201
|
Smith SJ, Neugut A, Heitjan D, Forde K, Holt P, Santella RM, Jiin-Chyuan L, Carney W, Ward L, Brandt-Rauf PW. In situ quantification of aberrant p53 in colorectal neoplasia. Biomarkers 2003; 8:311-32. [PMID: 12944180 DOI: 10.1080/1354750031000138676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Aberrant p53 protein accumulation was measured immunohistologically in 342 colorectal paraffin-embedded tissue sections from 115 patients (24 with adenocarcinoma, 59 with adenoma and 32 'hospital controls'). Subjective scoring was compared with quantitative cell imaging, including dichotomous (p53+/p53-) status, ng p53mut mg(-1) enterocyte protein, and tumour burden and patient body 'burden' of aberrant p53. A total of 62.5% cancer patients, 23.7% adenoma patients and 3.1% hospital controls were accorded p53+ status on the basis of p53 quantification. Quantitative p53+/p53- assignment had a stronger inverse association with survival (chi2=6.17, p=0.013, Kaplan-Meier test) than subjective 'visual estimation' (chi2=0.57, p=0.449). There was a strong inverse relationship between the p53 'body burden' and the months of post-diagnosis survival (hazard ratio=1.42, p=0.0004, Cox proportional hazards). Absolute quantification for inactivated p53 permits objective and reproducible scoring, adjusts for intra-laboratory immunostaining 'batch effects', corrects for fixation artefacts, and standardizes for inter-laboratory differences in fixation, antibody selection and staining method. Clinically, in situ quantification of p53 will permit more accurate survival prognoses and will inform therapy selection and dose. Ultimately, accurate quantitative tissue/blood p53 correlations may provide a minimally invasive and systemic surrogate measure for these same clinical purposes.
Collapse
Affiliation(s)
- Steven Jay Smith
- VA-New York Harbor Health Care System, Brooklyn, New York 11209, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Chowdhury IH, Radonovich M, Mahieux R, Pise-Masison C, Muralidhar S, Brady JN. P53 facilitates degradation of human T-cell leukaemia virus type I Tax-binding protein through a proteasome-dependent pathway. J Gen Virol 2003; 84:897-906. [PMID: 12655090 DOI: 10.1099/vir.0.18753-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Human T-cell leukaemia virus type 1 (HTLV-I), the aetiological agent of adult T-cell leukaemia (ATL) and tropical spastic paraparesis (TSP/HAM), transforms human T-cells in vivo and in vitro. The Tax protein of HTLV-I is essential for cellular transformation as well as viral and cellular gene transactivation. The interaction of Tax with cellular proteins is critical for these functions. We previously isolated and characterized a novel Tax-binding protein, TRX (TAX1BP2), by screening a Jurkat T-cell cDNA library. In the present study, we present evidence that the tumour suppressor p53 targets the TRX protein for proteasome degradation. Pulse-chase experiments revealed that p53 enhanced the degradation of TRX protein and reduced the half-life from 2.0 to 0.25 h. p53 mutants R248W and R273H enhance TRX degradation suggesting a transcriptionally independent mechanism. Both HTLV-I Tax and the proteasome-specific inhibitor MG132 inhibited p53-mediated TRX protein degradation. These results suggest that TRX degradation is mediated through activation of the proteasome protein degradation pathway independent of transcriptional function of p53. Our results provide the first experimental evidence that Tax inhibits transcription-dependent and independent functions of p53.
Collapse
Affiliation(s)
- Iqbal H Chowdhury
- Virus Tumor Biology Section, Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Building 41/B201, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Michael Radonovich
- Virus Tumor Biology Section, Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Building 41/B201, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Renaud Mahieux
- Institut Pasteur, 28 rue du Docteur Roux, 75724 Paris cedex 15, France
- Virus Tumor Biology Section, Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Building 41/B201, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Cynthia Pise-Masison
- Virus Tumor Biology Section, Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Building 41/B201, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Sumitra Muralidhar
- Virus Tumor Biology Section, Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Building 41/B201, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - John N Brady
- Virus Tumor Biology Section, Basic Research Laboratory, National Cancer Institute, National Institutes of Health, Building 41/B201, 9000 Rockville Pike, Bethesda, MD 20892, USA
| |
Collapse
|
203
|
Joshi AA, Wu Z, Reed RF, Suttle DP. Nuclear factor-Y binding to the topoisomerase IIalpha promoter is inhibited by both the p53 tumor suppressor and anticancer drugs. Mol Pharmacol 2003; 63:359-67. [PMID: 12527807 DOI: 10.1124/mol.63.2.359] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Expression of the human DNA topoisomerase IIalpha (topo IIalpha) gene is positively regulated by the binding of the nuclear factor Y (NF-Y) transcription factor to four of five inverted CCAAT boxes (ICBs) located in its promoter. We have demonstrated previously that expression of the p53 tumor suppressor inhibits human topo IIalpha promoter activity in murine (10)1 cells. In this report, we demonstrate that the inhibition of topo IIalpha gene expression by wild-type p53 correlates with the decreased binding of the transcription factor NF-Y to the first four ICBs of the topo IIalpha promoter. The expression of mutant p53 does not affect the binding of NF-Y. In NIH3T3 cells, we show that topo II-targeted drugs inhibit the binding of NF-Y to ICB sites in the topo IIalpha promoter. This effect is seen not only with drugs that result in DNA strand breaks but also with drugs that inhibit the catalytic activity of topo II, and even with the mitotic spindle inhibitor, vinblastine. Further experiments with p53-null (10)1 cells treated with these same drugs also demonstrate decreased NF-Y binding to the topo IIalpha ICBs. The data presented points to the existence of both p53-dependent and -independent mechanisms for regulating NF-Y binding to ICBs in the topo IIalpha promoter and thus the modulation of topo IIalpha gene expression.
Collapse
Affiliation(s)
- Ashish A Joshi
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | |
Collapse
|
204
|
Calpena R, Javier Lacueva F, Oliver I, Cansado P, Pérez F, Costa D, Compañ AF, Teresa Pérez M. Estado actual del tratamiento multidisciplinario del cáncer gástrico avanzado. Cir Esp 2003. [DOI: 10.1016/s0009-739x(03)72191-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
205
|
Hait WN, Rubin E, Goodin S. Tubulin-targeting agents. CANCER CHEMOTHERAPY AND BIOLOGICAL RESPONSE MODIFIERS 2003; 21:41-67. [PMID: 15338740 DOI: 10.1016/s0921-4410(03)21003-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- William N Hait
- UMDNJ/Robert Wood Johnson Medical School, Department of Medicine, New Brunswick, NJ 08901-1914, USA.
| | | | | |
Collapse
|
206
|
Lee CH, Ling V. Superinduction of P-glycoprotein messenger RNA in vivo in the presence of transcriptional inhibitors. JOURNAL OF EXPERIMENTAL THERAPEUTICS AND ONCOLOGY 2003; 3:14-26. [PMID: 12724855 DOI: 10.1046/j.1359-4117.2003.01063.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
P-Glycoprotein (P-gp) is comprised of a small family of plasma membrane proteins, and its presence in high amounts often correlates with multidrug resistance in cultured cells. Dramatically increased levels of a single member of P-gp mRNA (pgp2) have been observed in experimental liver carcinogenesis models, during liver regeneration, upon culturing of hepatocytes and in the uterus of pregnant animals. In all cases, the increase in mRNA level appears to be the result of an increase in mRNA half-life (stability). Previously, we have used transcriptional inhibitors alpha-amanitin and actinomycin D to measure P-gp mRNA half-life in normal liver and in liver tumors. We showed that the level of all three P-gp mRNAs decreased with time in the presence of transcriptional inhibitors, yielding measured half-lives of less than 2 h in liver but greater than 12 h in liver tumors. This observation raised the possibility that regulation of P-gp mRNA stability plays a role in liver carcinogenesis. In the present study, we measured P-gp mRNA half-life in other normal tissues to determine if a short P-gp mRNA half-life is unique to the liver. Our study reveals that in contrast to liver, measured P-gp mRNA half-lives in most tissues examined are greater than 12 h. Moreover, we observed an unexpected, marked increase in the level of pgp2 mRNA with time after injection of transcriptional inhibitors. This can only be explained if the transcriptional inhibitors directly or indirectly inhibit the normally high degradation rate of pgp2 mRNA, resulting in the superinduction of this mRNA. These findings have implications for our understanding of the regulation of P-gp gene expression and drug resistance in vivo.
Collapse
Affiliation(s)
- Chow H Lee
- Chemistry Program, University of Northern British Columbia, Prince George, BC, Canada.
| | | |
Collapse
|
207
|
Chaudhry S, Freebern WJ, Smith JL, Butscher WG, Haggerty CM, Gardner K. Cross-regulation of T cell growth factor expression by p53 and the Tax oncogene. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:6767-78. [PMID: 12471108 DOI: 10.4049/jimmunol.169.12.6767] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this study, we demonstrate that p53 directly inhibits expression of the T cell growth factor (IL-2) in activated T cells. This repression is independent of the intrinsic transcriptional activity of p53 and is mediated by the Tax-responsive CD28RE-3'-12-O-tetradecanoylphorbol-13-acetate response element (AP1) element of the IL-2 promoter. Coexpression of the Tax oncogene causes full reversal of this repression through coordinate targeting of p300, CREB, and the NF-kappaB pathways. Paradoxically, IL-2 repression by p53 is not reversed by mdm2. Instead, mdm2 represses the IL-2 promoter by a mechanism that is synergistic with p53 and resistant to Tax reversal. The p300 structure-function studies show that these effects are linked to competitive associations among p53, Tax, and mdm2 with multiple domains of p300. The functional outcome of these antagonistic associations is revealed further by the observation that Tax and p53 induce apoptosis in activated T cells through separate and mutually exclusive pathways. Interestingly, both pathways are abrogated by mdm2. These results provide evidence that a dynamic interplay, between Tax and specific elements of the p53 network, mediates growth factor expression and programmed cell death in activated T cells.
Collapse
Affiliation(s)
- Sohail Chaudhry
- Laboratory of Receptor Biology and Gene Expression, Advanced Technology Center, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4605, USA
| | | | | | | | | | | |
Collapse
|
208
|
Sauer G, Kafka A, Grundmann R, Kreienberg R, Zeillinger R, Deissler H. Basal expression of the multidrug resistance gene 1 (MDR-1) is associated with the TT genotype at the polymorphic site C3435T in mammary and ovarian carcinoma cell lines. Cancer Lett 2002; 185:79-85. [PMID: 12142082 DOI: 10.1016/s0304-3835(02)00232-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Resistance to established drugs for cancer therapy is in many cases associated with overexpression of the multidrug resistance gene 1 (MDR-1). Regulation of basal expression of MDR-1 and mechanisms of induction as a result of exposure to cytotoxic substances are still not completely understood. Recent reports have suggested an association of the C3435T polymorphism in exon 26 of the MDR-1 gene with MDR-1 expression in duodenal mucosa cells of healthy individuals. We analyzed the C3435T and G2677T genotypes of 38 mammary and ovarian carcinoma cell lines and measured basal MDR-1 expression by real-time reverse transcriptase-polymerase chain reaction. Cell lines were classified as non-expressing or showing weak basal expression that was found to be significantly associated (six/seven versus 13/31 expressing cell lines; P=0.0448, Fisher's exact test) with the TT genotype at position 3435 of the MDR-1 gene.
Collapse
Affiliation(s)
- Georg Sauer
- Department of Obstretrics and Gynecology, University of Ulm Medical School, Frauensteige 14, D-89075, Ulm, Germany
| | | | | | | | | | | |
Collapse
|
209
|
Hawes D, Liu PY, Muggia FM, Wilczynski S, Cote R, Felix J, Terada K, Belt RJ, Alberts DS. Correlation of p53 immunostaining in primary and residual ovarian cancer at the time of positive second-look laparotomy and its prognostic role: a Southwest Oncology Group ancillary study. Gynecol Oncol 2002; 87:17-23. [PMID: 12468337 DOI: 10.1006/gyno.2002.6755] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The objective of this study was to verify the correlation between p53 immunostaining at initial diagnosis and at positive reassessment after completing platinum-based chemotherapy and to assess prognostic differences between patients whose tumors display positive immunostaining versus those that have negative immunostaining at such reassessment. METHODS This study made use of samples from patients entered into a prospective randomized study of the Southwest Oncology Group (SWOG 8835) that treated patients with minimal residual disease at second-look laparotomy with either intraperitoneal (ip) mitoxantrone or fluorodeoxyuridine (FUDR). Unstained slides from tumor obtained at the initial diagnosis and at reassessment were retrospectively requested from individual institutions. The degree of nuclear staining was determined using the anti-p53 mouse monoclonal antibody Pab1801 and previously published techniques, with a cutoff of 10% or more staining of tumor cell nuclei for a positive result. Cox model regression analysis was performed for overall survival and progression-free survival, with p53 status, ip treatment, and baseline CA125 as independent variables. RESULTS p53 determination was feasible in 22 patients both at diagnosis and at the second-look samples; 9 additional patients had only either sample available. Since concordance between the 10 negative and 12 positive immunostained samples was 100%, all 31 patients were considered in the Cox model. The death hazard ratio of p53-positive versus p53-negative patients was 4.18 (two-sided P value of 0.006). CONCLUSION p53 immunostaining at second-look laparotomy correlates with the immunostaining at diagnosis. In this series confined to patients with minimal residual disease after initial therapy subjected to second-line intraperitoneal treatment, it appears to identify a poor prognostic (positive) subset for survival.
Collapse
Affiliation(s)
- Debra Hawes
- University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
210
|
Jennings MT, Iyengar S. The molecular genetics of therapeutic resistance in malignant astrocytomas. AMERICAN JOURNAL OF PHARMACOGENOMICS : GENOMICS-RELATED RESEARCH IN DRUG DEVELOPMENT AND CLINICAL PRACTICE 2002; 1:93-9. [PMID: 12174677 DOI: 10.2165/00129785-200101020-00002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The adverse prognosis associated with malignant astrocytomas (MA) is due in part to the development of resistance by the tumor to chemo- and radiotherapy-induced cytotoxic damage. The mechanisms of resistance are poorly understood but function at the level of the endothelial cell, the blood-brain barrier and the neoplastic cell itself. The classic examples of drug resistance proteins, such as the p-glycoprotein/multidrug resistance protein 1, have been identified within MA biopsy specimens. However, it is questionable to what degree, if at all, these proteins contribute directly to the evolution and prognosis of the MA. Surprisingly, there are specific genes, not traditionally associated with resistance, which appear increasingly relevant to both tumor progression and insensitivity to cytotoxic damage. These genes are involved in cell cycle regulation, and include the retinoblastoma susceptibility gene (Rb), the tumor suppressor gene p53, as well as those encoding the cyclins, their kinases and inhibitors. The interaction between the products of these genes and intratumoral environmental factors appears to involve a dynamic and prognostically adverse selection process. It is from this perspective that the mechanism(s) of hypoxic-ischaemic selection for resistance and its therapeutic repercussions will be analyzed.
Collapse
Affiliation(s)
- M T Jennings
- Vanderbilt Ingram Cancer Center, Vanderbilt Medical School, Nashville, Tennessee, USA.
| | | |
Collapse
|
211
|
Labialle S, Gayet L, Marthinet E, Rigal D, Baggetto LG. Transcriptional regulators of the human multidrug resistance 1 gene: recent views. Biochem Pharmacol 2002; 64:943-8. [PMID: 12213590 DOI: 10.1016/s0006-2952(02)01156-5] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The multidrug resistance (MDR) phenotype is the major cause of failure of cancer chemotherapy. This phenotype is mainly due to the overexpression of the human MDR1 (hMDR1) gene. Several studies have shown that transcriptional regulation of this gene is unexpectedly complex and is far from being completely understood. Current work is aimed mainly at defining unclear and new control regions in the hMDR1 gene promoter as well as clarifying corresponding signaling pathways. Such studies provide new insights into the mechanisms by which xenobiotic molecules might modify the physiological hMDR1 expression as well as the possible role of oncogenes in the pathological dysregulation of the gene. Here we report recent findings on the regulation of hMDR1 which may help define specific targets aimed at modulating its transcription.
Collapse
Affiliation(s)
- Stéphane Labialle
- IBCP UMR 5086 CNRS UCBL, 7 passage du Vercors, F-69367 Cedex 07, Lyon, France
| | | | | | | | | |
Collapse
|
212
|
Kim EJ, Park JS, Um SJ. Identification and characterization of HIPK2 interacting with p73 and modulating functions of the p53 family in vivo. J Biol Chem 2002; 277:32020-8. [PMID: 11925430 DOI: 10.1074/jbc.m200153200] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To study the biological role of p73 alpha, a member of the p53 tumor suppressor family, we performed a yeast two-hybrid screen of a human cDNA library. Using a p73 alpha fragment consisting of amino acids 49-636 as bait, we found that p73 alpha is functionally associated with the human homologue of mouse and hamster homeodomain-interacting protein kinase 2 (HIPK2). The hamster homologue, also known as haHIPK2 or PKM, was used for further characterization of interactions between HIPK2 and members of the p53 protein family. Systematic yeast two-hybrid assays indicated a physical interaction between the oligomerization domains of p73 alpha and p53 (amino acid regions 345-380 and 319-360, respectively) and amino acid region 812-907 of haHIPK2. This region of haHIPK2 includes a PEST sequence, an Ubc9-binding domain, and a partial speckle retention sequence and is identical to amino acid residues 846-941 of human HIPK2 (hHIPK2). The interaction was confirmed by glutathione S-transferase pull-down assays in vitro and immunoprecipitation assays in vivo. HIPK2 colocalized with p73 and p53 in nuclear bodies, as shown by confocal microscopy. Overexpression of HIPK2 stabilized the p53 protein and greatly increased the p73- and p53-induced transcriptional repression of multidrug-resistant and collagenase promoters in Saos2 cells but had little effect on the p73- or p53-mediated transcriptional activation of synthetic p53-responsive and p21WAF1 promoters. Stable expression of HIPK2 in U2OS cells enhanced the cisplatin response of sub-G(1) and G(2)/M populations, and it also increased the apoptotic response to cisplatin and adriamycin as demonstrated by fluorescence-activated cell sorter and 4',6-diamidino-2-phenylindole-staining analyses. HIPK2 potentiated the inhibition of colony formation by p73 and p53. These results suggest that physical interactions between HIPK2 and members of the p53 family may determine the roles of these proteins in cell cycle regulation and apoptosis.
Collapse
Affiliation(s)
- Eun-Joo Kim
- Department of Bioscience and Biotechnology, Institute of Bioscience, Sejong University, Seoul, 143-747 Korea
| | | | | |
Collapse
|
213
|
Stein T, Crighton D, Warnock LJ, Milner J, White RJ. Several regions of p53 are involved in repression of RNA polymerase III transcription. Oncogene 2002; 21:5540-7. [PMID: 12165852 DOI: 10.1038/sj.onc.1205739] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2001] [Revised: 05/16/2002] [Accepted: 06/07/2002] [Indexed: 01/21/2023]
Abstract
The tumour suppressor p53 has been shown to regulate RNA polymerase (pol) III transcription both in vitro and in vivo. We have characterized the regions of p53 that contribute to this effect. Repression of pol III transcription in vivo does not require residues 13-19 near the N-terminus of p53 that are highly conserved through evolution. However, amino acids 22 and 23 in the adjacent transactivation domain do contribute to the inhibition of pol III activity. Deletions within the central DNA-binding core domain (residues 102-292) of p53 can entirely abolish the repression function in these assays, despite the fact that pol III templates contain no recognized p53 binding site. Deletion or substitution within the C-terminal domain of p53 can also compromise its ability to repress pol III activity in vitro and in transfected cells. These observations reveal that repression of pol III transcription is a complex function involving multiple regions of p53 extending throughout much of the protein.
Collapse
Affiliation(s)
- Torsten Stein
- Institute of Biomedical and Life Sciences, Division of Biochemistry and Molecular Biology, Davidson Building, University of Glasgow, Glasgow G12 8QQ, UK
| | | | | | | | | |
Collapse
|
214
|
Pugacheva EN, Ivanov AV, Kravchenko JE, Kopnin BP, Levine AJ, Chumakov PM. Novel gain of function activity of p53 mutants: activation of the dUTPase gene expression leading to resistance to 5-fluorouracil. Oncogene 2002; 21:4595-600. [PMID: 12096336 DOI: 10.1038/sj.onc.1205704] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2002] [Revised: 05/15/2002] [Accepted: 05/22/2002] [Indexed: 12/22/2022]
Abstract
Mutated forms of p53 are often expressed in a variety of human tumors. In addition to loss of function of the p53 tumor suppressor, mutant p53s contribute to malignant process by acquisition of novel functions that enhance transformed properties of cells and resistance to anticancer therapy in vitro, and increase tumorigenecity, invasiveness and metastatic ability in vivo. Searching for genes that change expression in response to p53 gain of function mutants may give a clue to the mechanisms underlying their oncogenic effects. Recently by subtraction hybridization cloning we found that the dUTPase gene is transcriptionally upregulated in p53-null mouse fibroblasts expressing the exogenous human tumor-derived His175 p53 mutant. Here we show that conditional expression of His175 and Trp248 hot-spot p53 mutants in p53-negative mouse 10(1) fibroblasts and human SK-OV3 and H1299 tumor cells results in increase in dUTPase gene transcription, an important marker predicting the efficacy of cancer therapy with fluoropyrimidine drugs. Using tetracycline-regulated retroviral vectors for conditional expression of p53 mutants, we found that transcription of the dUTPase gene is increased within 24 h after tetracycline withdrawal, and the cells acquire higher resistance to 5-FU. Additional inactivation of the N-terminal transcription activation domain of mutant p53 (substitutions in amino-acid residues 22 and 23) results in abrogation of both induction of dUTPase transcripts and 5-FU resistance.
Collapse
Affiliation(s)
- Elena N Pugacheva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilov Street, 117984 Moscow, Russia
| | | | | | | | | | | |
Collapse
|
215
|
Schaich M, Illmer T. Mdr1 gene expression and mutations in Ras proto-oncogenes in acute myeloid leukemia. Leuk Lymphoma 2002; 43:1345-54. [PMID: 12389613 DOI: 10.1080/10428190290033279] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Resistance to cytotoxic therapy and development of refractory disease in acute myeloid leukemia (AML) is frequently associated with the expression of mdr1/P-gp. In the last years many potential signaling pathways leading to modulation of mdr1 expression have been described. Thus, it has been assumed that activated Ras may influence mdr1 expression. This activation can be realized by mutations in the Ras oncogene leading to constitutive signaling. Ras mutations are observed in many human cancers, including AML. Recently, we could show a negative correlation between Ras mutations and mdr1 expression in blast samples of AML patients. Taking this up the potential possibilities of Ras influence on mdr1 activity and their implications on treatment outcome in AML are discussed.
Collapse
Affiliation(s)
- Markus Schaich
- Department of Medicine I, University Hospital C.G. Carus, Dresden, Germany.
| | | |
Collapse
|
216
|
Romagnolo B, Jiang M, Kiraly M, Breton C, Begley R, Wang J, Lund J, Kim SK. Downstream targets of let-60 Ras in Caenorhabditis elegans. Dev Biol 2002; 247:127-36. [PMID: 12074557 DOI: 10.1006/dbio.2002.0692] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In Caenorhabditis elegans, let-60 Ras controls many cellular processes, such as differentiation of vulval epithelial cells, function of chemosensory neurons, and meiotic progression in the germ line. Although much is known about the let-60 Ras signaling pathway, relatively little is understood about the target genes induced by let-60 Ras signaling that carry out terminal effector functions leading to morphological change. We have used DNA microarrays to identify 708 genes that change expression in response to activated let-60 Ras.
Collapse
Affiliation(s)
- Béatrice Romagnolo
- Department of Developmental Biology and Genetics, Stanford University Medical School, California 94305, USA
| | | | | | | | | | | | | | | |
Collapse
|
217
|
Abstract
P53 is a tumor-suppressor gene that codes for a multifunctional DNA-binding protein involved in cell cycle arrest, DNA repair, differentiation, and apoptosis. The P53 gene is mutated in approximately 50% of human cancers and in germline DNA of families with inherited cancer syndromes. The role of P53 mutations in the program of carcinogenic genetic alterations differs among tumor sites ranging from the earliest mutations that can be detected in premalignant cells to mutations that trigger malignant transformation of a benign neoplasm. P53 mutations can cause expression of abnormal proteins or result in complete absence of P53 expression. For these reasons the role of P53 genetic disruption has different implications in different tumor types and may vary depending on the effect of the mutation on P53 protein function. Immunohistochemical detection of P53, commonly used as a surrogate for identification of a mutant gene, has imperfect sensitivity and specificity, further complicating correlations between P53 gene status and clinical outcomes. The presence of P53 mutations has been shown to affect prognosis of some cancers. The identity of P53 mutations can be used to determine tumor clonality. The detection of P53 mutations suggests the severity of premalignant lesions. Evolving technology for more accurate identification of P53 mutations, better understanding of the function of mutant P53 protein, and more detailed analysis of individual tumor types may expand the relevance of P53 gene analysis for clinical outcomes and therapeutic response.
Collapse
Affiliation(s)
- Minetta C Liu
- Department of Oncology, Lombardi Cancer Center, Georgetown University School of Medicine, Washington, DC 20007, USA
| | | |
Collapse
|
218
|
Cripe LD, Gelfanov VM, Smith EA, Spigel DR, Phillips CA, Gabig TG, Jung SH, Fyffe J, Hartman AD, Kneebone P, Mercola D, Burgess GS, Boswell HS. Role for c-jun N-terminal kinase in treatment-refractory acute myeloid leukemia (AML): signaling to multidrug-efflux and hyperproliferation. Leukemia 2002; 16:799-812. [PMID: 11986940 DOI: 10.1038/sj.leu.2402457] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2001] [Accepted: 01/15/2002] [Indexed: 11/09/2022]
Abstract
A relationship was proved between constitutive activity of leukemic cell c-jun-N-terminal kinase (JNK) and treatment failure in AML. Specifically, early treatment failure was predicted by the presence of constitutive JNK activity. The mechanistic origins of this association was sought. A multidrug resistant leukemic cell line, HL-60/ADR, characterized by hyperexpression of c-jun and JNK activity, was transfected with a mutant c-jun vector, whose substrate N-terminal c-jun serines were mutated. Down-regulated expression occurred of c-jun/AP-1-dependent genes, catalase and glutathione-S-transferase (GST) pi, which participate in cellular homeostasis to oxidative stress and xenobiotic exposure. MRP-efflux was abrogated in HL-60/ADR cells with dominant-negative c-jun, perhaps because MRP1 protein expression was also lost. Heightened sensitivity to daunorubicin resulted in cells subjected to this change. Biochemical analysis in 67 primary adult AML samples established a statistical correlation between cellular expression of c-jun and JNK activity, JNK activity with hyperleukocytosis at presentation of disease, and with exuberant MRP efflux. These findings reflect the survival role for c-jun/AP-1 and its regulatory kinase previously demonstrated for yeast in homeostatic response to oxidative stress and in operation of ATP-binding cassette efflux pumps, and may support evolutionary conservation of such function. Thus, JNK and c-jun may be salient drug targets in multidrug resistant AML.
Collapse
Affiliation(s)
- L D Cripe
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, 1044 W Walnut Street, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Brownlee NA, Hazen-Martin DJ, Garvin AJ, Re GG. Functional and gene expression analysis of the p53 signaling pathway in clear cell sarcoma of the kidney and congenital mesoblastic nephroma. Pediatr Dev Pathol 2002; 5:257-68. [PMID: 12007018 DOI: 10.1007/s10024-001-0215-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mutation of p53 has been implicated in progression of classical Wilms tumor (WT) into the anaplastic variant (AWT), drug resistance, and poor prognosis. Because of prognostic similarities, clear cell sarcoma of the kidney (CCSK) has been classified with AWT and other aggressive pediatric renal tumors, apart from congenital mesoblastic nephroma (CMN), which is instead a relatively benign tumor of neonates. Initially, CCSK and CMN were assumed to be ontologically related, but the role of p53 in the pathogenesis of either disease has not been sufficiently evaluated as in AWT. We examined the status of p53 in CMN and CCSK in comparison to AWT by immunohistochemistry and mRNA analysis of p53, the downstream effector p21(WAF-1/CIP-1) ( p21), the multidrug resistance gene MDR-1, a putative target of p53, and the p53-antagonist Mdm-2. Surprisingly, strong p53 nuclear immunoreactivity was found in cultures from two CMN specimens, but not in frozen or fixed tumor tissue from five other CMN specimens, nor in cell lines or tumor tissue from CCSK. Sequence analysis excluded p53 mutations. The size of the p53 mRNA in CMN and CCSK primary tumors excluded gross deletions or rearrangements. Low levels of Mdm-2 mRNA in CCSK and CMN primary tumors and cultures did not support a role for Mdm-2. Absence of MDR-1 mRNA excluded MDR-1 in the drug-resistant phenotype of CCSK. Cisplatin-induced p21 transactivation assays and G(1) cell cycle arrest analyses showed that p21 transactivation and G(1) arrest occurred in both CCSK and CMN cultures, demonstrating integrity of the p53 signal transduction pathway. Absence of p53 functional abnormalities excluded relationships between CCSK and CMN as in AWT, supporting the association of cellular CMN with congenital fibrosarcomas as more recently proposed.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Cyclin-Dependent Kinase Inhibitor p21
- Cyclins/genetics
- Cyclins/metabolism
- DNA, Neoplasm/analysis
- Gene Expression Profiling
- Humans
- Immunoenzyme Techniques
- Infant, Newborn
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Neoplasm Staging
- Nephroma, Mesoblastic/congenital
- Nephroma, Mesoblastic/genetics
- Nephroma, Mesoblastic/metabolism
- Nuclear Proteins
- Phenotype
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-mdm2
- RNA, Messenger/metabolism
- Sarcoma, Clear Cell/genetics
- Sarcoma, Clear Cell/metabolism
- Sarcoma, Clear Cell/pathology
- Signal Transduction
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Noel A Brownlee
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
220
|
Shi XB, Nesslinger NJ, Deitch AD, Gumerlock PH, deVere White RW. Complex functions of mutant p53 alleles from human prostate cancer. Prostate 2002; 51:59-72. [PMID: 11920959 DOI: 10.1002/pros.10072] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Few studies have used multiple assays to examine the functionality of mutant p53 in prostate cancer (CaP). We employed seven functional assays to study 16 representative mutant p53 alleles, six from localized and ten from metastatic CaP. METHODS Yeast assays were employed to determine loss of function (LOF), partial function (PF), and dominant-negative status. Assays using p53-null Saos2 cells were used to determine whether mammalian cells transfected with mutant p53 could up-regulate the MDR-1 or PCNA promoters, alter IL-6 expression or confer the ability to grow in soft agar. As a further test of gain of function (GOF), p53-null PC3 cells stably transfected with these mutant p53 alleles were examined for cell cycle distributions. RESULTS All 16 mutant p53 alleles demonstrated either total or partial LOF. All but one allele also had at least one gain of function; however, the pattern of GOF was different for each mutant allele. Alleles derived from both localized and metastatic CaP had similar GOF characteristics; however, only alleles from metastatic disease had significantly increased S-phase fractions. CONCLUSIONS Different mutant p53 alleles from CaP had different, complex functional profiles. The lack of predictable patterns for these alleles suggest that each mutation may uniquely affect p53 function.
Collapse
Affiliation(s)
- Xu-Bao Shi
- Department of Urology, University of California, Davis, School of Medicine, Sacramento, California 95817, USA
| | | | | | | | | |
Collapse
|
221
|
Abstract
OBJECTIVE To review the literature published in the past 6 years concerning the role of p53 tumor-suppressor protein in rheumatoid arthritis (RA). METHODS A MEDLINE search was performed to identify all publications that covered the role of p53 in RA. In addition, selected articles related to proto-oncogenes and matrix metalloproteinases were included in this review. RESULTS p53 protein is expressed in RA fibroblast-like synoviocytes (FLSs), and its overexpression is a characteristic feature of RA. The overexpression of p53 is probably induced by DNA strand breaks caused by the genotoxic environment of RA joints, in some cases because of p53 mutations. Independent studies from 3 groups indicated that p53 mutations can and do occur in RA synovial tissue samples derived from a subset of RA patients. Inactivation of p53 may contribute to the invasiveness of FLSs and to the high-level expression of cartilage degradation enzymes as well. Gene transfer or gene knockout studies using a collagen-II-induced RA animal model to examine the role of p53 in RA have been reported. Initial results are positive and indicate that gene transfer of p53 may be clinically useful for the management of RA. CONCLUSIONS p53 protein is expressed in RA FLSs, and its overexpression is a characteristic feature of RA. p53 mutations occur in the synovial tissues derived from a subset of RA patients. The clinical implications of p53 expression and the functional importance of somatic mutations in RA, however, are still unclear. Further research is needed to fully understand the implications of these findings and develop corresponding new therapeutic strategies.
Collapse
Affiliation(s)
- Yubo Sun
- Department of Medicine, University of Miami School of Medicine, FL, USA
| | | |
Collapse
|
222
|
Bush JA, Li G. Cancer chemoresistance: the relationship between p53 and multidrug transporters. Int J Cancer 2002; 98:323-30. [PMID: 11920581 DOI: 10.1002/ijc.10226] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Extensive studies indicate that both p53 and multidrug transporters play important roles in chemoresistance. Since the initial reports a decade ago demonstrating a transcriptional dependence of the ABCB1 gene (MDR) promoter by p53, much data have been accumulated. However, despite being the subject of intense study, this p53-MDR relationship remains unclear in human cancers. The data are confounded by variable and contrasting results when considering the in vitro regulation and attempting to draw parallels in tissue specimens. The original model suggested that wild-type p53 downregulates the ABCB1 promoter, whereas mutant p53 increases expression of ABCB1. This review summarizes the data for and against this hypothesis. What emerges from these studies is a complex picture, where data have been obtained in support of this hypothesis, but there are also many circumstances where it is not supported. Taken together, these data suggest that the relationship between p53 and multidrug transporters is conditional. It is dependent on cellular environment, the drug used, and the nature of the p53 mutation.
Collapse
Affiliation(s)
- Jason A Bush
- Division of Dermatology, Department of Medicine, Vancouver Hospital and Health Sciences Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
223
|
Emadi-Konjin HP, Zhang H, Anandan V, Sun D, Schuetz J, Furuya KN. Isolation of a genomic clone containing the promoter region of the human ATP binding cassette (ABC) transporter, ABCB6. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1574:117-30. [PMID: 11955620 DOI: 10.1016/s0167-4781(01)00340-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We previously reported on the isolation of a new rat ATP binding cassette (ABC) transporter, ABCB6. We now report the isolation of the full-length cDNA and genomic clones containing the human ABCB6 gene. ABCB6 is 100% identical to the cloned MTABC3 human ABC transporter and contains the typical ABC signature, Walker A and B motifs. We found that HuABCB6 is expressed at low levels in normal human liver. We found that ABCB6 was overexpressed in human hepatocellular carcinomas compared to paired surrounding non-malignant tissue. We found that there was no difference in ABCB6 gene copy between human liver cancer and its paired non-malignant tissue. Because HuABCB6 was overexpressed in human cancers compared to peri-tumoral tissue in the absence of gene amplification, transcriptional regulation may play an important role in its expression. Therefore, we isolated a 14 kb genomic DNA clone containing the HuABCB6 promoter and 5'-flanking region. The 5'-flanking region contains a CpG island, lacks an appropriately positioned TATA element and contains a number of putative transcription factor binding sites. Two transcription start sites were identified by S1 nuclease mapping at -274 and -296 bp from the start codon. Transient transfection of the HuABCB6 promoter constructs (HuABCB6/1.68, 1.39, 1.13, 0.90, 0.52) containing the luciferase reporter gene resulted in a 1100-2300-fold increase in luciferase activity compared to the empty vector control whereas HuABCB6/1.68 subcloned in the reverse orientation resulted in no activity. We observed a significant decrease in luciferase activity with the promoter constructs, HuABCB6/0.25, 0.15 and 0.06, which indicates that an orientation-dependent functional promoter is contained within our previously predicted promoter region of -315 bp to -565 bp as deletion of this 250 bp sequence resulted in a loss of promoter activity.
Collapse
Affiliation(s)
- H Pasha Emadi-Konjin
- Division of Gastroenterology and Nutrition, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
224
|
Deb D, Scian M, Roth KE, Li W, Keiger J, Chakraborti AS, Deb SP, Deb S. Hetero-oligomerization does not compromise 'gain of function' of tumor-derived p53 mutants. Oncogene 2002; 21:176-89. [PMID: 11803461 DOI: 10.1038/sj.onc.1205035] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2001] [Revised: 09/14/2001] [Accepted: 10/09/2001] [Indexed: 01/10/2023]
Abstract
Tumor-derived p53 mutants activate transcription from promoters of various growth-related genes. We tested whether this transactivation function of the mutant protein is sufficient to induce tumorigenesis ('gain of function'). Tumor-derived mutant p53-281G transactivates the promoters of human epidermal growth factor receptor (EGFR) and human multiple drug resistance gene (MDR-1). To determine whether the C-terminal domain functions only as an oligomerization domain in mutant p53-mediated transactivation, we have replaced the tetramerization domain of p53 by a heterologous tetramerization domain; although this mutant protein formed tetramers in solution, it failed to transactivate significantly. Therefore, for successful mutant p53-mediated transactivation, sequences near the C-terminus of mutant p53 are required to perform functions in addition to tetramerization. We also demonstrate that co-expression of a deletion mutant of p53 (p53 del 1-293), which retains the p53 oligomerization domain, inhibits this transactivation. p53 del 1-293 co-immunoprecipitates with p53-281G suggesting that hetero-oligomers of p53-281G and p53 del 1-293 are defective in transactivation. We also show that a cell line stably transfected with p53-281G expresses higher levels of endogenous NF-kappaB and proliferating cell nuclear antigen (PCNA) compared to that transfected with vector alone. On co-expression, p53 del 1-293 lowered the levels of NF-kappaB and PCNA in p53-281G-expressing cells. However, on co-expression, p53 del 1-293 did not inhibit the tumorigenicity and colony forming ability of p53-281G expressing cells. Our earlier work showed that a deletion of the C-terminal sequences of p53-281G overlapping the oligomerization domain obliterates 'gain of function'. Taken together, the above information suggests that the C-terminal sequences have some critical role in 'gain of function' in addition to transactivation.
Collapse
Affiliation(s)
- Debabrita Deb
- Department of Biochemistry and Molecular Biophysics, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia, VA 23298, USA
| | | | | | | | | | | | | | | |
Collapse
|
225
|
Bonovich M, Olive M, Reed E, O'Connell B, Vinson C. Adenoviral delivery of A-FOS, an AP-1 dominant negative, selectively inhibits drug resistance in two human cancer cell lines. Cancer Gene Ther 2002; 9:62-70. [PMID: 11916245 DOI: 10.1038/sj.cgt.7700409] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2001] [Indexed: 11/08/2022]
Abstract
Activator protein-1 (AP-1) transcription factor has been linked to chemotherapeutic resistance. To assess the clinical efficacy of AP-1 inhibition toward reversing drug resistance, we have developed an adenovirus expressing a dominant negative that inhibits AP-1 DNA binding, namedAdA-FOS. We examined the consequence of AdA-FOS infection on two paired human cancer cell lines, each pair consisting of a parental cell and the drug- resistant derivative. The first pair of cells is the parental human ovarian cancer cell line A2780 and the cisplatin-resistant A2780/CP70 cell line. The second pair of cells is the parental epidermal carcinoma cell line KB8 and the multidrug-resistant (mdr) KB85 cell line. Because of an association of up-regulated AP-1 activity with their drug resistance, these cell lines were considered good targets of AdA-FOS therapy. Following infection of the drug-sensitive and drug-resistant cells, we observed a significant decrease in cell viability of KB85 and A2780/CP70 cells at drug doses normally not lethal to the cell. The parental cell lines, A2780 and KB8 cells, were not similarly affected by AdA-FOS. This decrease in viability was specific to AdA-FOS as an adenovirus control (Advector) did not reverse drug resistance. Although the efficiency of AdA- FOS in therapy would need to be further analyzed with other cisplatin-resistant and mdr cell lines, these results suggest that AP-1 is a therapeutic molecular target and that inhibition of AP-1 DNA binding may be of clinical value in treating chemotherapeutic resistance.
Collapse
Affiliation(s)
- Maria Bonovich
- Laboratory of Metabolism, Medicine Branch, National Cancer Institute, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
226
|
Abstract
To characterize the multidrug resistance (MDR) phenotype in human oral squamous cell carcinomas (OSCCs), the expression levels of four MDR-related genes (multidrug resistance, mdr1; multidrug resistance-associated protein, MRP; glutathione S-transferase-pi, GST-pi; and DNA topoisomerase II, topoII) were analyzed in OSCCs. Fifty-two OSCC tissues and 22 normal oral mucosal tissues were involved in this study. The expression of each gene was analyzed with a reverse-transcription polymerase chain reaction (RT-PCR) method using beta(2)m microglobulin (beta(2)m) mRNA as an endogenous control. The mean values of mdr1, MRP, GST-pi, and topoII gene expression relative to the beta(2)m gene in OSCC tissues were 0.37, 0.75, 0.66, and 1.11; those of normal oral mucosa were 0.40, 0.27, 0.62, and 0.91, respectively. The averaged expression levels of the MRP and topoII gene in OSCC tissues were higher than those of normal oral mucosas (P=0.001 and P=0.02, respectively). The expression levels of four MDR-related genes in OSCCs were not related with the degree of histologic cell differentiation, tumor stage, primary or recurred tumor, or the presence or absence of chemotherapy. Linear regression analysis showed a correlation between the expression levels of MRP and GST-pi in normal oral mucosas (r=0.596, P=0.003) and in OSCCs (r=0.287, P=0.039). The results suggest that MRP expression is activated during the tumorigenesis of OSCCs and that this may play a role in de novo drug resistance in OSCCs. These results should provide further insight into the complex role postulated for MDR-related genes in chemotherapy, carcinogenesis and tumor progression.
Collapse
Affiliation(s)
- Y S Cho
- Department of Oral and Maxillofacial Surgery, College of Medicine and Medical Research Institute, Chungbuk National University, #62 Gaeshin-Dong, Heungdeok-Gu. Cheongju, Chungbuk, 361-240, South Korea.
| | | |
Collapse
|
227
|
Takagi S, Ueda Y, Hijikata M, Shimotohno K. Overproduced p73alpha activates a minimal promoter through a mechanism independent of its transcriptional activity. FEBS Lett 2001; 509:47-52. [PMID: 11734204 DOI: 10.1016/s0014-5793(01)03141-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
p73, the gene for a protein related to the tumor suppressor p53, encodes several variants which bear distinct carboxy-terminal structures as a result of alternative splicing. We and others showed that these splicing variants have different transcriptional effects on promoters with a p53-binding consensus sequence (p53BCS). Here we show that when transiently overexpressed, p73alpha but not p73beta activated several minimal promoters without the p53BCS, while p73gamma and p73epsilon activated them to a much lesser extent than p73alpha, and p53 suppressed the promoters without p53BCS as reported previously. Moreover, the results of RNase protection and RNA transfection assays suggested that this activation occurred at the transcriptional level. Deletion analysis of p73alpha revealed that the transactivation domain of p73 was not involved in this activity and the C-terminal region of p73alpha which is a specific structure of this variant was essential, suggesting that this phenomenon occurs independent of the transactivation activity of p73alpha and that the C-terminal extension of p73alpha may affect the basal level of transcription.
Collapse
Affiliation(s)
- S Takagi
- Department of Viral Oncology, Institute for Virus Research, Kyoto University, Sakyo-ku, 606-8507, Kyoto, Japan
| | | | | | | |
Collapse
|
228
|
Cocker HA, Hobbs SM, Tiffin N, Pritchard-Jones K, Pinkerton CR, Kelland LR. High levels of the MDM2 oncogene in paediatric rhabdomyosarcoma cell lines may confer multidrug resistance. Br J Cancer 2001; 85:1746-52. [PMID: 11742497 PMCID: PMC2363993 DOI: 10.1054/bjoc.2001.2145] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The MDM2 protein is known to be overexpressed in some sarcomas including rhabdomyosarcoma. However, the extent to which the MDM2 protein influences sensitivity to chemotherapeutic drugs is unclear. We have analysed this further using stable transfection of the mdm2 gene into 4 well-characterised human paediatric rhabdomyosarcoma cell lines. Transfection with the mdm2 gene resulted in increased levels of the MDM2 protein in all the cell lines. In 2 of the lines, SCMC and RD, the mdm2 gene caused between 2-fold and 61-fold increase in resistance to vincristine, etoposide and doxorubicin but not to cisplatin. In these lines there was an increase in expression of the mdr-1 gene which encodes P-glycoprotein, but not the mrp1 gene which encodes the multidrug resistance protein (MRP). The resistance was reversible using the MDR modulator PSC833, confirming the presence of P-glycoprotein. We conclude that MDM2 overexpression may be a mechanism by which multidrug resistance is regulated in some rhabdomyosarcomas.
Collapse
Affiliation(s)
- H A Cocker
- CRC Centre for Cancer Therapeutics, The Institute of Cancer Research, Cotswold Road, Sutton, Surrey, SM2 5NG, UK
| | | | | | | | | | | |
Collapse
|
229
|
Cadwell C, Zambetti GP. The effects of wild-type p53 tumor suppressor activity and mutant p53 gain-of-function on cell growth. Gene 2001; 277:15-30. [PMID: 11602342 DOI: 10.1016/s0378-1119(01)00696-5] [Citation(s) in RCA: 180] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The tumor suppressor p53 plays a central role in the protection against DNA damage and other forms of physiological stress primarily by inducing cell cycle arrest or apoptosis. Mutation of p53, which is the most frequent genetic alteration detected in human cancers, inactivates these growth regulatory functions and causes a loss of tumor suppressor activity. In some cases, mutation also confers tumor-promoting functions, such as the transcriptional activation of genes involved in cell proliferation, cell survival and angiogenesis. Consequently, cells expressing some forms of mutant p53 show enhanced tumorigenic potential with increased resistance to chemotherapy and radiation. Our current understanding of these activities is summarized in this review. By dissecting out mechanistic differences between wild-type and mutant p53 activities, it may be possible to develop therapeutics that restore tumor suppressor function to mutant p53 or that selectively inactivate mutant p53 tumor-promoting functions.
Collapse
Affiliation(s)
- C Cadwell
- Department of Biochemistry, St. Jude Children's Research Hospital, 332 N. Lauderdale, Memphis, TN 38105, USA
| | | |
Collapse
|
230
|
Kato A, Miyazaki M, Ambiru S, Yoshitomi H, Ito H, Nakagawa K, Shimizu H, Yokosuka O, Nakajima N. Multidrug resistance gene (MDR-1) expression as a useful prognostic factor in patients with human hepatocellular carcinoma after surgical resection. J Surg Oncol 2001; 78:110-5. [PMID: 11579388 DOI: 10.1002/jso.1129] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Multidrug resistance gene (MDR-1) overexpression has been correlated with tumor aggressiveness and worse prognosis in some human neoplasms. The aim of this study is to evaluate the clinical value of MDR-1 mRNA expression as a prognostic factor after surgical resection in human hepatocellular carcinoma (HCC). METHODS MDR-1 mRNA levels in tissue samples from 34 patients with HCC, who underwent surgical resection, were measured by quantitative northern blot analysis. We stratified these patients into two groups according to a ratio of MDR-1 mRNA levels of HCC to nontumorous tissue; MDR-1 mRNA ratio > or = 1.0 and < 1.0. The overall and disease-free survival rates were analyzed using multivariate regression analysis. RESULTS The median survival periods were 10.3 and 35.8 months for patients with the MDR-1 mRNA ratio > or = 1.0 and < 1.0, respectively, and the corresponding 5-year survival rates were 33 and 54%, respectively, P < 0.05. The multivariate analysis revealed that TNM stage and MDR-1 mRNA ratio were independent factors for predicting overall survival after surgical resection. CONCLUSION This study suggested that the measurement of the MDR-1 mRNA levels in HCC and nontumorous liver tissue might be a useful prognostic factor after surgical resection in patients with HCC.
Collapse
Affiliation(s)
- A Kato
- First Department of Surgery, School of Medicine, Chiba University, Chuo-Ku, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
231
|
Zhan M, Yu D, Lang A, Li L, Pollock RE. Wild type p53 sensitizes soft tissue sarcoma cells to doxorubicin by down-regulating multidrug resistance-1 expression. Cancer 2001; 92:1556-66. [PMID: 11745235 DOI: 10.1002/1097-0142(20010915)92:6<1556::aid-cncr1482>3.0.co;2-s] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND p53 mutations occur in almost half of all soft tissue sarcomas (STS) and may contribute to multidrug resistance (MDR) in patients with STS. Doxorubicin (Dox) is one of the most active single agents in STS but is less effective in STS with p53 mutations. The effect of reintroducing wild type (wt) p53 into STS cells harboring p53 mutations on the cytotoxicity of DOX in vitro and in vivo was studied. METHODS The following cell lines were used in this study: SKLMS-1 STS cells, which do not express wt p53; two wt p53 stable transfectant cells derived from SKLMS-1 cells; and SKLMS-1 transfectant cells from a p53 temperature-sensitive mutant that expresses wt p53 at 32 degrees C and mutant p53 at 38 degrees C. The cytotoxicity of Dox was examined by [3-(4,5-dimethylthiazzol-2-yl)-2,5-diphenltetrazolium] (MTT) and clonogenetic assay, and the effect of reintroducing wt p53 on tumor suppression by Dox was evaluated with a tumorigenicity assay. DNA fragmentation was used to detect apoptosis. MDR-1 P-glycoprotein (P-gp) expression was detected by Western blot and immunohistochemical analyses of protein levels and by Northern blot analysis of mRNA levels, respectively. The intracellular accumulation of Dox was detected by flow cytometric analysis. RESULTS The 50% inhibitory concentration (IC(50)) of Dox for the SKLMS-1 wt p53 transfectants decreased 16-fold compared with SKLMS-1 parental cells expressing mutant p53. Colony formation of SKLMS-1 cells after Dox treatment also was inhibited by wt p53 reintroduction. The tumorigenicity of SKLMS-1 cells was inhibited by wt p53 reintroduction alone or by Dox treatment alone and was inhibited further when p53 introduction was combined with Dox treatment in severe combined immunodeficient mice. Although no difference in DNA fragmentation, Bax expression, or Bcl-2 expression was detected among wt p53 transfectants and parental SKLMS-1 cells after Dox treatment, MDR-1 P-gp expression was decreased in wt p53 transfectants compared with parental SKLMS-1 cells. Furthermore, higher intracellular accumulations of Dox were found in wt p53 transfectants than that in SKLMS-1 cells. CONCLUSIONS Reintroduction of wt p53 into STS cells harboring p53 mutations can enhance their chemosensitivity to Dox through the inhibition of MDR-1 P-gp expression. Thus, the combination of p53 gene therapy and chemotherapy may increase the therapeutic efficacy in the treatment of patients with STS.
Collapse
Affiliation(s)
- M Zhan
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | |
Collapse
|
232
|
Yoon SS, Ahn KS, Kim SH, Shim YM, Kim J. In vitro establishment of cis-diammine-dichloroplatinum(II) resistant lung cancer cell line and modulation of apoptotic gene expression as a mechanism of resistant phenotype. Lung Cancer 2001; 33:221-8. [PMID: 11551417 DOI: 10.1016/s0169-5002(01)00205-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
After exposure of H460 cells to an increasing concentrations of cis-diammine-dichloroplatinum(II) (cisplatin, CDDP) for 6 months, cisplatin resistant cells were isolated (H460/CIS). The biologic behaviors of H460 and H460/CIS cells were tested using animal experiments. Only the resistant cells developed lung metastases despite cisplatin treatment. The characteristics of H460/CIS cells are as follows, MTT analyses revealed that H460/CIS cells were markedly resistant to cisplatin compared with their parental cells. Also, H460/CIS cells exhibited cross-resistance to DNA damaging agents such as doxorubicin (DXR) and etoposide. Cisplatin treatment dramatically increased p53 expression in parental cells but not in H460/CIS cells which expressed basal levels of p53. Without cisplatin treatment, Bcl-2 and Bax were expressed in H460/CIS cells, but not in parental cell. Our data suggested that p53, Bax and Bcl-2 were up-regulated in H460/CIS cells. These changes could explain some of the mechanisms of cisplatin resistance. Thus, H460/CIS could be useful to investigate the mechanisms of drug resistance to cisplatin including apoptotic gene expressions conferring drug resistance, thereby making progress in the treatment of cisplatin-resistant tumor cells.
Collapse
Affiliation(s)
- S S Yoon
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | | | | | | | | |
Collapse
|
233
|
Zelazny E, Li B, Anagnostopoulos AM, Coleman A, Perkins AS. Cooperating oncogenic events in murine mammary tumorigenesis: assessment of ErbB2, mutant p53, and mouse mammary tumor virus. Exp Mol Pathol 2001; 70:183-93. [PMID: 11417997 DOI: 10.1006/exmp.2001.2357] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We are investigating cooperating genetic events in the genesis of breast cancer, using the mouse as a model system. We have shown cooperativity between a mutant allele of p53 (p53-172H) and overexpressed ErbB2 in mammary tumorigenesis in transgenic mice. We are now performing additional crosses to further examine oncogene cooperativity with ErbB2 and p53-172H. We attempted to test the dominant oncogenic potential of p53-172H in an in vivo setting by crossing the p53-172H transgene together with ErbB2 onto either a p53(-/-) or a p53(+/-) background. We show that the p53-172H allele and the heterozygous p53 genotype have an identical impact on the latency of ErbB2-induced mammary tumors; there was no evidence of additivity or synergy between p53-172H and the p53(+/-) genotype. On the p53(-/-) background, we obtained no mammary tumors due to the early onset of lymphomas and sarcomas, thus precluding assessment of the effect of the p53-172H transgene on mammary tumorigenesis in a p53-null background. Thus, in this in vivo model for breast cancer, we failed to find evidence that p53-172H can function as a dominant oncogenic allele, but rather found support for its being essentially equivalent to a null allele in its impact on ErbB2-induced mammary tumorigenesis. By comparative genome analysis, we showed that a common feature of tumors arising in ErbB2/mutant p53 mice (p53-null allele with or without p53-172H) is a loss of chromosome 4, a feature of many epithelial tumors in mice and one that is consistent with a role for loss of INK4a/ARF in such tumors. We also attempted to accelerate ErbB2-induced mammary tumorigenesis with mouse mammary tumor virus (MMTV) proviral tagging mutagenesis, but we were surprised to find that mice with MMTV alone had the same latency as mice with both MMTV and ErbB2, indicating no cooperativity between ErbB2 and MMTV. This may have been due to the mixed C3H/HeN x FVB strain background used in this cross.
Collapse
MESH Headings
- Alleles
- Animals
- Breast Neoplasms/genetics
- Cell Transformation, Neoplastic
- Crosses, Genetic
- Disease Models, Animal
- Epithelial Cells/cytology
- Epithelial Cells/pathology
- Female
- Genes, erbB-2
- Genes, p53
- Genotype
- Humans
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/pathology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Knockout
- Mice, Transgenic
- Nucleic Acid Hybridization
- Receptor, ErbB-2/metabolism
- Tumor Suppressor Protein p53/deficiency
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- E Zelazny
- Yale University Department of Pathology, 310 Cedar Street, New Haven, Connecticut, 06520-8025, USA
| | | | | | | | | |
Collapse
|
234
|
Johnson RA, Ince TA, Scotto KW. Transcriptional repression by p53 through direct binding to a novel DNA element. J Biol Chem 2001; 276:27716-20. [PMID: 11350951 DOI: 10.1074/jbc.c100121200] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The tumor suppressor protein p53 has been well documented as a transcriptional activator involved in the regulation of a number of critical genes involved in the cell cycle, response to DNA damage, and apoptosis. Activation by p53 requires the interaction of the protein with a consensus binding site consisting of two half-sites, each comprising two copies of the sequence PuPuPuC(A/T) arranged head-to-head and separated by 0-13 base pairs. In addition to activation, p53 has been shown to be a potent repressor of transcription. However, the basis for p53-mediated repression is not well understood and has been proposed to occur indirectly through interactions with other promoter-bound transcription factors. In the present study, we show that p53 can repress transcription directly by binding to a novel head-to-tail (HT) site within the MDR1 promoter. A mutation that disrupted p53 binding to the MDR1 HT site blocked p53-mediated repression of the MDR1 promoter in transfection assays. Replacement of the HT site with a head-to-head (HH) site converted the activity of p53 from repression to activation, indicating that simple recruitment of p53 to the promoter is not sufficient for repression and that the orientation of the binding element determines the fate of p53-regulated promoters.
Collapse
Affiliation(s)
- R A Johnson
- Program in Molecular Pharmacology and Therapeutics, Memorial Sloan-Kettering Cancer Center, New York, New York 1002, USA
| | | | | |
Collapse
|
235
|
Abstract
Flavopiridol inhibits phosphokinases. Its activity is strongest on cyclin dependent kinases (cdk-1, -2, -4, -6, -7) and less on receptor tyrosine kinases (EGFR), receptor associates tyrosine kinases (pp60 Src) and on signal transducing kinases (PKC and Erk-1). Although the inhibiting activity of flavopiridol is strongest for cdk, the cytotoxic activity of flavopiridol is not limited to cycling cells. Resting cells are also killed. This fact suggests that inhibition of cdks involved in the control of cell cycle is not the only mechanism of action. Inhibition of cdk's with additional functions (i.e. involved in the control of transcription or function of proteins that do not control cell cycle) may contribute to the antitumoral effect. Moreover, direct and indirect inhibition of receptor activation (EGFR) and/or a direct inhibition of kinases (pp60 Src, PKC, Erk-1) involved in the signal transduction pathway could play a role in the antiproliferative activity of flavopiridol. From pharmacokinetic data in patients it can be concluded that the inhibitory activity (IC50) of flavopiridol on these kinases is in the range of concentrations that might be achieved intracellularly after systemic application of non-toxic doses of flavopiridol. However, no in situ data from flavopiridol treated cells have been published yet that prove that by inhibition of EGFR, pp60 Src, PKC and/or Erk-1 (in addition to inhibition of cdk's) flavopiridol is able to induce apoptosis. Thus many questions regarding the detailed mechanism of antitumoral action of flavopiridol are still open. For the design of protocols for future clinical studies this review covers the essential information available on the mechanism of antitumoral activity of flavopiridol. The characteristics of this antitumoral activity include: High rate of apoptosis, especially in leukemic cells; synergy with the antitumoral activity of many cytostatics; independence of its efficacy on pRb, p53 and Bcl-2 expression; lack of interference with the most frequent multidrug resistance proteins (P-glycoprotein and MRP-190); and a strong antiangiogenic activity. Based on these pharmacological data it can be concluded that flavopiridol could be therapeutically active in tumor patients: independent on the genetic status of their tumors or leukemias (i.e. mutations of the pRb and/or p53, amplification of bcl-2); in spite of drug resistance of their tumors induced by first line treatment (and caused by enhanced expression of multidrug resistance proteins); in combination with conventional chemotherapeutics preferentially given prior to flavopiridol; and due to a complex mechanism involving cytotoxicity on cycling and on resting tumor cells, apoptosis and antiangiogenic activity. In consequence, flavopiridol is a highly attractive, new antitumoral compound and deserves further elucidation of its clinical potency.
Collapse
Affiliation(s)
- H H Sedlacek
- Aventis Pharma Deutschland GmbH, Central Biotechnology, P.O. Box 1140, 35001, Marburg, Germany.
| |
Collapse
|
236
|
Ding BC, Whetstine JR, Witt TL, Schuetz JD, Matherly LH. Repression of human reduced folate carrier gene expression by wild type p53. J Biol Chem 2001; 276:8713-9. [PMID: 11106643 DOI: 10.1074/jbc.m005248200] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The relationship between loss of functional p53 and human reduced folate carrier (hRFC) levels and function was examined in REH lymphoblastic leukemia cells, which express wild type p53, and in p53-null K562 cells (K562(pTet-on/p53)) engineered to express wild type p53 under control of a tetracycline-inducible promoter. Activation of p53 in REH cells by treatment with daunorubicin was accompanied by decreased ( approximately 5-fold) levels of hRFC transcripts and methotrexate transport. Treatment of K562(pTet-on/p53) cells with doxycycline resulted in a dose-dependent expression of p53 protein and transcripts, increased p21 protein, decreased dihydrofolate reductase, and G(1) arrest with decreased numbers of cells in S-phase. p53 induction was accompanied by up to 3-fold decreases in hRFC transcripts transcribed from the upstream hRFC-B promoter and similar losses of hRFC protein and methotrexate uptake capacity. Expression of p15 in an analogous inducible system in K562 cells resulted in a nearly identical decrease of S-phase cells and dihydrofolate reductase without effects on hRFC levels or activity. When the hRFC-B promoter was expressed as full-length and basal promoter-luciferase reporter constructs in K562(pTet-on/p53) cells, induction of p53 with doxycycline resulted in a 3-fold loss of promoter activity, which was reversed by cotransfection with a trans-dominant-negative p53. These studies show that wild type p53 acts as a repressor of hRFC gene expression, via a mechanism that is independent of its effects on cell cycle progression.
Collapse
Affiliation(s)
- B C Ding
- Department of Pharmacology and the Experimental and Clinical Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan 48201 , USA
| | | | | | | | | |
Collapse
|
237
|
|
238
|
Climent MA, Seguí MA, Peiró G, Molina R, Lerma E, Ojeda B, López-López JJ, Alonso C. Prognostic value of HER-2/neu and p53 expression in node-positive breast cancer. HER-2/neu effect on adjuvant tamoxifen treatment. Breast 2001; 10:67-77. [PMID: 14965564 DOI: 10.1054/brst.2000.0225] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
HER-2/neu and p53 expression, conventional clinical and pathologic prognostic factors, were evaluated in a retrospective series of 283 node-positive breast cancer patients. Overexpression was determined by immunohistochemistry in formalin-fixed paraffin-embedded tissue blocks. Twenty one percent were HER-2/neu positive and 40% p53 positive. HER-2/neu expression was related to axillary lymph node metastasis (P=0.014), inflammatory infiltrates (P=0.004), and the absence of oestrogen (ER) (P=0.0026) and progesterone (P=0.01) receptors (PR). p53 expression was related to lymph node involvement (P=0.03), necrosis (P=0.036), absence of ER (P=0.028) and PR (P=0.065). p53 was not associated with outcome. HER-2/neu was an unfavourable prognostic factor for disease-free (DFS) (P=0.05) and overall survival (OS) (P=0.02) in univariate analysis. Multivariate analysis showed that the number of involved axillary nodes (P<0.00001), age (P=0.004), grade (P=0.04), and PR (P=0.04) were independent predictors for OS. ER-positive patients treated with adjuvant tamoxifen had shorter DFS and OS when they were HER-2/neu positive.
Collapse
Affiliation(s)
- M A Climent
- Instituto Valenciano de Oncología, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
239
|
Schaich M, Ritter M, Illmer T, Lisske P, Thiede C, Schäkel U, Mohr B, Ehninger G, Neubauer A. Mutations in ras proto-oncogenes are associated with lower mdr1 gene expression in adult acute myeloid leukaemia. Br J Haematol 2001; 112:300-7. [PMID: 11167822 DOI: 10.1046/j.1365-2141.2001.02562.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mutations in ras genes have been found to be the most frequent genetic aberrations in adult myeloid leukaemia (AML). Some reports have shown an improved outcome of ras-mutated AML. In order to understand the biology of ras mutation in AML, we studied a cohort of patients treated in a prospective multicentre trial for ras mutational status and resistance gene expression. Blast samples from 162 adult patients with de novo or secondary AML were examined for resistance gene expression (mdr1, mrp1 and lrp) and ras mutations using reverse transcription-polymerase chain reaction and protein nucleic acid-competitive polymerase chain reaction strategies respectively. Ras mutations were confirmed using DNA sequencing. Ras mutations leading to an exchange of amino acids were found in 40 (25%) patients. Thirty AML patients had N-ras mutations and nine patients had K-ras mutations. One patient showed both N-ras and K-ras mutations. Resistance gene expression was positive for mdr1 in 30%, for mrp1 in 43% and for lrp in 62% of patients. There was a strong inverse correlation between the presence of ras mutation and mdr1 expression (P = 0.005). However, no significant difference was seen between patients with or without ras mutations and mrp1 or lrp expression. Whereas mdr1 expression was associated with a lower complete remission rate (P < 0.04), ras mutations had no significant influence on remission status. Neither ras mutation nor mdr1 expression had a significant impact on overall or disease-free survival to date. For the first time, there is evidence that activated ras genes are associated with lower mdr1 expression in AML.
Collapse
Affiliation(s)
- M Schaich
- Department of Medicine I, University Hospital Dresden, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
240
|
Wu Y, Mehew JW, Heckman CA, Arcinas M, Boxer LM. Negative regulation of bcl-2 expression by p53 in hematopoietic cells. Oncogene 2001; 20:240-51. [PMID: 11313951 DOI: 10.1038/sj.onc.1204067] [Citation(s) in RCA: 164] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2000] [Revised: 10/26/2000] [Accepted: 11/01/2000] [Indexed: 11/09/2022]
Abstract
The p53 protein activates promoters containing p53 binding sites, and it represses other promoters. We examined the effect of p53 on bcl-2 expression in both the DHL-4 B cell line and the K562 erythroleukemia line. Transient transfection analyses revealed that wild-type p53 repressed the bcl-2 full-length promoter. The region of the bcl-2 promoter that was responsive to p53 was mapped to the bcl-2 P2 minimal promoter region, and we showed that p53 and the TATA binding protein bound to the bcl-2 TATA sequence. The TATA binding protein, p53, histone deacetylase-1 and mSin3a could be co-immunoprecipitated from K562 cell nuclear extract. The TATA binding protein and mSin3a could be recovered in a complex at the bcl-2 promoter TATA sequence, however, the formation of this complex was not dependent on the presence of p53. Treatment of K562 cells with the histone deacetylase inhibitor, trichostatin A, resulted in an increase in bcl-2 promoter activity whether p53 was present or not. Therefore, we demonstrated that p53 and the histone deacetylases repress the bcl-2 promoter independently. Similar results were obtained when endogenous bcl-2 mRNA or protein levels were measured in response to either p53 or trichostatin A, and p53 expression resulted in enhanced apoptosis. RNase protection assays demonstrated that transcription from the endogenous 3' bcl-2 promoter was decreased by p53. The regions of p53 that were required for repression of the bcl-2 promoter were defined. We conclude that the TATA sequence in the bcl-2 P2 minimal promoter is the target for repression by p53, and that the interaction between p53 and TBP is most likely responsible for the repression. Mutation of p53 may play a role in the up-regulation of bcl-2 expression in some B cell lymphomas.
Collapse
Affiliation(s)
- Y Wu
- The Center for Molecular Biology in Medicine, Palo Alto Veterans Affairs Medical Center and the Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305-5112, USA
| | | | | | | | | |
Collapse
|
241
|
Lee CH. Induction of P-glycoprotein mRNA transcripts by cycloheximide in animal tissues: evidence that class I Pgp is transcriptionally regulated whereas class II Pgp is post-transcriptionally regulated. Mol Cell Biochem 2001; 216:103-10. [PMID: 11216854 DOI: 10.1023/a:1011086716568] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
P-glycoprotein (Pgp) are a small family of plasma membrane proteins capable of transporting substrates across cell membranes. Class I and class II Pgp are able to transport drugs and have been shown to mediate multidrug resistance (MDR). Class III Pgp is a long chain phospholipid transporter and does not mediate MDR. The expression and regulation of Pgp genes in animal tissues are not well understood. In this study, the protein synthesis inhibitor cycloheximide was used as a tool to understand Pgp gene expression and regulation in animal tissues. The sensitive RNase protection assay was used to detect changes in Pgp mRNA levels and nuclear run-on assay was used to determine whether transcription or post-transcription is important. The results showed that cycloheximide significantly induced class II Pgp expression in all tissues examined. This was predominantly through post-transcriptional effect. In contrast, the relatively modest increase in class I Pgp expression by cycloheximide was found to be mainly due to increased transcriptional activity. On the other hand, cycloheximide induced class III Pgp expression in some tissues while caused decay of class III Pgp mRNA in other tissues. The transcriptional and post-transcriptional mechanisms exerted by cycloheximide on Pgp genes are discussed. These findings have implications for our understanding of gene regulation in animal tissues and MDR reversal strategies in vivo.
Collapse
Affiliation(s)
- C H Lee
- Department of Chemistry, University of Northern British Columbia, Prince George, Canada
| |
Collapse
|
242
|
Delatte SJ, Hazen-Martin DJ, Re GG, Kelly JR, Sutphin A, Tagge EP. Restoration of p53 function in anaplastic Wilms' tumor. J Pediatr Surg 2001; 36:43-50. [PMID: 11150436 DOI: 10.1053/jpsu.2001.20002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND/PURPOSE Recent studies have reported a high incidence of p53 mutations in anaplastic Wilms' tumors (WT). Restoration of the normal p53 state by current gene therapy techniques is thus an attractive potential mode of therapy for this tumor, which is poorly responsive to standard therapy. The purpose of this study is to determine whether gene delivery of normal p53 is possible and to characterize the subsequent effect of restoring the wild-type p53 state. METHODS Anaplastic WT RM1 cells (mutant p53) were transduced with replication-deficient adenoviral vectors containing either the wild-type p53 gene (rAd-p53) or the gene encoding a green fluorescent protein (rAd-GFP). The transduction efficiency of adenovirus for RM1 cells was determined by flow cytometric analysis of rAd-GFP-transduced cells. The effect of p53 transduction on cell viability was evaluated using a colorimetric proliferation assay. Apoptosis was evaluated by labeling DNA breaks using a TUNEL assay (Apo-Direct kit). RESULTS Cells treated with increasing concentrations of viral particles relative to tumor cells (multiplicity of infection-MOI) showed a dose-dependent increase in the number of cells transduced. Twenty-four hours after viral treatment, the percentage of cells transduced for MOIs of 10, 50, 100, and 500 was 29.5, 60.9, 74.6, and 92.4, respectively; at 48 hours the percentage of cells transduced increased to 70.8, 90.7, 93. 7, and 96.3, respectively. Viral treatment at an MOI of 50 reduced cell proliferation by 10% at 17 hours and 97% at 5 days; at an MOI of 100, the relative reduction in proliferation was 15% and 99.8%, respectively. When assayed, 30% of cells became apoptotic at an MOI of 50, and 48% at an MOI of 100. CONCLUSIONS Highly efficient delivery of the p53 tumor suppressor gene by adenoviral vector to anaplastic WT is possible. Subsequent restoration of the normal p53 state results in reduced viability and increased apoptosis. Gene replacement of p53 may represent a novel therapeutic agent for anaplastic Wilms' tumors.
Collapse
Affiliation(s)
- S J Delatte
- Division of Pediatric Surgery, Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | | | |
Collapse
|
243
|
Lackinger D, Kaina B. Primary mouse fibroblasts deficient for c-Fos, p53 or for both proteins are hypersensitive to UV light and alkylating agent-induced chromosomal breakage and apoptosis. Mutat Res 2000; 457:113-23. [PMID: 11106803 DOI: 10.1016/s0027-5107(00)00133-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The important regulatory proteins, c-Fos and p53 are induced by exposure of cells to a variety of DNA damaging agents. To investigate their role in cellular defense against genotoxic compounds, we comparatively analysed chromosomal aberrations and apoptosis induced by ultraviolet (UV-C) light and the potent alkylating agent methyl methanesulfonate (MMS) in primary diploid mouse fibroblasts knockout for either c-Fos or p53, or double knockout for both genes. We show that c-Fos and p53 deficient fibroblasts are more sensitive than the corresponding wild-type cells as to the induction of chromosomal aberrations and apoptosis. Double knockout fibroblasts lacking both c-Fos and p53 are viable and were even more sensitive, showing additivity of the chromosomal breakage effects observed in the single knockouts. Regarding the endpoint apoptosis, double knockout fibroblasts displayed a sensitivity similar to c-Fos and p53 deficient cells. The data indicate that (a) both c-Fos and p53 are involved in cellular protection against the clastogenic effect of genotoxic agents, (b) p53 is not required for induction of apoptosis by UV light and MMS, but rather prevents fibroblasts from undergoing apoptotic cell death upon DNA damage, and (c) c-Fos and p53 seem to act independently in determining genotoxic resistance, which is hypothesized to be achieved by impaired DNA repair or differential cell cycle check point control.
Collapse
Affiliation(s)
- D Lackinger
- Institute of Toxicology, Division of Applied Toxicology, University of Mainz, Obere Zahlbacher Str. 67, D-55131, Mainz, Germany
| | | |
Collapse
|
244
|
Chicas A, Molina P, Bargonetti J. Mutant p53 forms a complex with Sp1 on HIV-LTR DNA. Biochem Biophys Res Commun 2000; 279:383-90. [PMID: 11118296 DOI: 10.1006/bbrc.2000.3965] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Many mutants of p53 activate HIV-LTR driven transcription and promote HIV replication. The region of the HIV-LTR containing Sp1-binding sites is important for this effect. In this study we test the hypothesis that mutant p53 interacts with DNA-bound Sp1 and in this way can increase transcription from Sp1-dependent promoters. We have used the breast cancer cell line MDA-MB-468 that expresses endogenous mutant p53(His273) as our source of p53 protein. First, we demonstrated that this mutant p53 participates in activating transcription from the HIV-LTR by showing that HIV-LTR-directed transcription in MDA-MB-468 cells is inhibited in a dominant-negative manner by p53(Val135). Using HIV-LTR DNA affinity chromatography, we detected coelution of p53(His273) and Sp1. We also demonstrated that this mutant p53 binds sequence specifically to the super consensus sequence (SCS) and that Sp1 coeluted with p53(His273) from a column containing this site. These data indicate that p53(His273) can associate with DNA-bound Sp1 suggesting that activated HIV-LTR transcription associated with mutant p53 occurs through a DNA driven multi-protein complex.
Collapse
Affiliation(s)
- A Chicas
- Department of Biological Sciences, Institute for Biomolecular Structure and Function, Hunter College, 695 Park Avenue, New York, New York, 10021, USA
| | | | | |
Collapse
|
245
|
Okamoto T, Izumi H, Imamura T, Takano H, Ise T, Uchiumi T, Kuwano M, Kohno K. Direct interaction of p53 with the Y-box binding protein, YB-1: a mechanism for regulation of human gene expression. Oncogene 2000; 19:6194-202. [PMID: 11175333 DOI: 10.1038/sj.onc.1204029] [Citation(s) in RCA: 124] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The Y-box binding protein, YB-1, belongs to a family of multifunctional proteins which regulate gene expression on both transcriptional and translational levels. The tumor suppressor gene p53 displays growth suppressive properties by regulating gene expression through transcriptional regulation. We now demonstrate that YB-1 directly interacts with p53 using an in vitro pull-down assay. Using immunochemical co-precipitation methods, we also found that the two proteins are bound in vivo. Deletion analysis showed that three independent domains of YB-1, one at the N-terminal and two at the C-terminal, interact with p53. Conversely, a 14 amino acid sequence at the C-terminal of p53 was required for its interaction with YB-1. Gel mobility shift assays showed that the interaction of YB-1 with p53 stimulated the sequence-specific DNA binding of p53 to its consensus sequence. By contrast, this interaction inhibited the binding of YB-1. Using a p53-responsive p21 promoter linked to a reporter gene, it can be shown that antisense expression of YB-1 inhibits the induction of this promoter by p53 in transient transfection assays. These findings delineate a straightforward mechanism for gene expression through p53-YB-1 interaction.
Collapse
Affiliation(s)
- T Okamoto
- Department of Molecular Biology, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu 807-8555, Japan
| | | | | | | | | | | | | | | |
Collapse
|
246
|
Johnsen JI, Aurelio ON, Kwaja Z, Jörgensen GE, Pellegata NS, Plattner R, Stanbridge EJ, Cajot JF. p53-mediated negative regulation of stathmin/Op18 expression is associated with G(2)/M cell-cycle arrest. Int J Cancer 2000; 88:685-91. [PMID: 11072234 DOI: 10.1002/1097-0215(20001201)88:5<685::aid-ijc1>3.0.co;2-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Utilizing the technique of differential display of mRNA, we have identified p53-responsive genes that are transcriptionally up- or down-regulated as cells enter growth arrest. One gene that was down-regulated, pong16, was found to be identical to stathmin/Op18, a protein involved in the regulation of microtubule dynamics. Evidence that p53 is directly or indirectly involved in negative regulation of stathmin/Op18 expression includes the following: (i) p53-mediated growth inhibition is associated with repression of stathmin/Op18 expression following serum stimulation, (ii) reporter gene assays revealed p53-mediated repression of stathmin/Op18 promoter activity and (iii) constitutive over-expression of stathmin/Op18 bypasses a p53-mediated G(2)/M arrest in the cell cycle. These results suggest that p53-mediated negative regulation of stathmin/Op18 plays an important role in cell-cycle control.
Collapse
Affiliation(s)
- J I Johnsen
- Department of Virology, Faculty of Medicine, University of Tromso, Tromso Norway
| | | | | | | | | | | | | | | |
Collapse
|
247
|
Lange C, Walther W, Schwabe H, Stein U. Cloning and initial analysis of the human multidrug resistance-related MVP/LRP gene promoter. Biochem Biophys Res Commun 2000; 278:125-33. [PMID: 11071864 DOI: 10.1006/bbrc.2000.3782] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The lung resistance-related protein (LRP) was identified as the human major vault protein (MVP), and is overexpressed in various multidrug-resistant cancer cell lines and clinical samples. We characterized DNA sequences upstream to the transcription initiation site of the MVP gene in the human non-small cell lung cancer cell line SW-1573. A 1.9-kb and a shortened 0.7-kb fragment of the 5'-upstream genomic region show strong promoter activity in chloramphenicol acetyltransferase (CAT) reporter assays. The promoter is TATA-less and contains an inverted CCAAT-box and a Sp1 site located near to a p53 binding motif. An alternative 3'-splice site of intron 1 results in a splicing variant within the 5'-untranslated region of MVP mRNA.
Collapse
MESH Headings
- 5' Untranslated Regions
- Alternative Splicing
- Base Sequence
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Chloramphenicol O-Acetyltransferase/metabolism
- Cloning, Molecular
- Colonic Neoplasms/genetics
- Colonic Neoplasms/metabolism
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/genetics
- Electrophoresis, Agar Gel
- Enzyme-Linked Immunosorbent Assay
- Genes, Reporter
- Humans
- Introns
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Models, Genetic
- Molecular Sequence Data
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Plasmids/metabolism
- Promoter Regions, Genetic
- RNA, Messenger
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Analysis, DNA
- Sp1 Transcription Factor/genetics
- TATA Box
- Transcription, Genetic
- Transfection
- Tumor Cells, Cultured
- Tumor Suppressor Protein p53/metabolism
- Up-Regulation
- Vault Ribonucleoprotein Particles/genetics
- Vault Ribonucleoprotein Particles/metabolism
Collapse
Affiliation(s)
- C Lange
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13092 Berlin, Germany
| | | | | | | |
Collapse
|
248
|
Preuss U, Kreutzfeld R, Scheidtmann KH. Tumor-derived p53 mutant C174Y is a gain-of-function mutant which activates the fos promoter and enhances colony formation. Int J Cancer 2000; 88:162-71. [PMID: 11004663 DOI: 10.1002/1097-0215(20001015)88:2<162::aid-ijc3>3.0.co;2-m] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
SV40 large T antigen-induced primitive neuroectodermal tumors of the rat provide a model system to study induction and progression of primitive neuroectodermal tumors at the molecular level. A cell line derived from such a tumor reproducibly gave rise to malignant derivatives that ceased large T-antigen expression but harbored a mutant p53 allele with a common mutation at Cys(174) to Tyr (C174Y). To determine whether this p53 mutation contributes to tumor progression, we analyzed mutant C174Y functionally. Co-transfection experiments in Saos-2 cells with mutant or wild-type p53 and reporter genes linked to various p53-responsive promoters revealed that mutant C174Y failed to transcriptionally transactivate the Mdm2, Waf1, Cyclin G and Bax promoters. Loss of transcriptional activation correlated with loss of DNA-binding activity. Moreover, mutant C174Y exhibited a dominant negative effect on co-expressed wild-type p53. The ability of mutant p53 to repress the viral RSV, LTR or SV40 early promoters or the cellular fos promoter was likewise impaired. In contrast, it showed even induction of the fos promoter. Consistent with these observations, mutant C174Y was non-functional in the suppression of Saos-2 cell growth and even conferred a growth advantage to the cells. Surprisingly, mutant C174Y was also impaired in nuclear transport, as revealed by immunofluorescence analyses. Taken together, our results demonstrate that mutant C174Y possesses features that can positively contribute to cancer progression.
Collapse
Affiliation(s)
- U Preuss
- Institut für Genetik, Abteilung Molekulargenetik, Universität Bonn, Bonn, Germany.
| | | | | |
Collapse
|
249
|
Lacueva FJ, Calpena R, Medrano J, Teruel A, Mayol MJ, Graells ML, Camarasa MV, Perez-Vazquez MT, Ferragut JA. Changes in P-glycoprotein expression in gastric carcinoma with respect to distant gastric mucosa may be influenced by p53. Cancer 2000. [PMID: 10896996 DOI: 10.1002/1097-0142(20000701)89:1%3c21::aid-cncr4%3e3.0.co;2-t] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The effectiveness of some chemotherapeutic agents used to treat gastric carcinoma patients may be impaired by the presence of P-glycoprotein (P-gp) and the status of p53. A modulation of P-gp expression by p53 or other alterations during tumorigenesis have been reported. The authors analyzed P-gp expression in relation to p53 and histopathologic features in gastric carcinoma. METHODS Forty-one resected gastric carcinomas and mucosa distant from the tumor were assessed for P-gp expression by immunohistochemistry with C494 and JSB-1 antibodies. p53 expression was also immunohistochemically assessed by DO7 antibody in tumor samples. P-gp and p53 expression were semiquantitatively analyzed according to the percentage of stained cells. Histologic type, grade, vessel invasion, and stage were also studied. RESULTS Moderate or high P-gp expression was detected in gastric carcinoma in 29 cases (71%) and in gastric mucosa remote from the tumor in 36 cases (88%). This reduction in P-gp expression was observed in 22% of the carcinomas, all but 1 being p53 immunonegative tumors. Thus, 8 (42%) of the p53 immunonegative carcinomas showed a loss of P-gp expression compared with their distant gastric mucosa. All p53 immunopositive carcinomas coexpressed P-gp. No correlation between P-gp expression and histologic type, grade, vessel invasion, or stage was found. CONCLUSIONS P-gp expression in gastric carcinomas is frequent and coexpression with p53 is found. The analysis of P-gp expression in carcinomas and distant mucosa show that it is not regulated by p53, but a loss of P-gp detected in some of these carcinomas is mainly associated with a lack of p53 protein accumulation.
Collapse
Affiliation(s)
- F J Lacueva
- Department of Pathology and Surgery, School of Medicine, Miguel Hernandez University, Alicante, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
250
|
van Zuylen L, Nooter K, Sparreboom A, Verweij J. Development of multidrug-resistance convertors: sense or nonsense? Invest New Drugs 2000; 18:205-20. [PMID: 10958589 DOI: 10.1023/a:1006487003814] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review describes the clinical relevance of the two drug transporters P-glycoprotein (Pgp) and multidrug resistance-associated protein (MRP) and the in vitro phenomenon which is referred to as multidrug resistance (MDR). The attempts to try to block these resistance mechanisms are summarized with specific attention for the intentionally designed "second generation" MDR-convertors. Potential explanations of the limited clinical success rate are given and recommendations for the design of future studies provided.
Collapse
Affiliation(s)
- L van Zuylen
- Department of Medical Oncology, Rotterdam Cancer Institute (Daniel den Hoed Kliniek) and University Hospital, The Netherlands.
| | | | | | | |
Collapse
|