151
|
Abstract
In its wild-type form, p53 is a major tumor suppressor whose function is critical for protection against cancer. Many human tumors carry missense mutations in the TP53 gene, encoding p53. Typically, the affected tumor cells accumulate excessive amounts of the mutant p53 protein. Various lines of evidence indicate that, in addition to abrogating the tumor suppressor functions of wild-type p53, the common types of cancer-associated p53 mutations also endow the mutant protein with new activities that can contribute actively to various stages of tumor progression and to increased resistance to anticancer treatments. Collectively, these activities are referred to as mutant p53 gain-of-function. This article addresses the biological manifestations of mutant p53 gain-of-function, the underlying molecular mechanisms, and their possible clinical implications.
Collapse
Affiliation(s)
- Moshe Oren
- Department of Molecular Cell Biology, The Weizmann Institute, Rehovot 76100, Israel.
| | | |
Collapse
|
152
|
Gast A, Scherer D, Chen B, Bloethner S, Melchert S, Sucker A, Hemminki K, Schadendorf D, Kumar R. Somatic alterations in the melanoma genome: a high-resolution array-based comparative genomic hybridization study. Genes Chromosomes Cancer 2010; 49:733-45. [PMID: 20544847 DOI: 10.1002/gcc.20785] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We performed DNA microarray-based comparative genomic hybridization to identify somatic alterations specific to melanoma genome in 60 human cell lines from metastasized melanoma and from 44 corresponding peripheral blood mononuclear cells. Our data showed gross but nonrandom somatic changes specific to the tumor genome. Although the CDKN2A (78%) and PTEN (70%) loci were the major targets of mono-allelic and bi-allelic deletions, amplifications affected loci with BRAF (53%) and NRAS (12%) as well as EGFR (52%), MITF (40%), NOTCH2 (35%), CCND1 (18%), MDM2 (18%), CCNE1 (10%), and CDK4 (8%). The amplified loci carried additional genes, many of which could potentially play a role in melanoma. Distinct patterns of copy number changes showed that alterations in CDKN2A tended to be more clustered in cell lines with mutations in the BRAF and NRAS genes; the PTEN locus was targeted mainly in conjunction with BRAF mutations. Amplification of CCND1, CDK4, and other loci was significantly increased in cell lines without BRAF-NRAS mutations and so was the loss of chromosome arms 13q and 16q. Our data suggest involvement of distinct genetic pathways that are driven either through oncogenic BRAF and NRAS mutations complemented by aberrations in the CDKN2A and PTEN genes or involve amplification of oncogenic genomic loci and loss of 13q and 16q. It also emerges that each tumor besides being affected by major and most common somatic genetic alterations also acquires additional genetic alterations that could be crucial in determining response to small molecular inhibitors that are being currently pursued.
Collapse
Affiliation(s)
- Andreas Gast
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Huang CY, Chen JYF, Wu JE, Pu YS, Liu GY, Pan MH, Huang YT, Huang AM, Hwang CC, Chung SJ, Hour TC. Ling-Zhi polysaccharides potentiate cytotoxic effects of anticancer drugs against drug-resistant urothelial carcinoma cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2010; 58:8798-8805. [PMID: 20681668 DOI: 10.1021/jf1020158] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The combined effects of ling-zhi polysaccharide fraction 3 (LZP-F3) and anticancer drugs (cisplatin and arsenic trioxide) were examined in three human urothelial carcinoma (UC) cells (parental, NTUB1; cisplatin-resistant, N/P(14); and arsenic-resistant, N/As(0.5)). MTT assay and median-effect analysis revealed that LZP-F3 could profoundly reverse the chemosensitivity of N/P(14) and N/As(0.5) to cisplatin and arsenic, respectively, in a dose-dependent manner, which involved activation of p38 and down-regulation of Akt and XPA. A dose of 10 mug/mL of LZP-F3 induced significant G1 arrest in N/P(14) and N/As(0.5) cells by flow cytometry, which may be mediated by the induction of p21(WAF1/CIP1). The combination of LZP-F3 and arsenic trioxide produced a significant synergistic growth inhibition of NTUB1 and N/As(0.5) cells. Similar results were also found in N/P(14) cells. These molecular events of combined effects involved significant and earlier induction of Fas, caspase 3 and 8 activation, Bax and Bad up-regulation, Bcl-2 and Bcl-x(L) down-regulatuion, and cytochrome c release.
Collapse
Affiliation(s)
- Chao-Yuan Huang
- Department of Urology, National Taiwan University Hospital, and College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
154
|
Bruno T, Desantis A, Bossi G, Di Agostino S, Sorino C, De Nicola F, Iezzi S, Franchitto A, Benassi B, Galanti S, La Rosa F, Floridi A, Bellacosa A, Passananti C, Blandino G, Fanciulli M. Che-1 promotes tumor cell survival by sustaining mutant p53 transcription and inhibiting DNA damage response activation. Cancer Cell 2010; 18:122-34. [PMID: 20708154 DOI: 10.1016/j.ccr.2010.05.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 04/06/2010] [Accepted: 06/23/2010] [Indexed: 11/25/2022]
Abstract
Che-1 is a RNA polymerase II binding protein involved in the regulation of gene transcription and, in response to DNA damage, promotes p53 transcription. In this study, we investigated whether Che-1 regulates mutant p53 expression. We found that Che-1 is required for sustaining mutant p53 expression in several cancer cell lines, and that Che-1 depletion by siRNA induces apoptosis both in vitro and in vivo. Notably, loss of Che-1 activates DNA damage checkpoint response and induces transactivation of p73. Therefore, these findings underline the important role that Che-1 has in survival of cells expressing mutant p53.
Collapse
Affiliation(s)
- Tiziana Bruno
- Department of Therapeutic Programs Development, Regina Elena Cancer Institute, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
155
|
PBK/TOPK interacts with the DBD domain of tumor suppressor p53 and modulates expression of transcriptional targets including p21. Oncogene 2010; 29:5464-74. [DOI: 10.1038/onc.2010.275] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
156
|
Goebell PJ, Groshen SG, Schmitz-Dräger BJ. p53 immunohistochemistry in bladder cancer—a new approach to an old question. Urol Oncol 2010; 28:377-88. [DOI: 10.1016/j.urolonc.2010.03.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2010] [Revised: 03/30/2010] [Accepted: 03/31/2010] [Indexed: 10/19/2022]
|
157
|
|
158
|
Zhang J, Chen YH, Lu Q. Pro-oncogenic and anti-oncogenic pathways: opportunities and challenges of cancer therapy. Future Oncol 2010; 6:587-603. [PMID: 20373871 PMCID: PMC2886915 DOI: 10.2217/fon.10.15] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Carcinogenesis is the uncontrolled growth of cells gaining the potential to invade and disrupt vital tissue functions. This malignant process includes the occurrence of 'unwanted' gene mutations that induce the transformation of normal cells, for example, by overactivation of pro-oncogenic pathways and inactivation of tumor-suppressive or anti-oncogenic pathways. It is now recognized that the number of major signaling pathways that control oncogenesis is not unlimited; therefore, suppressing these pathways can conceivably lead to a cancer cure. However, the clinical application of cancer intervention has not matched up to scientific expectations. Increasing numbers of studies have revealed that many oncogenic-signaling elements show double faces, in which they can promote or suppress cancer pathogenesis depending on tissue type, cancer stage, gene dosage and their interaction with other players in carcinogenesis. This complexity of oncogenic signaling poses challenges to traditional cancer therapy and calls for considerable caution when designing an anticancer drug strategy. We propose future oncology interventions with the concept of integrative cancer therapy.
Collapse
Affiliation(s)
- Jiao Zhang
- Department of Anatomy & Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Yan-Hua Chen
- Department of Anatomy & Cell Biology, Leo Jenkins Cancer Center, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Qun Lu
- Associate Professor, Department of Anatomy & Cell Biology, Leo Jenkins Cancer Center, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA, Tel.: +1 252 744 2844, Fax: +1 252 744 2850,
| |
Collapse
|
159
|
Oxidative and nitrosative stress in the metastatic microenvironment. Cancers (Basel) 2010; 2:274-304. [PMID: 24281071 PMCID: PMC3835079 DOI: 10.3390/cancers2020274] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Revised: 03/02/2010] [Accepted: 03/25/2010] [Indexed: 12/17/2022] Open
Abstract
Metastases that are resistant to conventional therapies are the main cause of most cancer-related deaths in humans. Tumor cell heterogeneity, which associates with genomic and phenotypic instability, represents a major problem for cancer therapy. Additional factors, such as the attack of immune cells or organ-specific microenvironments, also influence metastatic cell behavior and the response to therapy. Interaction of cancer and endothelial cells in capillary beds, involving mechanical contact and transient adhesion, is a critical step in the initiation of metastasis. This interaction initiates a cascade of activation pathways that involves cytokines, growth factors, bioactive lipids and reactive oxygen and nitrogen species (ROS and RNS) produced by either the cancer cell or the endothelium. Vascular endothelium-derived NO and H2O2 are cytotoxic for the cancer cells, but also help to identify some critical molecular targets that appear essential for survival of invasive metastatic cell subsets. Surviving cancer cells that extravasate and start colonization of an organ or tissue can still be attacked by macrophages and be influenced by specific intraorgan microenvironment conditions. At all steps; from the primary tumor until colonization of a distant organ; metastatic cells undergo a dynamic process of constant adaptations that may lead to the survival of highly resistant malignant cell subsets. In this sequence of molecular events both ROS and RNS play key roles.
Collapse
|
160
|
Blanco-Aparicio C, Cañamero M, Cecilia Y, Pequeño B, Renner O, Ferrer I, Carnero A. Exploring the gain of function contribution of AKT to mammary tumorigenesis in mouse models. PLoS One 2010; 5:e9305. [PMID: 20174572 PMCID: PMC2824815 DOI: 10.1371/journal.pone.0009305] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 01/27/2010] [Indexed: 12/31/2022] Open
Abstract
Elevated expression of AKT has been noted in a significant percentage of primary human breast cancers, mainly as a consequence of the PTEN/PI3K pathway deregulation. To investigate the mechanistic basis of the AKT gain of function-dependent mechanisms of breast tumorigenesis, we explored the phenotype induced by activated AKT transgenes in a quantitative manner. We generated several transgenic mice lines expressing different levels of constitutively active AKT in the mammary gland. We thoroughly analyzed the preneoplastic and neoplastic mammary lesions of these mice and correlated the process of tumorigenesis to AKT levels. Finally, we analyzed the impact that a possible senescent checkpoint might have in the tumor promotion inhibition observed, crossing these lines to mammary specific p53(R172H) mutant expression, and to p27 knock-out mice. We analyzed the benign, premalignant and malignant lesions extensively by pathology and at molecular level analysing the expression of proteins involved in the PI3K/AKT pathway and in cellular senescence. Our findings revealed an increased preneoplastic phenotype depending upon AKT signaling which was not altered by p27 or p53 loss. However, p53 inactivation by R172H point mutation combined with myrAKT transgenic expression significantly increased the percentage and size of mammary carcinoma observed, but was not sufficient to promote full penetrance of the tumorigenic phenotype. Molecular analysis suggest that tumors from double myrAKT;p53(R172H) mice result from acceleration of initiated p53(R172H) tumors and not from bypass of AKT-induced oncogenic senescence. Our work suggests that tumors are not the consequence of the bypass of senescence in MIN. We also show that AKT-induced oncogenic senescence is dependent of pRb but not of p53. Finally, our work also suggests that the cooperation observed between mutant p53 and activated AKT is due to AKT-induced acceleration of mutant p53-induced tumors. Finally, our work shows that levels of activated AKT are not essential in the induction of benign or premalignant tumors, or in the cooperation of AKT with other tumorigenic signal such as mutant p53, once AKT pathway is activated, the relative level of activity seems not to determine the phenotype.
Collapse
Affiliation(s)
- Carmen Blanco-Aparicio
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Marta Cañamero
- Biotechnology Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Yolanda Cecilia
- Biotechnology Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Belén Pequeño
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Oliver Renner
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Irene Ferrer
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre, Madrid, Spain
| | - Amancio Carnero
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre, Madrid, Spain
- * E-mail:
| |
Collapse
|
161
|
Abstract
Currently, around 11 million people are living with a tumour that contains an inactivating mutation of TP53 (the human gene that encodes p53) and another 11 million have tumours in which the p53 pathway is partially abrogated through the inactivation of other signalling or effector components. The p53 pathway is therefore a prime target for new cancer drug development, and several original approaches to drug discovery that could have wide applications to drug development are being used. In one approach, molecules that activate p53 by blocking protein-protein interactions with MDM2 are in early clinical development. Remarkable progress has also been made in the development of p53-binding molecules that can rescue the function of certain p53 mutants. Finally, cell-based assays are being used to discover compounds that exploit the p53 pathway by either seeking targets and compounds that show synthetic lethality with TP53 mutations or by looking for non-genotoxic activators of the p53 response.
Collapse
|
162
|
Abstract
Cancers are rare because their evolution is actively restrained by a range of tumour suppressors. Of these p53 seems unusually crucial as either it or its attendant upstream or downstream pathways are inactivated in virtually all cancers. p53 is an evolutionarily ancient coordinator of metazoan stress responses. Its role in tumour suppression is likely to be a relatively recent adaptation, which is only necessary when large, long-lived organisms acquired the sufficient size and somatic regenerative capacity to necessitate specific mechanisms to reign in rogue proliferating cells. However, such evolutionary reappropriation of this venerable transcription factor entails compromises that restrict its efficacy as a tumour suppressor.
Collapse
Affiliation(s)
- Melissa R Junttila
- Department of Pathology and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco, 513 Parnassus Avenue, Room HSW-450A, UCSF Box 0502, San Francisco, California 94143-0502, USA
| | | |
Collapse
|
163
|
Abstract
Ample data indicate that mutant p53 proteins not only lose their tumour suppressive functions, but also gain new abilities that promote tumorigenesis. Moreover, recent studies have modified our view of mutant p53 proteins, portraying them not as inert mutants, but rather as regulated proteins that influence the cancer cell transcriptome and phenotype. This influence is clinically manifested as association of TP53 mutations with poor prognosis and drug resistance in a growing array of malignancies. Here, we review recent studies on mutant p53 regulation, gain-of-function mechanisms, transcriptional effects and prognostic association, with a focus on the clinical implications of these findings.
Collapse
Affiliation(s)
- Ran Brosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
164
|
Yacoubi-Hadj Amor I, Smaoui K, Belguith H, Djemal L, Dardouri M, Mokdad-Gargouri R, Gargouri A. Selection of cell death-deficient p53 mutants in Saccharomyces cerevisiae. Yeast 2009; 26:441-50. [PMID: 19579214 DOI: 10.1002/yea.1677] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The budding yeast Saccharomyces cerevisiae is a useful system for the detection and transcriptional evaluation of mutant p53 in cancer. In previous work we showed that the overexpression of wild-type p53 induces yeast cell death on minimal medium; however, the R248W p53 mutant was completely inactive, and we suggested that ROS production is a key event in p53-induced yeast cell death. In this study we explored the effect of other p53 mutants, such as the hot-spot mutant R282W and the double mutant N268S::I332V. Unexpectedly, both mutants behaved inversely to R248W, as they completely inhibited yeast growth on minimal medium and induced ROS production. This phenotype 'yeast cell death on minimal medium' allowed for the subsequent screening of intragenic p53-inactivating mutations. In all cases, the 'revertant yeast clones' display a complete p53 inactivation through either gross deletion or nonsense mutations. More interestingly, missense mutations were also found: the deletion of I255 or substitution of R337G completely inactivated the p53 mutant R282W in the yeast context. Taken together, these results suggest that p53 tumour-derived mutants could be classified according to their ability to induce yeast cell death and not uniquely by their transcriptional activity on a selected target reporter gene.
Collapse
|
165
|
Haupt S, di Agostino S, Mizrahi I, Alsheich-Bartok O, Voorhoeve M, Damalas A, Blandino G, Haupt Y. Promyelocytic leukemia protein is required for gain of function by mutant p53. Cancer Res 2009; 69:4818-26. [PMID: 19487292 DOI: 10.1158/0008-5472.can-08-4010] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mutations in the p53 tumor suppressor are the most common genetic events in human cancer. These mutations not only result in a loss of wild-type p53 activity, but can also lead to a gain of new oncogenic properties. Understanding how these gained functions are regulated is in its infancy. In this study, we show that the promyelocytic leukemia (PML) protein is an important regulator of mutant p53. We show that PML interacts with mutant p53. Importantly, PML enhances the transcriptional activity of mutant p53. Unexpectedly, PML is required for the proliferation and colony formation of cancer cells bearing mutant p53. Down-regulation of PML expression inhibits the growth of mutant p53-expressing cancer cells, predominantly by promoting cell cycle arrest. Our results suggest that the tumor suppression function of PML depends on the status of p53. In the context of mutant p53, PML enhances its cancer-promoting activities.
Collapse
Affiliation(s)
- Sue Haupt
- Research Division, The Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
166
|
Lavra L, Ulivieri A, Rinaldo C, Dominici R, Volante M, Luciani E, Bartolazzi A, Frasca F, Soddu S, Sciacchitano S. Gal-3 is stimulated by gain-of-function p53 mutations and modulates chemoresistance in anaplastic thyroid carcinomas. J Pathol 2009; 218:66-75. [PMID: 19199318 DOI: 10.1002/path.2510] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Galectin-3 (Gal-3) is an anti-apoptotic molecule of the beta-galactoside-binding lectin family. Gal-3 is down-regulated by wt-p53 and this repression is required for p53-induced apoptosis. Since poorly differentiated thyroid carcinomas (PDTCs) and anaplastic thyroid carcinomas (ATCs) frequently harbour p53 mutations, we asked whether Gal-3 expression and activity could be influenced by such mutations in these tumours. We found a positive correlation between Gal-3 expression and p53 mutation in human thyroids and in thyroid carcinoma cell lines (TCCLs) harbouring different p53 mutations. Gal-3 was over-expressed in most ATCs and TCCLs, especially those with the most frequently detected p53 mutation (p53(R273H)). Over-expression of p53(R273H) in two p53-null cells (SAOS-2 and SW-1736) as well as in two wt-p53-carrying TCCLs (TPC-1 and K1), stimulated Gal-3 expression, while interference with p53(R273H) endogenous expression in ARO cells down-regulated Gal-3 expression. Conversely, over-expression of wt-p53 in ARO cells restored the inhibitory effect on Gal-3 expression. ARO cells are highly resistant to apoptosis and express both p53 and Gal-3, which are increased upon cisplatin treatment. Interference with Gal-3 expression in these cells stimulated their chemosensitivity. In conclusion, gain-of-function p53 mutant acquires the de novo ability to stimulate Gal-3 expression and to increase chemoresistance in ATCs.
Collapse
Affiliation(s)
- Luca Lavra
- S. Pietro Fatebenefratelli Hospital, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Bonuccelli G, Casimiro MC, Sotgia F, Wang C, Liu M, Katiyar S, Zhou J, Dew E, Capozza F, Daumer KM, Minetti C, Milliman JN, Alpy F, Rio MC, Tomasetto C, Mercier I, Flomenberg N, Frank PG, Pestell RG, Lisanti MP. Caveolin-1 (P132L), a common breast cancer mutation, confers mammary cell invasiveness and defines a novel stem cell/metastasis-associated gene signature. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1650-62. [PMID: 19395651 DOI: 10.2353/ajpath.2009.080648] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Here we used the Met-1 cell line in an orthotopic transplantation model in FVB/N mice to dissect the role of the Cav-1(P132L) mutation in human breast cancer. Identical experiments were performed in parallel with wild-type Cav-1. Cav-1(P132L) up-regulated the expression of estrogen receptor-alpha as predicted, because only estrogen receptor-alpha-positive patients have been shown to harbor Cav-1(P132L) mutations. In the context of primary tumor formation, Cav-1(P132L) behaved as a loss-of-function mutation, lacking any tumor suppressor activity. In contrast, Cav-1(P132L) caused significant increases in cell migration, invasion, and experimental metastasis, consistent with a gain-of-function mutation. To identify possible molecular mechanism(s) underlying this invasive gain-of-function activity, we performed unbiased gene expression profiling. From this analysis, we show that the Cav-1(P132L) expression signature contains numerous genes that have been previously associated with cell migration, invasion, and metastasis. These include i) secreted growth factors and extracellular matrix proteins (Cyr61, Plf, Pthlh, Serpinb5, Tnc, and Wnt10a), ii) proteases that generate EGF and HGF (Adamts1 and St14), and iii) tyrosine kinase substrates and integrin signaling/adapter proteins (Akap13, Cdcp1, Ddef1, Eps15, Foxf1a, Gab2, Hs2st1, and Itgb4). Several of the P132L-specific genes are also highly expressed in stem/progenitor cells or are associated with myoepithelial cells, suggestive of an epithelial-mesenchymal transition. These results directly support clinical data showing that patients harboring Cav-1 mutations are more likely to undergo recurrence and metastasis.
Collapse
Affiliation(s)
- Gloria Bonuccelli
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Cheng C, Li LM, Alves P, Gerstein M. Systematic identification of transcription factors associated with patient survival in cancers. BMC Genomics 2009; 10:225. [PMID: 19442316 PMCID: PMC2686740 DOI: 10.1186/1471-2164-10-225] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 05/15/2009] [Indexed: 12/23/2022] Open
Abstract
Background Aberrant activation or expression of transcription factors has been implicated in the tumorigenesis of various types of cancer. In spite of the prevalent application of microarray experiments for profiling gene expression in cancer samples, they provide limited information regarding the activities of transcription factors. However, the association between transcription factors and cancers is largely dependent on the transcription regulatory activities rather than mRNA expression levels. Results In this paper, we propose a computational approach that integrates microarray expression data with the transcription factor binding site information to systematically identify transcription factors associated with patient survival given a specific cancer type. This approach was applied to two gene expression data sets for breast cancer and acute myeloid leukemia. We found that two transcription factor families, the steroid nuclear receptor family and the ATF/CREB family, are significantly correlated with the survival of patients with breast cancer; and that a transcription factor named T-cell acute lymphocytic leukemia 1 is significantly correlated with acute myeloid leukemia patient survival. Conclusion Our analysis identifies transcription factors associating with patient survival and provides insight into the regulatory mechanism underlying the breast cancer and leukemia. The transcription factors identified by our method are biologically meaningful and consistent with prior knowledge. As an insightful tool, this approach can also be applied to other microarray cancer data sets to help researchers better understand the intricate relationship between transcription factors and diseases.
Collapse
Affiliation(s)
- Chao Cheng
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
| | | | | | | |
Collapse
|
169
|
Khromova NV, Kopnin PB, Stepanova EV, Agapova LS, Kopnin BP. p53 hot-spot mutants increase tumor vascularization via ROS-mediated activation of the HIF1/VEGF-A pathway. Cancer Lett 2008; 276:143-51. [PMID: 19091459 DOI: 10.1016/j.canlet.2008.10.049] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 05/13/2008] [Accepted: 10/30/2008] [Indexed: 12/11/2022]
Abstract
The function of p53 tumor suppressor is often altered in various human tumors predominantly through missense-mutations resulting in accumulation of mutant proteins. We revealed that expression of p53 proteins with amino-acid substitutions at codons 175 (R175H), 248 (R248W), and 273 (R273H), representing the hot-spots of mutations in various human tumors, increased the number of vessels in HCT116 human colon carcinoma xenografts and, as a result, accelerated their growth. Stimulation of tumor angiogenesis was connected with about 2-fold increase in intracellular level of reactive oxygen species (ROS). Antioxidant N-acetyl-l-aspartate (NAC) decreased vessels number in tumors formed by cells with inactivated p53 and inhibited their growth. Effect of ROS on angiogenesis in tumors expressing hot-spot p53 mutants was correlated with their ability to increase a content of HIF1 transcriptional factor responsible for up-regulation of VEGF-A mRNAs.
Collapse
Affiliation(s)
- N V Khromova
- Blokhin Memorial Russian Cancer Research Center, Kashirskoye shosse 24, Moscow, Russia
| | | | | | | | | |
Collapse
|
170
|
Wen J, Zhu X, Liu B, You L, Kong L, Lee HI, Han KP, Wong JL, Rhode PR, Wong HC. Targeting activity of a TCR/IL-2 fusion protein against established tumors. Cancer Immunol Immunother 2008; 57:1781-94. [PMID: 18369620 PMCID: PMC11031049 DOI: 10.1007/s00262-008-0504-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 03/07/2008] [Indexed: 01/10/2023]
Abstract
We have previously reported that a single-chain T cell receptor/IL-2 fusion protein (scTCR-IL2) exhibits potent targeted antitumor activity in nude mice bearing human tumor xenografts that display cognate peptide/HLA complexes. In this study, we further explore the mechanism of action of this molecule. We compared the biological activities of c264scTCR-IL2, a scTCR-IL2 protein recognizing the aa264-272 peptide of human p53, with that of MART-1scTCR-IL2, which recognizes the MART-1 melanoma antigen (aa27-35). In vitro studies showed that c264scTCR-IL2 and MART-1scTCR-IL2 were equivalent in their ability to bind cell-surface IL-2 receptors and stimulate NK cell responses. In mice, MART-1scTCR-IL2 was found to have a twofold longer serum half-life than c264scTCR-IL2. However, despite its shorter serum half-life, c264scTCR-IL2 showed significantly better antitumor activity than MART-1scTCR-IL2 against p53(+)/HLA-A2(+) tumor xenografts. The more potent antitumor activity of c264scTCR-IL2 correlated with an enhanced capacity to promote NK cell infiltration into tumors. Similar differences in antigen-dependent tumor infiltration were observed with activated splenocytes pre-treated in vitro with c264scTCR-IL2 or MART-1scTCR-IL2 and then transferred into p53(+)/HLA-A2(+) tumor bearing recipients. The data support a model where c264scTCR-IL2 activates immune cells to express IL-2 receptors. Following stable interactions with cell-surface IL-2 receptors, c264scTCR-IL2 fusion molecule enhances the trafficking of immune cells to tumors displaying target peptide/HLA complexes where the immune cells mediate antitumor effects. Thus, this type of fusion molecule could be used directly as a targeted immunotherapeutic or in adoptive cell transfer approaches to activate and improve the anti-cancer activities of immune cells by providing them with pre-selected antigen recognition capability.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Female
- Flow Cytometry
- Half-Life
- Humans
- Immunohistochemistry
- Immunotherapy/methods
- Interleukin-2/immunology
- Interleukin-2/metabolism
- Interleukin-2/therapeutic use
- Killer Cells, Natural/immunology
- Lymphocyte Activation/immunology
- MART-1 Antigen
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Mice
- Mice, Nude
- Neoplasm Proteins/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/therapeutic use
- Recombinant Fusion Proteins/pharmacology
- Tumor Suppressor Protein p53/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jinghai Wen
- Altor Bioscience Corporation, 2810 N Commerce Parkway, Miramar, FL 33025 USA
| | - Xiaoyun Zhu
- Altor Bioscience Corporation, 2810 N Commerce Parkway, Miramar, FL 33025 USA
| | - Bai Liu
- Altor Bioscience Corporation, 2810 N Commerce Parkway, Miramar, FL 33025 USA
| | - Lijing You
- Altor Bioscience Corporation, 2810 N Commerce Parkway, Miramar, FL 33025 USA
| | - Lin Kong
- Altor Bioscience Corporation, 2810 N Commerce Parkway, Miramar, FL 33025 USA
| | - Hyung-il Lee
- Altor Bioscience Corporation, 2810 N Commerce Parkway, Miramar, FL 33025 USA
| | - Kai-ping Han
- Altor Bioscience Corporation, 2810 N Commerce Parkway, Miramar, FL 33025 USA
| | - Jeffrey L. Wong
- University of Pittsburgh Medical Center, Pittsburgh, PA 15232 USA
| | - Peter R. Rhode
- Altor Bioscience Corporation, 2810 N Commerce Parkway, Miramar, FL 33025 USA
| | - Hing C. Wong
- Altor Bioscience Corporation, 2810 N Commerce Parkway, Miramar, FL 33025 USA
| |
Collapse
|
171
|
Faria MHG, Patrocínio RMDSVD, Moraes Filho MOD, Rabenhorst SHB. Immunoexpression of tumor suppressor genes p53, p21 WAF1/CIP1 and p27 KIP1 in humam astrocystic tumors. ARQUIVOS DE NEURO-PSIQUIATRIA 2008; 65:1114-22. [PMID: 18345413 DOI: 10.1590/s0004-282x2007000700004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 10/06/2007] [Indexed: 11/22/2022]
Abstract
The aim of the present study was to evaluate the tumor suppressor genes p53, p21 WAF1/CIP1 and p27 KIP1 expression in astrocytic tumors, correlating the findings with the histopathological grade (WHO). An immunohistochemical study of the p53, p21 and p27 proteins using the streptavidin-biotin-peroxidase method was performed in fifty-five astrocytomas (13 grade I, 14 grade II, 7 grade III and 21 grade IV) and five samples of non-tumor brain tissue (negative control). p53 positive indices (PI) and labeling indices (LI) showed tendency to increase according to malignant progression. The nuclear expression of p27 presented similar inclination, except for the PI reduction verified in grade IV tumors. Otherwise, the cytoplasmic p27 staining was more evident between high-grade tumors (III and IV). p53 and nuclear p27 expression was correlated with the histological classification (p<0.01; test H). On the other hand, p21 indices revealed a propensity to reduction in agreement with malignant evolution of the astrocytic tumors, except for high scores observed in grade IV tumors. The non-tumor samples did not show any expression of these proteins. These results indicated the p53 mutation as an initial, relevant and potentially predictor of tumor progression event in astrocytomas, with the detection of p21 protein as an important resource for the deduction of functional situation of this gene. Moreover, the activation of p27 KIP1 was preserved in the astrocytic tumors and its cytoplasmic manifestation seems to be resultant of its nuclear expression, not demonstrating a direct impact in astrocytomas tumorigenesis.
Collapse
Affiliation(s)
- Mário Henrique Girão Faria
- Department of Physiology and Pharmacology, Molecular Genetics Laboratory, School of Medicine, Federal University of Ceará, Fortaleza CE, Brazil.
| | | | | | | |
Collapse
|
172
|
Chen KF, Yeh PY, Yeh KH, Lu YS, Huang SY, Cheng AL. Down-regulation of phospho-Akt is a major molecular determinant of bortezomib-induced apoptosis in hepatocellular carcinoma cells. Cancer Res 2008; 68:6698-707. [PMID: 18701494 DOI: 10.1158/0008-5472.can-08-0257] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bortezomib, a proteasome inhibitor, has been clinically approved for the treatment of myeloma and lymphoma. Here, we report a differential effect of bortezomib on apoptosis in four hepatocellular carcinoma (HCC) cell lines and identify the major molecular event that determines sensitivity. Although bortezomib inhibited proteasome activity to a similar extent in all HCC cell lines, it showed differential effects on their viability: Huh-7 (IC(50) 196 nmol/L), Sk-Hep1 (IC(50) 180 nmol/L), Hep3B (IC(50) 112 nmol/L), and resistant PLC5 (IC(50) >1,000 nmol/L). Bortezomib caused cell cycle arrest at G(2)-M phase in all HCC cells tested whereas apoptotic induction was found only in sensitive cells but not in PLC5 cells. No significant bortezomib-induced NF-kappaB changes were noted in Huh-7 and PLC5. Bortezomib down-regulated phospho-Akt (P-Akt) in a dose- and time-dependent manner in all sensitive HCC cells whereas no alterations of P-Akt were found in PLC5. Down-regulation of Akt1 by small interference RNA overcame the apoptotic resistance to bortezomib in PLC5 cells, but a constitutively activated Akt1 protected Huh-7 cells from bortezomib-induced apoptosis. Furthermore, bortezomib showed suppression of tumor growth with down-regulation of P-Akt in Huh-7 tumors but not in PLC5 tumors. Down-regulation of P-Akt represents a major molecular event of bortezomib-induced apoptosis in HCC cell lines and may be a biomarker for predicting clinical response to HCC treatment. Targeting Akt signaling overcomes drug resistance to bortezomib in HCC cells, which provides a new approach for the combinational therapy of HCC.
Collapse
Affiliation(s)
- Kuen-Feng Chen
- Department of Medical Research, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei
| | | | | | | | | | | |
Collapse
|
173
|
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]
|
174
|
Bajgelman MC, Strauss BE. Development of an adenoviral vector with robust expression driven by p53. Virology 2008; 371:8-13. [PMID: 18076963 DOI: 10.1016/j.virol.2007.11.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 10/23/2007] [Accepted: 11/14/2007] [Indexed: 11/26/2022]
Abstract
Here we introduce a new adenoviral vector where transgene expression is driven by p53. We first developed a synthetic promoter, referred to as PGTxbeta, containing a p53-responsive element, a minimal promoter and the first intron of the rabbit beta-globin gene. Initial assays using plasmid-based vectors indicated that expression was tightly controlled by p53 and was 5-fold stronger than the constitutive CMV immediate early promoter/enhancer. The adenoviral vector, AdPG, was also shown to offer p53-responsive expression in prostate carcinoma cells LNCaP (wt p53), DU-145 (temperature sensitive mutant of p53) and PC3 (p53-null, but engineered to express temperature-sensitive p53 mutants). AdPG served as a sensor of p53 activity in LNCaP cells treated with chemotherapeutic agents. Since p53 can be induced by radiotherapy and chemotherapy, this new vector could be further developed for use in combination with conventional therapies to bring about cooperation between the genetic and pharmacologic treatment modalities.
Collapse
Affiliation(s)
- Marcio C Bajgelman
- Viral Vector Group, Laboratory of Genetics and Molecular Cardiology/LIM 13, Heart Institute (InCor), University of Sao Paulo School of Medicine, Brazil
| | | |
Collapse
|
175
|
Crispen PL, Boorjian SA, Lohse CM, Leibovich BC, Kwon ED. Predicting disease progression after nephrectomy for localized renal cell carcinoma: the utility of prognostic models and molecular biomarkers. Cancer 2008; 113:450-60. [PMID: 18523999 PMCID: PMC2785036 DOI: 10.1002/cncr.23566] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Disease progression after nephrectomy for pathologically localized renal cell carcinoma (RCC) is associated with a significant mortality rate, given the limited efficacy of available treatment regimens for metastatic disease. As such, several adjuvant trials have been designed to treat patients at particularly high risk for postsurgical RCC progression. Several different prognostic models designed to identify patients at high risk of disease progression are available. Although these available predictive models provide a reasonable assessment of patients' risks of disease progression, the accuracy of these models may further be improved via the incorporation of molecular prognostic biomarkers. Although numerous candidate molecules have been described, few have been specifically assessed for the association with disease progression after nephrectomy. IMP-3, CXCR3, p53, Survivin, cIAP1, B7-H1, and B7-H4 have all been associated with disease progression after nephrectomy. The incorporation of 1 or several of these biomarkers may increase the accuracy of currently available prognostic models and thereby facilitate the appropriate use of adjuvant therapies aimed at preventing future disease progression. As such, the authors review the current prognostic tools for predicting disease progression for localized RCC, and detail studies to date that have evaluated various biomarkers in this setting.
Collapse
Affiliation(s)
| | | | - Christine M. Lohse
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Eugene D. Kwon
- Department of Urology, Mayo Clinic, Rochester, Minnesota
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
176
|
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
|
177
|
Abstract
Inactivation of tumor suppressor gene is a key event in carcinogenesis. p53 is one of the most important tumor suppressor genes in the genome, and its mutations are found in approximately 50% of human cancers. p53 mutation is also the main cause for human Li-Fraumeni syndrome. The vast majority of p53 mutations are missense mutations, and the corresponding mutant p53 proteins not only lose wild-type p53 tumor suppressor activities, but also gain new oncogenic properties favoring cancer development. Here, we mainly discussed the structural and functional alterations of mutant p53, the molecular mechanisms underlying gain of oncogenic functions, and the strategies and explorations of suppressing mutant p53 activities.
Collapse
|
178
|
Li Q, Wang G. Research progress of the relationship between microRNAs and p53 gene in oncogenesis. Shijie Huaren Xiaohua Zazhi 2008; 16:1332-1336. [DOI: 10.11569/wcjd.v16.i12.1332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [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
The initiation and development of oncogenesis are a multi-step and complicated process, in which activation of oncogenes and inactivation of tumor suppressor genes are involved. MicroRNAs (miRNAs) are a new class of endogenous, non-coding small RNA molecules. It has been demonstrated that their expression levels are closely associated with human pathogenesis of cancers. They may participate in regulating the abnormal expression of oncogenes and tumor suppressor genes. Mutation in tumor suppressor gene p53 is the most frequent phenomenon in human cancer, and up to now, almost 50% human cancers are demonstrated associated with p53 mutation. Recent studies showed that miRNAs might play a role in regulating the tumor-suppressor activity of p53 gene. In this review, the research progress in this field is discussed.
Collapse
|
179
|
Cuddihy AR, Jalali F, Coackley C, Bristow RG. WTp53 induction does not override MTp53 chemoresistance and radioresistance due to gain-of-function in lung cancer cells. Mol Cancer Ther 2008; 7:980-92. [DOI: 10.1158/1535-7163.mct-07-0471] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
180
|
Grove CA, Walhout AJM. Transcription factor functionality and transcription regulatory networks. MOLECULAR BIOSYSTEMS 2008; 4:309-14. [PMID: 18354784 PMCID: PMC2673723 DOI: 10.1039/b715909a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Now that numerous high-quality complete genome sequences are available, many efforts are focusing on the "second genomic code", namely the code that determines how the precise temporal and spatial expression of each gene in the genome is achieved. In this regard, the elucidation of transcription regulatory networks that describe combined transcriptional circuits for an organism of interest has become valuable to our understanding of gene expression at a systems level. Such networks describe physical and regulatory interactions between transcription factors (TFs) and the target genes they regulate under different developmental, physiological, or pathological conditions. The mapping of high-quality transcription regulatory networks depends not only on the accuracy of the experimental or computational method chosen, but also relies on the quality of TF predictions. Moreover, the total repertoire of TFs is not only determined by the protein-coding capacity of the genome, but also by different protein properties, including dimerization, co-factor interactions and post-translational modifications. Here, we discuss the factors that influence TF functionality and, hence, the functionality of the networks in which they operate.
Collapse
Affiliation(s)
- Christian A. Grove
- Program in Gene Function and Expression and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA. E-mail:
| | - Albertha J. M. Walhout
- Program in Gene Function and Expression and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA. E-mail:
| |
Collapse
|
181
|
|
182
|
Benjamin JM, Nelson WJ. Bench to bedside and back again: molecular mechanisms of alpha-catenin function and roles in tumorigenesis. Semin Cancer Biol 2008; 18:53-64. [PMID: 17945508 PMCID: PMC2692220 DOI: 10.1016/j.semcancer.2007.08.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2007] [Accepted: 08/28/2007] [Indexed: 12/17/2022]
Abstract
The cadherin/catenin complex, comprised of E-cadherin, beta-catenin and alpha-catenin, is essential for initiating cell-cell adhesion, establishing cellular polarity and maintaining tissue organization. Disruption or loss of the cadherin/catenin complex is common in cancer. As the primary cell-cell adhesion protein in epithelial cells, E-cadherin has long been studied in cancer progression. Similarly, additional roles for beta-catenin in the Wnt signaling pathway has led to many studies of the role of beta-catenin in cancer. Alpha-catenin, in contrast, has received less attention. However, recent data demonstrate novel functions for alpha-catenin in regulating the actin cytoskeleton and cell-cell adhesion, which when perturbed could contribute to cancer progression. In this review, we use cancer data to evaluate molecular models of alpha-catenin function, from the canonical role of alpha-catenin in cell-cell adhesion to non-canonical roles identified following conditional alpha-catenin deletion. This analysis identifies alpha-catenin as a prognostic factor in cancer progression.
Collapse
Affiliation(s)
- Jacqueline M. Benjamin
- Departments of Biological Sciences and Molecular and Cellular Physiology, Stanford University, Stanford CA 94305
| | - W. James Nelson
- Departments of Biological Sciences and Molecular and Cellular Physiology, Stanford University, Stanford CA 94305
| |
Collapse
|
183
|
Ehrhardt H, Häcker S, Wittmann S, Maurer M, Borkhardt A, Toloczko A, Debatin KM, Fulda S, Jeremias I. Cytotoxic drug-induced, p53-mediated upregulation of caspase-8 in tumor cells. Oncogene 2008; 27:783-93. [PMID: 17637740 DOI: 10.1038/sj.onc.1210666] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2007] [Revised: 06/06/2007] [Accepted: 06/11/2007] [Indexed: 11/08/2022]
Abstract
Apoptosis resistance is crucially involved in cancer development and progression, represents the leading cause for failure of anticancer therapy and is caused, for example, by downregulation of proapoptotic intracellular signaling molecules such as caspase-8. We found that the cytotoxic drugs methotrexate (MTX) and 5-fluorouracil (5-FU) were both able to sensitize resistant tumor cells for induction of apoptosis by p53-mediated upregulation of caspase-8. Increase in caspase-8 messenger RNA and protein expression disabled tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced proliferation and restored sensitivity toward TRAIL-induced apoptosis which was inhibited by transfection of p53 decoy oligonucleotides, p53 shRNA and caspase-8 shRNA. Upregulation of caspase-8 and sensitization toward TRAIL-induced apoptosis was found both in a broad panel of tumor cell lines with downregulated caspase-8 and in TRAIL-resistant primary tumor cells of children with acute leukemia. Taken together, we have identified caspase-8 as an important p53 target gene regulated by cytotoxic drugs. These findings highlight a new drug-induced modulation of physiological apoptosis pathways, which may be involved in successful anticancer therapy using MTX and 5-FU in leukemia and solid tumors over decades.
Collapse
Affiliation(s)
- H Ehrhardt
- Department of Oncology/Hematology, Dr von Haunersches Kinderspital, München, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Supiot S, Zhao H, Wiman K, Hill RP, Bristow RG. PRIMA-1(met) radiosensitizes prostate cancer cells independent of their MTp53-status. Radiother Oncol 2008; 86:407-11. [PMID: 18237796 DOI: 10.1016/j.radonc.2008.01.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Revised: 12/21/2007] [Accepted: 01/03/2008] [Indexed: 10/22/2022]
Abstract
The novel agent PRIMA-1(met) can reactivate WTp53 function in MTp53-expressing cells. We investigated PRIMA-1(met) as a radiosensitizer of WTp53, p53Null or MTp53 prostate cancer cells. Radiosensitization was observed in PC3 (p53Null) cells, even under hypoxia. In certain circumstances, PRIMA-1(met) may therefore act independently of MTp53 status and WTp53 transactivation.
Collapse
Affiliation(s)
- Stéphane Supiot
- Princess Margaret Hospital (University Health Network), Toronto, Canada
| | | | | | | | | |
Collapse
|
185
|
|
186
|
Real FX. p53: it has it all, but will it make it to the clinic as a marker in bladder cancer? J Clin Oncol 2007; 25:5341-4. [PMID: 18048811 DOI: 10.1200/jco.2007.13.1904] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
187
|
Abstract
Since the very early days of p53 research, the gain of oncogenic activities by some mutant p53 proteins had been suspected as an important factor contributing to cancer progression. Considerable progress towards understanding the biology of mutant p53 has been made during the last years, the quintessence being the realization that the impact of mutant p53 proteins on the transcriptome of a tumor cell is much more global than previously thought. The emerging role of mutant p53 proteins in coordinating oncogenic signaling and chromatin modifying activities reveals an until now unsuspected function of these proteins as important modifiers of the oncogenic transcriptional response. Notwithstanding the fact that the sequence-specific DNA binding activity of mutant p53 proteins is impaired, they are still able to associate with specific loci on DNA by utilizing different mechanisms. The ability to associate with DNA appears to be crucial for the master role of mutant p53 proteins in coordinating oncogenic transcriptional responses.
Collapse
Affiliation(s)
- E Kim
- Translational Neuro-Oncology Group, Department of Neurosurgery, Georg-August-University of Goettingen, Robert-Koch-Strasse 40, Goettingen, Germany.
| | | |
Collapse
|
188
|
Watanabe T, Tobe K, Nakachi Y, Kondoh Y, Nakajima M, Hamada S, Namiki C, Suzuki T, Maeda S, Tadakuma A, Sakurai M, Arai Y, Hyogo A, Hoshino M, Tashiro T, Ito H, Inazumi H, Sakaki Y, Tashiro H, Furihata C. Differential Gene Expression Induced by Two Genotoxic N-nitroso Carcinogens, Phenobarbital and Ethanol in Mouse Liver Examined with Oligonucleotide Microarray and Quantitative Real-time PCR. Genes Environ 2007. [DOI: 10.3123/jemsge.29.115] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|