 genes are rarely mutated in cancers and the stark absence of tumors in the p73 null mouse. Moreover, the high expression of dominant negative (dn) versions of the p73 and p63 proteins supports an anti-p53 function and therefore possibly an oncogenic effect. Indeed, the p63 gene is located in a region of chromosome three amplified in squamous cell carcinomas and the number of reports of dn-p63 overexpression in these diseases is increasing. This review will examine both sides of these arguments in an attempt to decipher common themes and to identify opportunities these genes represent for understanding tumorigenesis.
Collapse
Affiliation(s)
- Frank D McKeon
- Department of Cell Biology, Harvard Medical School, USA.
| |
Collapse
|
334
|
Bergamaschi D, Samuels Y, Jin B, Duraisingham S, Crook T, Lu X. ASPP1 and ASPP2: common activators of p53 family members. Mol Cell Biol 2004; 24:1341-50. [PMID: 14729977 PMCID: PMC321425 DOI: 10.1128/mcb.24.3.1341-1350.2004] [Citation(s) in RCA: 184] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
We recently showed that ASPP1 and ASPP2 stimulate the apoptotic function of p53. We show here that ASPP1 and ASPP2 also induce apoptosis independently of p53. By binding to p63 and p73 in vitro and in vivo, ASPP1 and ASPP2 stimulate the transactivation function of p63 and p73 on the promoters of Bax, PIG3, and PUMA but not mdm2 or p21(WAF-1/CIP1). The expression of ASPP1 and ASPP2 also enhances the apoptotic function of p63 and p73 by selectively inducing the expression of endogenous p53 target genes, such as PIG3 and PUMA, but not mdm2 or p21(WAF-1/CIP1). Removal of endogenous p63 or p73 with RNA interference demonstrated that (16) the p53-independent apoptotic function of ASPP1 and ASPP2 is mediated mainly by p63 and p73. Hence, ASPP1 and ASPP2 are the first two identified common activators of all p53 family members. All these results suggest that ASPP1 and ASPP2 could suppress tumor growth even in tumors expressing mutant p53.
Collapse
Affiliation(s)
- Daniele Bergamaschi
- Ludwig Institute for Cancer Research, Imperial College School of Medicine, St. Mary's Campus, London W2 1PG, United Kingdom
| | | | | | | | | | | |
Collapse
|
335
|
Liu G, Nozell S, Xiao H, Chen X. DeltaNp73beta is active in transactivation and growth suppression. Mol Cell Biol 2004; 24:487-501. [PMID: 14701724 PMCID: PMC343790 DOI: 10.1128/mcb.24.2.487-501.2004] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
p73, a p53 family protein, shares significant sequence homolog and functional similarity with p53. However, unlike p53, p73 has at least seven alternatively spliced isoforms with different carboxyl termini (p73alpha-eta). Moreover, the p73 gene can be transcribed from a cryptic promoter located in intron 3, producing seven more proteins (DeltaNp73alpha-eta). DeltaNp73, which does not contain the N-terminal activation domain in p73, has been thought to be transcriptionally inactive and dominant negative over p53 or p73. To systemically analyze the activity of the DeltaN variant, we generated stable cell lines, which inducibly express DeltaNp73alpha, DeltaNp73beta, and various DeltaNp73beta mutants by using the tetracycline-inducible expression system. Surprisingly, we found that DeltaNp73beta is indeed active in inducing cell cycle arrest and apoptosis. Importantly, we found that, when DeltaNp73beta is expressed at a physiologically relevant level, it is capable of suppressing cell growth. We then demonstrated that these DeltaNp73beta activities are not cell type specific. We showed that the 13 unique residues at the N terminus are required for DeltaNp73beta to suppress cell growth. We also found that, among the 13 residues, residues 6 to 10 are critical to DeltaNp73beta function. Furthermore, we found that DeltaNp73beta is capable of inducing some p53 target genes, albeit to a lesser extent than does p73beta. Finally, we found that the 13 unique residues, together with the N-terminal PXXP motifs, constitute a novel activation domain. Like DeltaNp73beta, DeltaNp73gamma is active in transactivation. However, unlike DeltaNp73beta, DeltaNp73alpha is inactive in suppressing cell growth. Our data, together with others' previous findings, suggest that DeltaNp73beta may have distinct functions under certain cellular circumstances.
Collapse
Affiliation(s)
- Gang Liu
- Department of Cell Biology, The University of Alabama at Birmingham, 1530 3rd Avenue, Birmingham, AL 35294-0005, USA
| | | | | | | |
Collapse
|
336
|
Tessema M, Lehmann U, Kreipe H. Cell cycle and no end. Virchows Arch 2004; 444:313-23. [PMID: 14968363 DOI: 10.1007/s00428-003-0971-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2003] [Accepted: 12/17/2003] [Indexed: 12/19/2022]
Abstract
Our knowledge about the molecular circuits regulating the duplication of the genetic material and the subsequent division of a cell into two daughter cells has exploded over the last decade. Aberrations in the regulation of the cell cycle belong to the hallmarks of malignant transformation, leading, in turn, to the development of tumours. After introducing the basics of eukaryotic cell-cycle regulation and describing the four phases of the cell cycle (namely, G1, S, G2 and M) in more detail, alterations of key components of the cell-cycle machinery in human malignancies and their functional consequences are presented. Principally, deregulation of the cell cycle can be caused by unrestricted activity of cell-cycle promoting factors (many oncogenes fall into this class) or by inactivation of inhibitory factors (many tumour suppressor genes belong to this class). Both types of deregulation have been described in human tumours and are discussed in detail. Perspectives concerning the translation of this knowledge into daily routine practice and future applications are discussed at the end. The molecular mechanisms of actual cell division (sister chromatid segregation and cytokinesis) are mentioned only briefly.
Collapse
Affiliation(s)
- Mathewos Tessema
- Institute of Pathology, Medizinische Hochschule Hannover, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | | | | |
Collapse
|
337
|
Sayan AE, Rossi M, Melino G, Knight RA. p73: in silico evidence for a putative third promoter region. Biochem Biophys Res Commun 2004; 313:765-70. [PMID: 14697257 DOI: 10.1016/j.bbrc.2003.12.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The TP73 gene, first identified in 1997, encodes the p73 protein, a p53 tumour suppressor homologue. The lack of mutations in the TP73 gene in cancers and the developmentally abnormal phenotype of the TP73 knockout mouse suggest a different function for TP73 gene derived proteins. An alternative promoter in the third intron of the TP73 gene, which produces a transcription deficient and dominant negative protein (DeltaNp73), produces increased complexity in the function of p73 proteins. Functional studies of transcriptionally active (TAp73; regulated by the first promoter) and DeltaNp73 (regulated by the second promoter) show that the TP73 gene encodes both a candidate tumour suppressor (TAp73) and an oncogene (DeltaNp73), with pro-apoptotic and anti-apoptotic properties, respectively. This "two in one" gene architecture leads us to make an in silico search for other probable promoter regions and transcription start sites in different introns of the TP73 gene. To identify such regulatory regions, we have analysed the genomic structure of human and mouse TP73 genes. We have found introns 1 and 4 to be extremely large and relatively conserved in size in mouse and human, in addition to intron 3, which includes the DeltaNp73 promoter. We have further characterized these introns by transcription factor binding motifs, transcription initiation sites, open reading frames, and splicing using six programs that can successfully identify the already characterized promoters. Our results suggest the presence of a candidate 2 kbp genomic DNA in the first intron of human TP73 gene, harbouring 2 putative promoter regions, together with a similar region within intron 1 of the mouse gene.
Collapse
Affiliation(s)
- A Emre Sayan
- Medical Research Council Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK
| | | | | | | |
Collapse
|
338
|
Billon N, Terrinoni A, Jolicoeur C, McCarthy A, Richardson WD, Melino G, Raff M. Roles for p53 and p73 during oligodendrocyte development. Development 2004; 131:1211-20. [PMID: 14960496 DOI: 10.1242/dev.01035] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Oligodendrocytes make myelin in the vertebrate central nervous system (CNS). They develop from oligodendrocyte precursor cells (OPCs), most of which divide a limited number of times before they stop and differentiate. OPCs can be purified from the developing rat optic nerve and stimulated to proliferate in serum-free culture by PDGF. They can be induced to differentiate in vitro by either thyroid hormone (TH) or PDGF withdrawal. It was shown previously that a dominant-negative form of p53 could inhibit OPC differentiation induced by TH but not by PDGF withdrawal, suggesting that the p53 family of proteins might play a part in TH-induced differentiation. As the dominant-negative p53 used inhibited all three known p53 family members - p53, p63 and p73 - it was uncertain which family members are important for this process. Here, we provide evidence that both p53 and p73, but not p63, are involved in TH-induced OPC differentiation and that p73 also plays a crucial part in PDGF-withdrawal-induced differentiation. This is the first evidence for a role of p73 in the differentiation of a normal mammalian cell.
Collapse
Affiliation(s)
- Nathalie Billon
- MRC Laboratory for Molecular Cell Biology and Cell Biology Unit, University College London, London WC1E 6BT, UK.
| | | | | | | | | | | | | |
Collapse
|
339
|
Melino G, Bernassola F, Ranalli M, Yee K, Zong WX, Corazzari M, Knight RA, Green DR, Thompson C, Vousden KH. p73 Induces Apoptosis via PUMA Transactivation and Bax Mitochondrial Translocation. J Biol Chem 2004; 279:8076-83. [PMID: 14634023 DOI: 10.1074/jbc.m307469200] [Citation(s) in RCA: 274] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
p73, an important developmental gene, shares a high sequence homology with p53 and induces both G(1) cell cycle arrest and apoptosis. However, the molecular mechanisms through which p73 induces apoptosis are unclear. We found that p73-induced apoptosis is mediated by PUMA (p53 up-regulated modulator of apoptosis) induction, which, in turn, causes Bax mitochondrial translocation and cytochrome c release. Overexpression of p73 isoforms promotes cell death and bax promoter transactivation in a time-dependent manner. However, the kinetics of apoptosis do not correlate with the increase of Bax protein levels. Instead, p73-induced mitochondrial translocation of Bax is kinetically compatible with the induction of cell death. p73 is localized in the nucleus and remains nuclear during the induction of cell death, indicating that the effect of p73 on Bax translocation is indirect. The ability of p73 to directly transactivate PUMA and the direct effect of PUMA on Bax conformation and mitochondrial relocalization suggest a molecular link between p73 and the mitochondrial apoptotic pathway. Our data therefore indicate that PUMA-mediated Bax mitochondrial translocation, rather than its direct transactivation, correlates with cell death. Finally, human DeltaNp73, an isoform lacking the amino-terminal transactivation domain, inhibits TAp73-induced as well as p53-induced apoptosis. The DeltaNp73 isoforms seem therefore to act as dominant negatives, repressing the PUMA/Bax system and, thus, finely tuning p73-induced apoptosis. Our findings demonstrate that p73 elicits apoptosis via the mitochondrial pathway using PUMA and Bax as mediators.
Collapse
Affiliation(s)
- Gerry Melino
- Biochemistry Laboratory, Instituto Dermopatico Dell'Immacolata-Istituto di Ricovero e Cura a Carattere Scientifico, Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
340
|
Cox RL, Stephens RE, Reinisch CL. p63/73 homologues in surf clam: novel signaling motifs and implications for control of expression. Gene 2004; 320:49-58. [PMID: 14597388 DOI: 10.1016/j.gene.2003.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To understand the role of p53 gene family members during invertebrate embryonic development, we used polymerase chain reaction (PCR) to identify p63/73 homologues in the marine mollusc Spisula solidissima. Here, we report the sequences of two distinct p63/73-like homologues, both cloned from Spisula embryos. The first, Ssp63/73alpha is 2699 nucleotide (nt); the second, Spp63/73beta is 3920 nt. The nucleotide sequences of the two variants are nearly identical up to their stop codons but diverge in their 3'-untranslated regions (UTRs). The deduced amino acid sequence of both Ssp63/73 variants is 597 amino acids, coding for a protein with predicted molecular weight of approximately 68 kDa. We conclude that the two unique transcripts, containing 3' UTRs of variable lengths, represent tandem alternate polyadenylation sites for the Ssp63/73 gene. While alternative splicing has been well documented in the p63/73 gene family, this is the first report of alternate polyadenylation site choice as a control point for p63/73 gene expression in any species. In order to identify specific post-transcriptional as well as post-translational signals potentially involved in regulation of p63/73-like expression, we compared Ssp63/p73 nucleotide and Ssp63/73 deduced amino acid sequences to corresponding regions of other mammalian and nonmammalian p63 and p73 homologues. Within the Spisula 3' UTRs we identified multiple AU-rich elements (AREs) which may control translation activation. Within the deduced amino acid sequence, we identified potential sites for sumoylation, a post-translational process that has been identified in mammalian p63 and p73 proteins. Identification of these novel signaling sites provides information about potential mechanisms controlling expression of multiple p63/73 isoforms during development.
Collapse
Affiliation(s)
- Rachel L Cox
- Laboratory of Aquatic Biomedicine, Woods Hole, MA 02543, USA.
| | | | | |
Collapse
|
341
|
Abstract
The p53 tumor suppressor acts to integrate multiple stress signals into a series of diverse antiproliferative responses. One of the most important p53 functions is its ability to activate apoptosis, and disruption of this process can promote tumor progression and chemoresistance. p53 apparently promotes apoptosis through transcription-dependent and -independent mechanisms that act in concert to ensure that the cell death program proceeds efficiently. Moreover, the apoptotic activity of p53 is tightly controlled, and is influenced by a series of quantitative and qualitative events that influence the outcome of p53 activation. Interestingly, other p53 family members can also promote apoptosis, either in parallel or in concert with p53. Although incomplete, our current understanding of p53 illustrates how apoptosis can be integrated into a larger tumor suppressor network controlled by different signals, environmental factors, and cell type. Understanding this network in more detail will provide insights into cancer and other diseases, and will identify strategies to improve their therapeutic treatment.
Collapse
|
342
|
Abstract
In eukaryotes, transcription of the diverse array of tens of thousands of protein-coding genes is carried out by RNA polymerase II. The control of this process is predominantly mediated by a network of thousands of sequence-specific DNA binding transcription factors that interpret the genetic regulatory information, such as in transcriptional enhancers and promoters, and transmit the appropriate response to the RNA polymerase II transcriptional machinery. This review will describe some early advances in the discovery and characterization of the sequence-specific DNA binding transcription factors as well as some of the properties of these regulatory proteins.
Collapse
Affiliation(s)
- James T Kadonaga
- Section of Molecular Biology, 0347, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
343
|
Tomimori Y, Katoh I, Kurata SI, Okuyama T, Kamiyama R, Ikawa Y. Evolutionarily conserved expression pattern and trans-regulating activity of Xenopus p51/p63. Biochem Biophys Res Commun 2004; 313:230-6. [PMID: 14684151 DOI: 10.1016/j.bbrc.2003.11.113] [Citation(s) in RCA: 9] [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
p51/p63, a member of the p53 gene family, is structurally conserved among a wide range of organisms, although the transactivator (TA) and N-terminally truncated (deltaN) isotype producing property seems to vary. Since p51/p63 is thought to play important roles in skin, limb, and craniofacial development in mammals, we examined Xenopus laevis larval and adult tissues for expression of p51/p63. Temporal analyses indicated enhanced transcription of the deltaN form of p51/p63 in premetamorphosis phase (at stage 44-48). p51/p63-positive cells in the inner layer of larval skin expanded to the suprabasal layers during the stratification. The epithelium of limb buds and the maxillofacial ectodermal tissues in tadpoles had a high level expression of p51/p63. The cloned deltaN-A/gamma type Xenopus p51/p63 exhibited a dominant-negative activity against the human TA-A/gamma isotype in a reporter assay. These results suggest that tissue-specific p51/p63-inducing mechanism and isotype-specific transcriptional regulator activities of p51/p63 are conserved between mammals and frogs.
Collapse
Affiliation(s)
- Yoshiya Tomimori
- Molecular Pathophysiology, Department of Moleculo-genetic Sciences, Division of Biomedical Laboratory Sciences, Graduate School of Allied Health Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | | | | | | | | | | |
Collapse
|
344
|
Ribeiro-Silva A, Ramalho LNZ, Garcia SB, Zucoloto S. Does the correlation between EBNA-1 and p63 expression in breast carcinomas provide a clue to tumorigenesis in Epstein-Barr virus-related breast malignancies? Braz J Med Biol Res 2004; 37:89-95. [PMID: 14689049 DOI: 10.1590/s0100-879x2004000100013] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Several investigators have identified Epstein-Barr virus (EBV) particles in breast carcinomas, a fact that supports a role for EBV in mammary tumorigenesis. The possible mechanism involved in this process is not clear. The present study was carried out in an attempt to determine whether there is a relationship between latent infection with EBV and p53 and p63 expression in breast carcinomas. Immunohistochemistry developed with 3.3-diaminobenzidine tetrahydrochloride was performed in 85 formalin-fixed paraffin-embedded breast carcinomas using anti-EBV EBNA-1, anti-p63, anti-p53, anti-estrogen receptor (ER) and anti-progesterone receptor (PR) antibodies. The cases were selected to represent each of the various histologic types: intraductal carcinoma (N=12), grade I invasive ductal carcinoma (N=15), grade II invasive ductal carcinoma (N=15), grade III invasive ductal carcinoma (N=15), tubular carcinoma (N=8), lobular carcinoma (N=10), and medullary carcinoma (N=10). The ductal breast carcinomas were graded I, II and III based on the Scarff-Bloom and Richardson grading system modified by Elston and Ellis. One slide containing at least 1000 neoplastic cells was examined in each case. ER, PR, p63, p53 and EBNA-1 were positive in 60, 40, 11.8, 21.2 and 37.6% of carcinomas, respectively. There was a correlation between EBNA-1 and p63 expression (P<0.001), but not between EBNA-1 and p53 (P=0.10). These data suggest a possible role for p63 in the mammary tumorigenesis associated with Epstein-Barr virus infection.
Collapse
MESH Headings
- Adult
- Aged
- Biomarkers, Tumor
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/virology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/virology
- DNA-Binding Proteins
- Epstein-Barr Virus Nuclear Antigens/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Genes, Tumor Suppressor
- Herpesvirus 4, Human/isolation & purification
- Humans
- Immunohistochemistry
- Middle Aged
- Phosphoproteins/genetics
- Receptors, Estrogen/analysis
- Receptors, Progesterone/analysis
- Trans-Activators/genetics
- Transcription Factors
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Proteins
Collapse
Affiliation(s)
- A Ribeiro-Silva
- Departamento de Patologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil.
| | | | | | | |
Collapse
|
345
|
Tullo A, Mastropasqua G, Bourdon JC, Centonze P, Gostissa M, Costanzo A, Levrero M, Del Sal G, Saccone C, Sbisà E. Adenosine deaminase, a key enzyme in DNA precursors control, is a new p73 target. Oncogene 2003; 22:8738-48. [PMID: 14647469 DOI: 10.1038/sj.onc.1206967] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery of the p73 and p63 genes, homologous to p53 tumor suppressor has uncovered a family of transcription factors and widened the scenario of cell cycle control and apoptosis. We have identified a putative p53-responsive element in the human adenosine deaminase (ADA) gene, an important enzyme involved in nucleotide metabolism, the deficit of which causes the inhibition of DNA synthesis and repair. Here, we demonstrate that the ectopic expression of p73 isoforms leads to the ADA gene upregulation, showing for the first time a correlation between p73 and ADA. We found that p73 promotes ADA gene expression following a dNTP unbalance generated by ADA enzyme deficiency and 2'deoxyadenosine accumulation. The abrogation of p73 transcriptional activity by the specific dominant-negative p73DD abolishes ADA induction. By contrast, the ADA gene does not appear to be a functional p53 target in the physiological conditions we tested. On the whole, our results contribute to the emerging picture that p73 could play a different role from p53 in normal growth and development by inducing alternative target genes, which are not shared by p53.
Collapse
Affiliation(s)
- Apollonia Tullo
- Istituto di Tecnologie Biomediche, Sezione di Bari di Bioinformatica e Genomica CNR, Via Amendola, 165/A Bari 70126, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
346
|
Maisse C, Guerrieri P, Melino G. p73 and p63 protein stability: the way to regulate function? Biochem Pharmacol 2003; 66:1555-61. [PMID: 14555234 DOI: 10.1016/s0006-2952(03)00511-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
While the p53 homologue p73 has been found to be involved in tumorigenesis, the molecular mechanisms involved in this function are still not fully evident. The presence of two distinct promoters allows the formation of two proteins with opposite effects: while TA-p73 shows pro-apoptotic effects, DeltaN-p73 has an evident anti-apoptotic function. The relative expression of the two proteins is in fact related to the prognosis of several cancers. Since both p73 and p63, the other member of the same family, share the ability to interact with each other, it is important to understand the mechanisms that control the degradation and stability of both proteins, and their relative isoforms. p73 and p63 stability is regulated not only by protein modifications (phosphorylation, acetylation) but also by its degradation in the proteasome. To this end, the interaction with Mdm2, p300/CBP, and SUMO-1 are discussed in details.
Collapse
Affiliation(s)
- Carine Maisse
- Department of Experimental Medicine, Biochemistry Laboratory, IDI-IRCCS, University of Rome Tor Vergata, Rome, Italy
| | | | | |
Collapse
|
347
|
Harmes DC, Bresnick E, Lubin EA, Watson JK, Heim KE, Curtin JC, Suskind AM, Lamb J, DiRenzo J. Positive and negative regulation of ΔN-p63 promoter activity by p53 and ΔN-p63-α contributes to differential regulation of p53 target genes. Oncogene 2003; 22:7607-16. [PMID: 14576823 DOI: 10.1038/sj.onc.1207129] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mammary epithelial regeneration implies the existence of cellular progenitors with retained replicative capacity, prolonged lifespan and developmental potency. Evidence exists that deltaN-p63 isoforms preserve these features by modulating p53 activity in basal epithelia. deltaN-p63 mRNA levels decline at the onset of differentiation suggesting that its transcriptional regulation may contribute to the initiation of differentiation. To study transcriptional regulation of deltaN-p63, a 10.3 kbp fragment containing the deltaN-p63 promoter was isolated. We report here that deltaN-p63 is a positive and negative transcriptional target of p53 and deltaN-p63-alpha, respectively. Disruption of p53 activity or expression abolishes the expression of deltaN-p63-alpha. This regulation is mediated by a p53-binding element sufficient to confer these activities to a heterologous promoter. Chromatin immune-precipitation indicates that, in asynchronously growing cells, p53 occupies this element. In response to DNA damage, deltaN-p63-alpha is recruited to this element as transcription of deltaN-p63 declines. Disruption of deltaN-p63-alpha expression had differential effects on the transcriptional regulation of several p53-target genes. These findings indicate that p53 contributes to the preservation of basal epithelia by driving the expression of deltaN-p63 isoforms. These studies also suggest that in response to genotoxic stress, deltaN-p63-alpha mediates the silencing of its own promoter thereby altering the pattern of p53-target gene expression.
Collapse
Affiliation(s)
- David Christopher Harmes
- Department of Pharmacology and Toxicology, Dartmouth Medical School, 7650 Remsen, Hanover, NH 03755, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
348
|
Yamada G, Satoh Y, Baskin LS, Cunha GR. Cellular and molecular mechanisms of development of the external genitalia. Differentiation 2003; 71:445-60. [PMID: 14641326 DOI: 10.1046/j.1432-0436.2003.7108001.x] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The limb and external genitalia are appendages of the body wall. Development of these structures differs fundamentally in that masculine development of the external genitalia is androgen dependent, whereas development of the limb is not. Despite this fundamental difference in developmental regulation, epithelial-mesenchymal interactions play key roles in the development of both structures, and similar regulatory molecules are utilized as mediators of morphogenetic cell-cell interactions during development of both the limb and external genitalia. Given the relatively high incidence of hypospadias, a malformation of penile development, it is appropriate and timely to review the morphological, endocrine, and molecular mechanisms of development of the genital tubercle (GT), the precursor of the penis in males and the clitoris in females. Morphological observations comparing development of the GT in humans and mouse emphasize the validity of the mouse as an animal model of GT development and validate the results of experimental studies. Accordingly, the use of mutant mice provides important insights into the roles of specific regulatory molecules in development of the external genitalia. While our current understanding of the morphological and molecular mechanisms of mammalian external genitalia development is still rudimentary, this review summarizes the current state of our knowledge and whenever possible draws from the rich experimental embryology literature on other relevant organs such as the developing limb. Future research on the hormonal and molecular mechanisms of GT development may yield strategies to prevent or reduce the incidence of hypospadias and to elucidate the molecular genetic mechanisms of GT morphogenesis, especially in relation to common organogenetic pathways utilized in other organ systems.
Collapse
Affiliation(s)
- Gen Yamada
- Center for Animal Resources and Development (CARD) and Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, Honjo, Kumamoto 860-0811, Japan
| | | | | | | |
Collapse
|
349
|
Kim EJ, Park JS, Um SJ. Identification of Daxx interacting with p73, one of the p53 family, and its regulation of p53 activity by competitive interaction with PML. Nucleic Acids Res 2003; 31:5356-67. [PMID: 12954772 PMCID: PMC203324 DOI: 10.1093/nar/gkg741] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
We performed a yeast two-hybrid screen using p73alpha, which is a member of the p53 family, as bait. We found that the p53 family members were functionally associated with Daxx, which was described originally as a cytoplasmic mediator of Fas signaling, but has been identified recently as a nuclear protein that co-localizes with the promyelocytic leukemia (PML) protein and regulates transcription. Extensive yeast two-hybrid assays indicated a physical interaction between a region including the oligomerization domain (OD) of p73alpha (amino acids 345-380) or p53 (amino acids 319-360) and amino acids 161-311 and 667-740 (C-terminal S/P/T-rich domain) of hDaxx, which is the common binding region of Fas, ASK1 and PML. This interaction was further confirmed by in vitro GST pull-down and in vivo immunoprecipitation assays. Both Daxx and p73/p53 co-localized in nuclear dot-like structures, which are probably nuclear PML oncogenic domains (PODs) or the nuclear domain NB10. Transient co-expression of Daxx resulted in strong inhibition of p73- and p53-mediated transcriptional activation of the synthetic p53-responsive and p21WAF1 promoters. Consequently, Gal4-Daxx repressed basal transcription in a dose-dependent manner. Treatment with trichostatin A, which is an inhibitor of histone deacetylase, or PML over-expression relieved Daxx-mediated transcriptional repression of p53. The mechanism underlying PML-mediated derepression appears to be competitive binding between Daxx, p53 and PML. Taken together, these findings delineate a transcriptional regulatory network that is modulated by differential Daxx-p53-PML interactions in the nuclear PODs. Therefore, Daxx is implicated in the regulation of the cell cycle and apoptosis through transcriptional regulation of p53 and possibly its family members.
Collapse
Affiliation(s)
- Eun-Joo Kim
- Department of Bioscience and Biotechnology/Institute of Bioscience, Sejong University, 98 Kunja-dong, Kwangjin-gu, Seoul 143-747, Korea
| | | | | |
Collapse
|
350
|
Momota H, Ichimiya S, Ikeda T, Yamaki T, Kikuchi T, Houkin K, Sato N. Immunohistochemical analysis of the p53 family members in human craniopharyngiomas. Brain Tumor Pathol 2003; 20:73-7. [PMID: 14756444 DOI: 10.1007/bf02483450] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Craniopharyngiomas are intracranial tumors that usually arise in the site around the sella turcica. They are composed of distinctive sheets of epithelial cells showing adamantinomatous or squamous-papillary histologic types. Because little is known about the tumorigenesis of craniopharyngiomas, we retrieved samples from 15 tumor cases to investigate the functional significance of the p53 family of transcription factors, which are known to be expressed in various human epithelia. Immunohistochemical analysis of these cases demonstrated similar expression profiles of p53 family members in the two histologic types of the tumor; i.e., strong nuclear expression of p63 was observed in all cell layers, and moderate to intense nuclear expression of p73 was observed in the basal cell layers. In contrast to p63 and p73, the reactivity of an archetypal tumor suppressor, p53, was occasional and weak in the two histologic types. Because p63 was widely expressed in the tumors, reverse transcription-polymerase chain reaction (RT-PCR) analysis was conducted to elucidate which spliced variant of p63 was expressed. The results showed that deltaNp63, lacking a terminal transactivation domain of p63, was the dominant isoform. Together with the reported evidence that the deltaNp63 isoform is highly expressed in human squamous-cell carcinomas, these data suggest that the cellular architecture characteristic of the expression of p53 family members may be required for the histogenesis of craniopharyngiomas, where deltaNp63 has a possible role in maintaining proliferative activity of the tumor cells, like squamous-cell carcinomas in other tissues.
Collapse
Affiliation(s)
- Hiroyuki Momota
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo 060-8556, Japan.
| | | | | | | | | | | | | |
Collapse
|