Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
Oncogenesis ; 3: e84, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24418892

ABSTRACT

We have investigated some roles of splicing factor polypyrimidine tract-binding protein (PTBP1) in human breast cancer. We found that PTBP1 was upregulated in progressively transformed human mammary epithelial cells (HMECs), as well as in breast tumor cell lines compared with HMECs with finite growth potential and found that the level of PTBP1 correlated with the transformation state of HMECs. Knockdown of PTBP1 expression substantially inhibited tumor cell growth, colony formation in soft agar and in vitro invasiveness of breast cancer cell lines, a result similar to what we have reported in ovarian cancer. However, ectopic expression of PTBP1 (as a PTBP1-EGFP fusion protein) did not enhance the proliferation of immortalized HMEC. Rather, PTBP1 expression promoted anchorage-independent growth of an immortalized HMEC as assessed by increased colony formation in soft agar. In addition, we found that knockdown of PTBP1 expression led to upregulation of the expression of the M1 isoform of pyruvate kinase (PKM1) and increase of the ratio of PKM1 vs PKM2. PKM1 has been reported to promote oxidative phosphorylation and reduce tumorigenesis. Correspondingly, we observed increased oxygen consumption in PTBP1-knockdown breast cancer cells. Together, these results suggest that PTBP1 is associated with breast tumorigenesis and appears to be required for tumor cell growth and maintenance of transformed properties. PTBP1 exerts these effects, in part, by regulating the splicing of pyruvate kinase, and consequently alters glucose metabolism and contributes to the Warburg effect.

2.
Oncogene ; 30(3): 356-65, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20856201

ABSTRACT

Our previous study revealed that two splicing factors, polypyrimidine tract-binding protein (PTB) and SRp20, were upregulated in epithelial ovarian cancer (EOC) and knockdown of PTB expression inhibited ovarian tumor cell growth and transformation properties. In this report, we show that knockdown of SRp20 expression in ovarian cancer cells also causes substantial inhibition of tumor cell growth and colony formation in soft agar and the extent of such inhibition appeared to correlate with the extent of suppression of SRp20. Massive knockdown of SRp20 expression triggered remarkable apoptosis in these cells. These results suggest that overexpression of SRp20 is required for ovarian tumor cell growth and survival. Immunohistochemical staining for PTB and SRp20 of two specialized tissue microarrays, one containing benign ovarian tumors, borderline/low malignant potential (LMP) ovarian tumors as well as invasive EOC and the other containing invasive EOC ranging from stage I to stage IV disease, reveals that PTB and SRp20 are both expressed differentially between benign tumors and invasive EOC, and between borderline/LMP tumors and invasive EOC. There were more all-negative or mixed staining cases (at least two evaluable section cores per case) in benign tumors than in invasive EOC, whereas there were more all-positive staining cases in invasive EOC than in the other two disease classifications. Among invasive EOC, the majority of cases were stained all positive for both PTB and SRp20, and there were no significant differences in average staining or frequency of positive cancer cells between any of the tumor stages. Therefore, the expression of PTB and SRp20 is associated with malignancy of ovarian tumors but not with stage of invasive EOC.


Subject(s)
Gene Knockdown Techniques , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , RNA-Binding Proteins/physiology , Base Sequence , Cell Division/genetics , DNA Primers , Female , Humans , Immunohistochemistry , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , RNA Interference , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors
3.
Oncogene ; 26(34): 4961-8, 2007 Jul 26.
Article in English | MEDLINE | ID: mdl-17310993

ABSTRACT

Polypyrimidine tract-binding protein (PTB) is an RNA-binding protein with multiple functions in the regulation of RNA processing and IRES-mediated translation. We report here overexpression of PTB in a majority of epithelial ovarian tumors revealed by immunoblotting and tissue microarray (TMA) staining. By western blotting, we found that PTB was overexpressed in 17 out of 19 ovarian tumor specimens compared to their matched-normal tissues. By TMA staining, we found PTB expression in 38 out of 44 ovarian cancer cases but only in two out of nine normal adjacent tissues. PTB is also overexpressed in SV40 large T-antigen immortalized ovarian epithelial cells compared to normal human ovarian epithelial cells. Using doxycycline-inducible small interfering RNA technology, we found that knockdown of PTB expression in the ovarian tumor cell line A2780 substantially impaired tumor cell proliferation, anchorage-independent growth and in vitro invasiveness. These results suggest that overexpression of PTB is an important component of the multistep process of tumorigenesis, and might be required for the development and maintenance of epithelial ovarian tumors. Moreover, because of its novel role in tumor cell growth and invasiveness, shown here for the first time, PTB may be a novel therapeutic target in the treatment of ovarian cancer.


Subject(s)
Ovarian Neoplasms/pathology , Polypyrimidine Tract-Binding Protein/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation , Female , Humans , Immunohistochemistry , Neoplasm Invasiveness , Ovarian Neoplasms/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , RNA Interference , Tissue Array Analysis
4.
Biochem Soc Trans ; 29(Pt 6): 702-3, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11709058

ABSTRACT

DNA topoisomerase (topo) II inhibitors either stabilize DNA-topo II complexes by blocking DNA religation (e.g. etoposide) or block the enzyme's catalytic activity (e.g. dexrazoxane). The former class of drugs causes direct DNA damage through topo II, while the latter class does not, but both classes cause apoptosis. We cloned the Fas ligand (FasL) promoter and coupled it to the luciferase gene. Treatment of cells transfected with this construct revealed that complex-stabilizing (DNA-damaging) agents induce FasL expression, but the catalytic inhibitors do not, suggesting that the FasL pathway may not be involved in all cases of topoisomerase-mediated apoptosis. Some topo II inhibitors activate a pathway involving stress-activated protein kinases, which include c-Jun N-terminal kinase-1 (JNK-1). We will discuss the effects of these agents on components of this pathway. Our earlier work revealed that topo IIalpha interacts with the cell cycle regulatory protein, retinoblastoma protein (Rb). This interaction and the subcellular distribution of these proteins are altered by topo II inhibitory drugs and lead to apoptosis. In addition, agents that affect Rb, such as E1A and E2F1/DP-1, when transfected into cells, also alter topo IIalpha-Rb localization, activate jun kinase pathways and cause apoptosis. This paper discusses current studies that are designed to determine the contributions of these signalling events to the alterations in subcellular protein distribution and apoptosis. We suggest that protein-protein interactions are important for mediation of cytotoxic signalling by anticancer drugs.


Subject(s)
Enzyme Inhibitors/pharmacology , Signal Transduction , Topoisomerase II Inhibitors , Animals , Apoptosis , Fas Ligand Protein , Humans , Membrane Glycoproteins/metabolism , Protein Binding , Tumor Cells, Cultured
5.
J Med Chem ; 44(18): 2959-65, 2001 Aug 30.
Article in English | MEDLINE | ID: mdl-11520204

ABSTRACT

Three classes of hydroxy-tethered platinum(II) complexes have been synthesized from K(2)PtCl(4) and appropriate amino alcohols. A sequence of selective oxidation and hydrolysis has been developed to prepare hydroxy-tethered platinum(IV) complexes. A novel procedure for the synthesis of amminetrichloroplatinate(II) anion has been generated and used to synthesize a number of monohydroxy-tethered nonchelating platinum complexes. These tethered platinum complexes, including hydroxy-tethered, phosphoramidite-tethered, and monodeoxyribonucleotide-tethered platinum(II) and -(IV) complexes, have been examined in vitro for antitumor activity in both leukemia and ovarian cancer cell lines. Activity of some of these complexes was similar to cis-platin, and most of them showed much better potency than carboplatin. We observed an interesting structure-activity correlation for platinum(II) complexes for both PA-1 and SK-OV-3 ovarian cancer cell lines. However, platinum(IV) complexes showed much more diversified response among cancer cell lines studied. We observed enhanced selectivity among different cancer cell lines for some agents. The most promising is the monodeoxyribonucleotide-tethered platinum(IV) complex, which is the first analogue of the conjugates between a platinum fragment and monodeoxyribonucleotides, showing antitumor activity and selectivity among the cell lines. Finally, the p53 status of the cells appears to contribute to the effectiveness of these agents in that cells harboring wild-type p53 appear to be more sensitive to these agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Oligonucleotides/chemistry , Organoplatinum Compounds/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Division , Drug Screening Assays, Antitumor , Humans , Magnetic Resonance Spectroscopy , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured
6.
Mol Pharmacol ; 59(2): 203-11, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160854

ABSTRACT

DNA topoisomerase (topo) IIalpha gene expression or activity is altered in tumor cells selected for resistance to inhibitors of topoII. To better understand the mechanisms by which topoIIalpha expression levels are modulated, we examined topoIIalpha transcriptional regulation in ICRF-187-sensitive and ICRF-187-resistant human leukemic cell lines that express an increased amount of topoIIalpha protein and mRNA. Transient transfections of luciferase reporter plasmids containing either the full-length human topoIIalpha promoter or fragments of it revealed that topoIIalpha transcriptional activity was significantly increased in the drug-resistant CEM/ICRF-8 cells, compared with CEM cells. Specifically, the transcriptional activity of the full-length topoIIalpha promoter (nucleotides -557 to +90) was doubled in CEM/ICRF-8 compared with CEM cells. Serial deletion of the topoIIalpha promoter permitted localization of the region responsible for its up-regulation in the drug-resistant cells between nucleotides -557 and -162, which includes the last three inverted CCAAT elements (ICE) 3 to 5. Note that construction of a point mutation in ICE3 resulted in a significant increase in transcriptional activity of the topoIIalpha promoter in the drug-sensitive CEM cells. In addition, by electrophoretic mobility shift assay, ICE3 was recognized by a protein complex containing NF-YB that was present at reduced levels in the topoIIalpha-overexpressing CEM/ICRF-8 extracts, suggesting that ICE3 plays a negative regulatory role in human topoIIalpha gene expression. This is the first study to show that topoIIalpha transcriptional up-regulation in ICRF-187-resistant cells is mediated in part by altered regulation of the third inverted CCAAT box in the topoIIalpha promoter.


Subject(s)
DNA Topoisomerases, Type II , DNA Topoisomerases, Type II/genetics , Gene Expression Regulation , Isoenzymes/genetics , Promoter Regions, Genetic/physiology , Razoxane/pharmacology , Transcription Factors , Antigens, Neoplasm , Antineoplastic Agents/pharmacology , CCAAT-Binding Factor/analysis , CCAAT-Binding Factor/physiology , CCAAT-Enhancer-Binding Proteins/analysis , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/analysis , Down-Regulation , Drug Resistance, Neoplasm/genetics , Humans , Isoenzymes/metabolism , NFI Transcription Factors , Nuclear Proteins , Promoter Regions, Genetic/genetics , Tumor Cells, Cultured , Y-Box-Binding Protein 1
7.
J Biol Chem ; 275(52): 41107-13, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11016921

ABSTRACT

DNA topoisomerase (topo) I is a nuclear enzyme that plays an important role in DNA metabolism. Based on conserved nuclear targeting sequences, four classic nuclear localization signals (NLSs) have been proposed at the N terminus of human topo I, but studies with yeast have suggested that only one of them (amino acids (aa) 150-156) is sufficient to direct the enzyme to the nucleus. In this study, we expressed human topo I fused to enhanced green fluorescent protein (EGFP) in mammalian cells and demonstrated that whereas aa 150-156 are sufficient for nuclear localization, the nucleolar localization requires aa 157-199. More importantly, we identified a novel NLS within aa 117-146. In contrast to the classic NLSs that are rich in basic amino acids, the novel NLS identified in this study is rich in acidic amino acids. Furthermore, this novel NLS alone is sufficient to direct not only EGFP into the nucleus but also topo I; and the EGFP.topo I fusion driven by the novel NLS is as active in vivo as the wild-type topo I in response to the topo I inhibitor topotecan. Together, our results suggest that human topo I carries two independent NLSs that have opposite amino acid compositions.


Subject(s)
Cell Nucleus/enzymology , DNA Topoisomerases, Type I/chemistry , Amino Acid Sequence , Cell Nucleolus/enzymology , DNA Topoisomerases, Type I/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Topoisomerase I Inhibitors , Topotecan/pharmacology
8.
Oncogene ; 19(43): 5010-9, 2000 Oct 12.
Article in English | MEDLINE | ID: mdl-11042688

ABSTRACT

We examined p53 protein stability and DNA damage-induced p53-dependent responses in a human leukemic CEM cell line and two teniposide-resistant sublines, CEM/VM-1 and CEM/VM-1-5 ( approximately 40 and 400-fold resistant to teniposide, respectively). Although all cell lines contain the same p53 mutations at codons 175 (Arg-->His) and 248 (Arg-->Gln), the constitutive levels of p53 were progressively increased with the resistance of the cells to teniposide. By pulse-chase experiments, we found that the half-lives of mutant p53 protein were approximately 12, 17, and >30 h in CEM, CEM/VM-1, and CEM/VM-1-5 cells, respectively. The prolonged half-lives of p53 in these cells is consistent with the fact that the protein harbors the indicated mutations. Of note, however, is the fact that the increased p53 protein half-lives in the two drug-resistant cell lines corresponds to a proportional decrease in MDM2 protein levels but an increase in p53-MDM2 binding interactions. This suggests that MDM2-mediated p53 degradation may be altered in our leukemic cell lines. The DNA damage-induced p53 response is fully functional in the drug-sensitive CEM cells containing a mutant p53, but this pathway is attenuated in the drug-resistant cells. Specifically, while the mutant p53 was phosphorylated at serine-15 in response to ionizing radiation in all these cell lines, mutant p53 induction in response to teniposide or ionizing radiation and induction of the p53-target genes, p21 and GADD45 only occurred in the drug-sensitive CEM cells. As assessed by MTT cytotoxicity assay, CEM cells were also significantly more sensitive to ionizing radiation, compared to the drug-resistant cell lines, and this correlated with p53 induction. Collectively, these results suggest that changes in constitutive mutant p53 protein levels, p53-MDM2 binding interactions, and altered regulation of the DNA damage-inducible p53-dependent pathway may play a role in drug- and radiation-responsiveness in these cells.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Damage/genetics , Nuclear Proteins , Teniposide/pharmacology , Tumor Suppressor Protein p53/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Drug Resistance, Neoplasm , Humans , Intracellular Signaling Peptides and Proteins , Leukemia/drug therapy , Leukemia/genetics , Leukemia/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphorylation/radiation effects , Protein Binding , Protein Biosynthesis , Proteins/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-mdm2 , Transcriptional Activation/drug effects , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein , GADD45 Proteins
9.
Exp Cell Res ; 256(2): 480-90, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10772820

ABSTRACT

DNA topoisomerase (topo) I plays an important role in DNA metabolism by relieving the torsional restraints of DNA topology through ATP-independent single-strand DNA breakage. In the present study, we expressed human topo I in HeLa cells by fusing it to enhanced green fluorescent protein (EGFP). The EGFP-topo I fusion protein is functionally active in that it relaxes supercoiled plasmid DNA; forms complexes with DNA, as revealed by band depletion assays; and increases the sensitivity of cells to topo I inhibitors such as topotecan, as determined by growth inhibition assays. In contrast, a mutant form of the EGFP-topo I fusion protein, in which the active Tyr has been replaced by Phe (Y723F), has no such activities. Furthermore, the fusion protein localizes to the nucleus at interphase and completely associates with chromatids at every stage of mitosis. Of importance, the mutant fusion protein (Y723F) displays a pattern of subcellular localization identical to that of the wild-type fusion protein, although the mutant fusion protein is catalytically inactive. These results suggest that in addition to its role in DNA metabolism, topo I might also play a structural role in chromosomal organization; moreover, the association of topo I with chromosomal DNA is independent of its catalytic activity. Finally, the fusion constructs may provide a useful tool to study drug action in tumor cells, as demonstrated by nucleolar delocalization of the fusion proteins in response to treatment with the topo I inhibitor topotecan.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Amino Acid Substitution , Cell Nucleus/enzymology , Chromosomes, Human , DNA/metabolism , DNA Topoisomerases, Type I/genetics , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins , HeLa Cells , Humans , Immunoblotting , Interphase , Luminescent Proteins/genetics , Microscopy, Fluorescence , Mitosis , Mutation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Subcellular Fractions/enzymology , Topoisomerase I Inhibitors , Topotecan/pharmacology
10.
Mol Pharmacol ; 57(2): 296-307, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10648639

ABSTRACT

ICRF-187 is a bisdioxopiperazine anticancer drug that inhibits the catalytic activity of DNA topoisomerase (topo) II without stabilizing DNA-topoII cleavable complexes. To better understand the mechanisms of action of and resistance to topoII catalytic inhibitors, human leukemic CEM cells were selected for resistance to ICRF-187. The clones CEM/ICRF-8 and CEM/ICRF-18 are approximately 40- and 69-fold resistant to ICRF-187, and 12- and 67-fold cross-resistant to ICRF-193, respectively, but are sensitive to other topoII catalytic inhibitors (merbarone and aclarubicin), as well as collaterally sensitive to the DNA-topoII complex-stabilizing drug etoposide (VP-16). Both the number of VP-16- induced DNA-topoII complexes formed and the amount of in vitro topoII catalytic activity are enhanced in the drug-resistant cells. The ICRF-187-resistant clones contain approximately 5-fold increase in topoIIalpha protein levels and approximately 2.2-fold increase in topoIIalpha mRNA levels. Furthermore, CEM/ICRF-8 expresses approximately 3.5-fold increase in topoIIalpha promoter activity, suggesting that up-regulation of topoIIalpha in this clone occurs at the transcriptional level. Treatment of the drug-resistant or -sensitive cells with equitoxic doses of merbarone or teniposide results in a G(2)/M arrest. In marked contrast, when treated with equitoxic ICRF-187 doses, the drug-resistant clones exhibit either a transient arrest or completely lack the G(2)/M checkpoint compared with the drug-sensitive cells. This aberrant cell cycle profile is associated with a 48-h delay in drug-induced apoptotic cell death, as revealed by fluorescent-end labeling of DNA and poly (ADP-ribose) polymerase cleavage. In summary, resistance to ICRF-187 in CEM cells is associated with increased levels of catalytically active topoIIalpha and altered G(2)/M checkpoint and apoptotic responses.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , DNA Topoisomerases, Type II/metabolism , Enzyme Inhibitors/pharmacology , Isoenzymes/metabolism , Razoxane/pharmacology , Topoisomerase II Inhibitors , Antigens, Neoplasm , Antineoplastic Agents, Phytogenic/pharmacology , DNA Fragmentation/drug effects , DNA, Complementary/analysis , DNA-Binding Proteins , Drug Resistance, Neoplasm , Etoposide/pharmacology , G2 Phase/drug effects , Humans , Mitosis/drug effects , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , S Phase/drug effects , Sequence Analysis , Tumor Cells, Cultured
11.
Exp Cell Res ; 252(1): 50-62, 1999 Oct 10.
Article in English | MEDLINE | ID: mdl-10502399

ABSTRACT

DNA topoisomerase (topo) II is an essential nuclear enzyme that plays an important role in DNA metabolism and chromosome organization. In the present study, we expressed human topo IIalpha in mammalian cells by fusion to an enhanced green fluorescent protein (EGFP). Decatenation assays indicated that the EGFP-topo IIalpha is catalytically active in vitro. Assays for band depletion, growth inhibition, and cytotoxicity by topo II inhibitors suggested that the fusion protein is also functional in vivo. By following its subcellular localization throughout the cell cycle in living cells, we found that the fusion protein is localized to the nucleus and nucleolus at interphase, and it is bound to chromosomal DNA at every stage of mitosis. Of importance, a mutant EGFP-topo IIalpha, in which the active Tyr 805 is replaced by Phe (Y805F) and is catalytically inactive, still binds to chromosomal DNA throughout the cell cycle like the wild-type enzyme. Together, our results suggest that the ability of topo IIalpha to bind to chromosomal DNA in the cell, a presumed requirement for its structural role, can be separated from its catalytic activity.


Subject(s)
Chromosomes/enzymology , DNA Topoisomerases, Type II , DNA Topoisomerases, Type II/metabolism , Isoenzymes/metabolism , Mitosis/physiology , Antigens, Neoplasm , Base Sequence , Cell Line , Cell Nucleus/enzymology , Cytoplasm/enzymology , DNA Primers/genetics , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Gene Expression , Green Fluorescent Proteins , HeLa Cells , Humans , Interphase/physiology , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mutagenesis, Site-Directed , Point Mutation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Teniposide/pharmacology , Topoisomerase II Inhibitors
12.
Biochem Pharmacol ; 58(8): 1247-57, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10487526

ABSTRACT

Dexrazoxane (ICRF-187) is an inhibitor of the catalytic activity of DNA topoisomerase II (topo II) that does not stabilize DNA-topo II covalent complexes. Here, we examined cytotoxic signaling by ICRF-187 in human leukemic CEM cells and a teniposide (VM-26)-resistant subline, CEM/VM-1. Treatment of CEM and CEM/VM-1 cells with ICRF-187 induced apoptotic cell death characterized by internucleosomal DNA fragmentation, nuclear condensation, and induction of at least caspase-3- and -7-like protease activities (but not caspase 1). Treatment of these cells with Z-Asp-2,6-dichlorobenzoyloxymethyl-ketone, a potent inhibitor of apoptosis, inhibited ICRF-187-induced DEVD-specific caspase activity and apoptosis in a concentration-dependent manner. ICRF-187-induced apoptosis in CEM cells was associated with transient induction of c-jun and activation of c-Jun NH2-terminal kinase 1 (JNK1). However, CEM/VM-1 cells, which were 3-fold more sensitive than CEM cells to ICRF-187 due to a decrease in topo II activity, exhibited ICRF-187-induced apoptosis in the absence of c-jun induction and JNK1 activation. These results indicate that catalytic inhibition of topo II by ICRF-187 leads to apoptosis through at least a caspase-3- and -7-like protease-dependent mechanism and suggest that c-jun and JNK1 are not required in ICRF-187-induced apoptosis in CEM cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinases , Proto-Oncogene Proteins c-jun/metabolism , Razoxane/pharmacology , Teniposide/pharmacology , Asparagine/analogs & derivatives , Asparagine/pharmacology , Caspases/biosynthesis , Cell Division/drug effects , Drug Resistance, Neoplasm , Enzyme Induction , Gene Expression/drug effects , Humans , JNK Mitogen-Activated Protein Kinases , Signal Transduction , Topoisomerase II Inhibitors , Tumor Cells, Cultured
13.
Proc Natl Acad Sci U S A ; 96(14): 7859-64, 1999 Jul 06.
Article in English | MEDLINE | ID: mdl-10393912

ABSTRACT

DNA topoisomerase II-an essential nuclear enzyme in DNA replication and transcription, chromatin segregation, and cell cycle progression-is also a target of clinically useful anticancer drugs. Preliminary observations of a positive correlation between the expression of topoisomerase (topo) IIalpha and the retinoblastoma protein (Rb) in a series of rhabdomyosarcoma cells prompted us to ask whether these two proteins interact in vivo. Using human rhabdomyosarcoma and leukemic cell lines, we found a physical association between topo IIalpha and Rb protein by reciprocal immunoprecipitation and immunoblotting, in which topo IIalpha appeared to interact primarily with the underphosphorylated form of Rb. Experiments with truncated glutathione S-transferase-Rb fusion proteins and nuclear extracts of Rh1 rhabdomyosarcoma cells indicated that topo IIalpha binds avidly to the A/B pocket domain of Rb, which contains the intact spacer amino acid sequence. To determine whether this interaction has functional consequences in vivo, we expressed wild-type and mutant Rb in human cervical carcinoma cells lacking functional Rb. Wild-type, but not mutant, Rb inhibited topo II activity in nuclear extracts of these transfected cells. Moreover, purified wild-type Rb inhibited the activity of purified human topo IIalpha, indicating a direct interaction between these two proteins. We conclude that topo IIalpha associates physically with Rb in interactions that appear to have functional significance.


Subject(s)
DNA Topoisomerases, Type II , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Isoenzymes/chemistry , Isoenzymes/metabolism , Retinoblastoma Protein/chemistry , Retinoblastoma Protein/metabolism , Animals , Antigens, Neoplasm , Binding Sites , Child , DNA Topoisomerases, Type II/isolation & purification , DNA-Binding Proteins , Female , Glutathione Transferase , Humans , Isoenzymes/isolation & purification , Kinetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Retinoblastoma Protein/isolation & purification , Rhabdomyosarcoma , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured , Uterine Cervical Neoplasms
14.
Mol Pharmacol ; 55(3): 548-56, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10051540

ABSTRACT

Merbarone (5-[N-phenyl carboxamido]-2-thiobarbituric acid) is an anticancer drug that inhibits the catalytic activity of DNA topoisomerase II (topo II) without damaging DNA or stabilizing DNA-topo II cleavable complexes. Although the cytotoxicity of the complex-stabilizing DNA-topo II inhibitors such as VP-16 (etoposide) has been partially elucidated, the cytotoxicity of merbarone is poorly understood. Here, we report that merbarone induces programmed cell death or apoptosis in human leukemic CEM cells, characterized by internucleosomal DNA cleavage and nuclear condensation. Treatment of CEM cells with apoptosis-inducing concentrations of merbarone caused activation of c-Jun NH2-terminal kinase/stress-activated protein kinase, c-jun gene induction, activation of caspase-3/CPP32-like protease but not caspase-1, and the proteolytic cleavage of poly(ADP-ribose) polymerase. Treatment of CEM cells with a potent inhibitor of caspases, Z-Asp-2. 6-dichlorobenzoyloxymethyl-ketone, inhibited merbarone-induced caspase-3/CPP32-like activity and apoptosis in a dose-dependent manner. These results indicate that the catalytic inhibition of topo II by merbarone leads to apoptotic cell death through a caspase-3-like protease-dependent mechanism. These results further suggest that c-Jun and c-Jun NH2-terminal kinase/stress-activated protein kinase signaling may be involved in the cytotoxicity of merbarone.


Subject(s)
Apoptosis , Cysteine Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinases , Thiobarbiturates/pharmacology , Topoisomerase II Inhibitors , Asparagine/analogs & derivatives , Asparagine/pharmacology , Caenorhabditis elegans Proteins , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Enzyme Activation , Gene Expression/drug effects , Genes, jun/physiology , Humans , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 9 , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinases/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
15.
Mol Pharmacol ; 55(2): 216-22, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9927611

ABSTRACT

Many anticancer agents exert their cytotoxicity through DNA damage and induction of apoptosis. Fas ligand (FasL), a key component of T lymphocytes, has been shown to be induced by some of those agents. To address what is an early signal for this induction, we constructed a FasL promoter-luciferase reporter gene to investigate effects of DNA topoisomerase (Topo) II inhibitors on FasL promoter activity. Transient transfection assays in HeLa and other tumor cell lines demonstrated that induction of FasL promoter activity in response to Topo II inhibitors such as VM-26 mimicked endogenous FasL expression under the same conditions. The ability of these agents to induce FasL expression correlated with their ability to cause DNA damage. For instance, complex-stabilizing Topo II inhibitors such as etoposide, teniposide, and doxorubicin, which cause DNA damage, strongly induce FasL expression; by contrast, non-DNA-damaging catalytic Topo II inhibitors such as ICRF-187 and merbarone do not do this. In support of the notion that DNA damage triggers FasL induction, we found that DNA-damaging irradiation also induced FasL promoter activity in a dose-dependent manner. Finally, the catalytic Topo II inhibitor ICRF-187 suppressed VM-26-induced-FasL expression. This suppression correlated with the ability of this drug to inhibit VM-26-induced DNA strand breaks. Together, our results suggest that DNA damage in response to agents such as etoposide and teniposide might serve as an early signal to induce FasL expression.


Subject(s)
DNA Damage/physiology , Membrane Glycoproteins/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , CHO Cells , Cricetinae , DNA/chemistry , DNA/drug effects , DNA Damage/genetics , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Fas Ligand Protein , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Jurkat Cells , Nucleic Acid Synthesis Inhibitors/pharmacology , Promoter Regions, Genetic/genetics , Razoxane/pharmacology , Recombinant Fusion Proteins/genetics , Signal Transduction/drug effects , Teniposide/pharmacology , Thiobarbiturates/pharmacology , Topoisomerase II Inhibitors , Tumor Cells, Cultured , Up-Regulation
16.
Biotechniques ; 25(6): 1052-7, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9863061

ABSTRACT

DNA topoisomerase (topo) II alpha is a major target for many anticancer agents. However, progress towards understanding how these agents interact with this enzyme in human cells and how resistance to these agents arises is greatly impeded by difficulties in expressing this gene. Here, we report on achieving a high level of expression of a full-length human topo II alpha gene in human cells. We started with the topo II alpha cDNA driven by a strong cytomegalovirus promoter and transiently transfected HeLa cells. Although topo II alpha mRNA was consistently detected in transfected cells, no exogenous topo II alpha protein was detected. By contrast, when the same cDNA was fused to an enhanced green fluorescent protein (EGFP), we detected a high level of expression at both mRNA and protein levels. The exogenous topo II alpha was localized to cell nuclei as expected, indicating that the fusion protein is properly folded. Furthermore, overexpression of the EGFP-topo II alpha fusion protein increased the sensitivity of the transfected cells to teniposide, suggesting that it functions as the endogenous counterpart. Thus, in addition to being used as a gene tag, the GFP fusion approach may be generally applicable for expressing genes, such as topo II alpha, that are difficult to express by conventional methods.


Subject(s)
DNA Topoisomerases, Type II/genetics , Gene Expression Regulation, Viral , Indicators and Reagents/metabolism , Luminescent Proteins/genetics , Recombinant Fusion Proteins/genetics , Base Sequence , Cell Nucleus/chemistry , DNA Topoisomerases, Type II/analysis , Enzyme Inhibitors/pharmacology , Genes, Reporter , Green Fluorescent Proteins , HeLa Cells , Humans , Immunoblotting , Microscopy, Fluorescence , Molecular Sequence Data , RNA, Messenger/analysis , Recombinant Fusion Proteins/analysis , Teniposide/pharmacology , Topoisomerase II Inhibitors , Transfection
17.
Cancer Res ; 58(24): 5762-9, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9865734

ABSTRACT

Multidrug resistance is a major obstacle to the success of cancer chemotherapy. The multidrug resistance-associated protein (MRP) has been shown to confer multidrug resistance. To study MRP gene expression at the transcriptional level, we have fused the MRP gene promoter with the luciferase reporter gene and studied its regulation. Cotransfection of MRP promoter constructs with p53 expression plasmids in p53-null human H1299 and mouse (10)1 cells demonstrated that the wild-type (wt) p53 markedly suppressed MRP promoter activity, whereas mutant p53 had little inhibitory effect. Transfections using 5' deletion mutant constructs of the MRP promoter showed that inhibition of the promoter activity by wt p53 mainly resided in the region from -91 to +103 bp, where several Sp1 transcription factor binding sites are localized. Cotransfection of the MRP promoter into Drosophila SL2 cells with an Sp1 expression vector increased the promoter activity in a dose-related manner up to approximately 200-fold. The stimulation of MRP promoter activity by Sp1 was attenuated by the cotransfection of a wt p53-expression plasmid. Furthermore, we have determined that endogenous MRP mRNA levels were down-regulated by restoration of wt p53-expression in a human lung cancer cell line. The relevance of MRP regulation in drug resistance was studied in a drug-resistant cell line, CEM/VM-1-5, that is approximately 140-fold more resistant to the epipodophyllotoxin, teniposide (VM-26), than the parental CEM cells. CEM/VM-1-5 cells express a much higher amount of MRP mRNA and protein than CEM cells, indicating that the resistant phenotype is at least partly due to increased MRP production. Transient transfection of the promoter constructs revealed that CEM/VM-1-5 cells had higher (7-fold) MRP promoter activity than CEM cells. Cotransfection of a wt p53-expression plasmid caused a reduction of MRP promoter activity in both CEM and CEM/VM-1-5 cells, but the inhibition was more than double in CEM/VM-1-5 cells compared with CEM cells. Our results demonstrated that wt p53 acts as a negative regulator of MRP gene transcription, at least in part by diminishing the effect of a powerful transcription activator Sp1. Therefore, a loss of wt p53 function and/or an increase in Sp1 activity in tumor cells could contribute to an up-regulation of the MRP gene.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, MDR , Genes, p53 , Animals , Down-Regulation , Humans , Mice , Mutation , Promoter Regions, Genetic , Sp1 Transcription Factor/physiology , Transcription, Genetic , Transfection , Tumor Cells, Cultured
18.
Biochem Pharmacol ; 56(6): 719-27, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9751076

ABSTRACT

P-Glycoprotein (Pgp) is responsible for the energy-dependent efflux of many natural product oncolytics. Overexpression of Pgp may result in multidrug resistance (MDR). Modulators can block Pgp efflux and sensitize multidrug resistant cells to these oncolytics. To study the interaction of modulators with Pgp, Pgp-ATPase activity was examined, using plasma membranes isolated from the multidrug-resistant cell line CEM/VLB100. A survey of modulators indicated that verapamil, trifluoperazine, and nicardipine stimulated ATPase activity by 1.3- to 1.8-fold, whereas two others, trimethoxybenzoylyohimbine (TMBY) and vindoline, had no effect. Further evaluation showed that TMBY completely blocked the stimulation by verapamil of ATPase activity by competitive inhibition, with a Ki of 2.1 microM. When the effects of these two modulators on the formation of the enzyme-nucleotide complex important in the catalytic cycle were examined, verapamil increased the amount of vanadate-trapped 8-azido-[alpha-32P]ATP bound to Pgp by two-fold, whereas TMBY had no effect. Moreover, TMBY blocked the verapamil stimulation of vanadate-8-azido-[alpha-32P]ATP. Together, these data indicate that verapamil and TMBY bind to Pgp at a common site or overlapping sites, but only verapamil results in enhanced Pgp-ATP hydrolysis and formation of the vanadate-nucleotide-enzyme complex.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphatases/drug effects , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/analogs & derivatives , Azides/metabolism , Vanadates/pharmacology , Adenosine Triphosphate/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Binding Sites , Calcium Channel Blockers/pharmacology , Cell Membrane/enzymology , Cell Membrane/metabolism , Drug Interactions , Drug Resistance, Multiple , Humans , Hydrolysis , Leukemia, Lymphoid/metabolism , Phosphorus Radioisotopes , Protein Conformation , Stimulation, Chemical , Tumor Cells, Cultured , Verapamil/pharmacology , Vinblastine/pharmacology , Yohimbine/analogs & derivatives , Yohimbine/pharmacology
19.
Anticancer Drugs ; 9(5): 437-48, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9660542

ABSTRACT

The sensitivity of human tumor and rat prostate tumor cells to a series of naphthoquinones, including tricyclic compounds of the beta-lapachone and dunnione families as well as 4-alkoxy-1,2-naphthoquinones, was evaluated. To better understand the mechanism of cytotoxicity of 1,2-naphthoquinones, the roles of various resistance mechanisms including P-glycoprotein, multidrug resistant associated protein, glutathione (GSH) and related enzymes, altered topoisomerase activity, and overexpression of genes that control apoptosis (bcl-2 and bc-xL) were studied. MCF7 cells were most sensitive to the naphthoquinones with IC50 values ranging from 1.1 to 10.8 microM, as compared to 2.5 to >32 microM for HT29 human colon, A549 human lung, CEM leukemia and AT3.1 rat prostate cancer cells. MCF7 ADR cells, selected for resistance to adriamycin (ADR), displayed cross-resistance to the tricyclic 1,2-naphthoquinones. Drug efflux via a P-glycoprotein mechanism was ruled out as a mechanism of resistance to 1,2-naphthoquinones, since KB-V1 cells expressing high levels of P-glycoprotein and the KB-3.1 parent line were equally sensitive to these compounds. Any resistance of the tricyclic naphthoquinones noted in ADR-resistant cells appeared to relate to the GSH redox cycle and could be circumvented by exposure to buthionine sulfoximine or by changing the structure from a tricyclic derivative to a 4-alkoxy-1,2-naphthoquinone. The 1,2-naphthoquinones were found to be cytotoxic against CEM/VM-1 and CEM/M70-B1 cells that were selected for resistance to teniposide or merbarone, respectively. In addition, cells overexpressing bcl-2 or bcl-xL proteins were as sensitive to 1,2-naphthoquinones as were control cells. Because of their effectiveness in drug-resistant cells, these agents appear to hold promise as effective chemotherapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Naphthoquinones/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/physiology , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Division/drug effects , Doxorubicin/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Glutathione/drug effects , Glutathione/physiology , Humans , Multidrug Resistance-Associated Proteins , Naphthoquinones/chemistry , Naphthoquinones/toxicity , Oxidation-Reduction/drug effects , Peroxidases/drug effects , Peroxidases/physiology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/physiology , Rats , Topoisomerase II Inhibitors , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , bcl-X Protein
20.
Cancer Res ; 57(22): 5004-8, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9371492

ABSTRACT

DNA topoisomerase II (topo II) is a target for many clinically useful anticancer drugs. However, a major concern in the use of these drugs is the development of resistance, often manifested by reduced drug accumulation or reduced topo IIalpha activity, due to mutant enzyme or the enzyme's decreased expression. To date, little is known of how the topo IIalpha is down-regulated in the resistant cells. In this study, using CEM cells selected for resistance to merbarone, we found that topo IIalpha RNA levels were reduced, compared to the parental cells, and this corresponded to reduced protein levels, whereas there was no significant difference in the RNA stability among these cell lines. Furthermore, we detected a lower level of topo IIalpha promoter activity in these resistant cells compared to the drug-sensitive parents. Thus, the down-regulation of topo IIalpha appeared to occur at the transcriptional level. Nucleotide sequencing of the topo IIalpha promoter regions up to -1200 bp revealed no mutations, suggesting that some trans-acting factors are possibly involved in this down-regulation of topo Ilalpha. In this context, we found by Northern blot analysis that the transcription factor, sp3, was reduced in the drug-resistant cell lines compared to the parental cells. Furthermore, cotransfection experiments revealed that Sp3 induced topo IIalpha promoter activity in a dose-dependent manner in drug-sensitive CEM cells, but its induction of topo IIalpha promoter activity was attenuated in the resistant B12 cells. Our results suggest that down-regulation of Sp3 might contribute to the reduced expression of topo IIalpha in certain drug-resistant tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Thiobarbiturates/pharmacology , Transcription Factors/metabolism , Drug Resistance, Neoplasm , Half-Life , Humans , RNA, Messenger/metabolism , Sp3 Transcription Factor , Topoisomerase II Inhibitors , Transcriptional Activation , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...