Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Chemotherapy ; 56(3): 223-33, 2010.
Article in English | MEDLINE | ID: mdl-20551639

ABSTRACT

BACKGROUND: The purpose of this study was: (1) to document the critical requirement of cystine for growth of human tumor cells in vitro, and (2) to determine the effect of the anticancer agent irinotecan on the cystine transporter x(c)(-) in head and neck FaDu xenografts. METHODS: Cell growth was measured by sulforhodamine B assay. xCT protein, glutathione (GSH) and DNA damage were determined using Western blot, spectrophotometry, and immunohistochemistry, respectively. RESULTS: Depletion of cystine from the medium inhibited tumor cell growth. Treatment of FaDu tumor with a therapeutic dose of irinotecan resulted in depression of xCT protein levels, leading to tumor growth retardation and downregulation of GSH with increased reactive oxygen species (ROS). The accumulation of ROS correlated with increased DNA damage as evidenced by increased H2AX. CONCLUSION: Depression of xCT protein by irinotecan resulted in downregulation of GSH and increase in ROS, which could be the other possible mechanisms of DNA damage by irinotecan.


Subject(s)
Amino Acid Transport Systems/metabolism , Camptothecin/analogs & derivatives , Cystine/physiology , Down-Regulation/physiology , Head and Neck Neoplasms/metabolism , Xenograft Model Antitumor Assays , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/biosynthesis , Animals , Camptothecin/pharmacology , Cell Line, Tumor , Cystine/antagonists & inhibitors , Cystine/metabolism , Down-Regulation/drug effects , Female , Humans , Irinotecan , Mice , Mice, Nude , Xenograft Model Antitumor Assays/methods
2.
Mol Pharmacol ; 66(1): 153-60, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15213307

ABSTRACT

Methylselenocysteine (MSC) is an organic selenium compound in preventative clinical trials involving prostate, lung, and colon carcinoma. We found that methioninase-activated MSC potentiates 7-ethyl-10-hydroxycamptothecin (SN-38)-induced cell lethality in vitro in the p53-defective human head and neck carcinoma A253 cells. Activated MSC increases chk2 phosphorylation at threonine-68 induced by SN-38, with no significant effect on chk1 phosphorylation. Cell cycle arrest induced by SN-38, however, was not abrogated or potentiated by MSC. These results suggest that the enhanced cellular lethality of SN-38 by MSC was not associated with cell cycle regulation pathways. Because chk2, in addition to its role in cell cycle arrest, can induce apoptosis by phosphorylation/activation, we examined whether increased chk2 phosphorylation could induce preapoptotic DNA fragmentation. DNA damage analysis showed that megabase DNA fragmentation is decreased, accompanied by the increased 30 to 300 kilobase pairs of DNA fragmentation after exposure to SN-38 with MSC, compared with SN-38 alone. No significant changes in the amount of DNA fragments were observed in cells treated with SN-38 or MSC alone. Moreover, proteolytic destruction of DNA replication-associated proteins cdc6, MCM2, and cdc25A may induce a DNA damage checkpoint response. The observed down-regulation of DNA replication proteins cdc6, MCM2, and cdc25A after exposure to SN-38 with MSC further indicates a relationship between drug response and DNA damage. Exposure to SN-38 with MSC resulted in a significant increase of poly(ADP-ribose) polymerasecleavage and caspase 3 activation. All together, the data support the hypothesis that enhanced lethality of this combination is associated with increased chk2 phosphorylation at Thr68 and down-regulation of specific DNA replication-associated proteins, which result in poly(ADP-ribose) polymerase cleavage, caspase 3 activation, and the induction of 30 to 300 kilobase pairs of DNA fragmentation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cysteine/analogs & derivatives , Cysteine/pharmacology , Organoselenium Compounds/pharmacology , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins , Carbon-Sulfur Lyases/metabolism , Caspase 3 , Caspases/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cell Survival/drug effects , Checkpoint Kinase 1 , Checkpoint Kinase 2 , DNA Fragmentation/drug effects , Drug Interactions , Enzyme Activation , Gene Expression/drug effects , Humans , Irinotecan , Minichromosome Maintenance Complex Component 2 , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinases/metabolism , Selenocysteine/analogs & derivatives , cdc25 Phosphatases/metabolism
3.
Clin Cancer Res ; 10(3): 1121-9, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14871992

ABSTRACT

PURPOSE: Although the combination of irinotecan and 5-Fluorouracil is clinically active, it is associated with significant toxicity and resistance. Studies were carried out to define the optimal dosage, sequence, and timing for the combination in mice bearing xenografted human tumors. EXPERIMENTAL DESIGN: The maximum tolerated dose of irinotecan and 5-Fluorouracil in combination was determined in nude mice. Therapeutic efficacy against established human colon carcinoma xenografts, HCT-8 and HT-29, and human head and neck squamous cell carcinoma xenografts, FaDu and A253, was determined using the rugs individually, simultaneously, and in sequence with various intervals in between. Treatments were i.v. weekly x 4. Immunohistochemical and reverse transcription-PCR measurements of relevant drug-metabolizing enzymes, apoptosis-related proteins, cell cycle distribution, cyclin A, and S phase fraction expression were carried out and compared with the therapeutic outcome. RESULTS: The maximum tolerated dose of irinotecan resulted in cure rates of 30% or less in all xenografts. No cures were achieved with FUra alone. Concurrent administration of irinotecan and FUra, or of FUra 24 h before irinotecan, resulted in cure rates of <20%, except for FaDu (60%). Administration of irinotecan 24 h before FUra resulted in the highest cure rates, 80% in HCT-8, 0% in HT-29, 100% in FaDu, and 10% in A253. CONCLUSIONS: The optimal therapeutic synergy was achieved when irinotecan was administered 24 h before 5-Flurouracil. Sensitivity to this combination was associated with poor differentiation status, higher cyclin A index, recruitment of cells into S phase, and induction of Bax expression and apoptosis.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Carcinoma, Squamous Cell/pathology , Fluorouracil/therapeutic use , Head and Neck Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Blotting, Western , Carcinoma, Squamous Cell/drug therapy , Cell Cycle , Cell Line, Tumor , Female , Head and Neck Neoplasms/drug therapy , Humans , Immunohistochemistry , Irinotecan , Kinetics , Maximum Tolerated Dose , Mice , Mice, Nude , Neoplasm Transplantation , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
4.
Int J Oncol ; 21(5): 1059-66, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12370755

ABSTRACT

Human head and neck squamous carcinoma cell lines, A253 and FaDu, were utilized to identify mediators associated with response to topoisomerase I poison, SN-38, a metabolite of irinotecan. The drug sensitivity of FaDu cells to SN-38 was significantly higher than that of the A253 cells. In A253 cells, G2/M arrest following drug treatment (0.35 microM SN-38, 2-h exposure) was accompanied by DNA fragmentation in the 50-300 kb range, but FaDu cells accumulated in S-phase concurrently with induction of smaller DNA fragmentation in the 4-80 kb range. Because the critical regulatory step in activating cdc2 during progression into mitosis appears to be dephosphorylation of Tyrosine 15 (Tyr15), we examined the Tyr15 phosphorylation status of cdc2 in both cell lines. Slightly increased levels of cdc2 phosphorylation was observed in the A253 cells, while reduced levels of cdc2 phosphorylation was noted in the FaDu cells, corresponding to the abrogation of the G2-phase arrest. Increased chk1 phosphorylation at Ser345 induced by SN-38 was accompanied by the observed G2 phase arrest in the A253 cell line, while significant downregulation of chk1 and cdc25C phosphorylation, which resulted in the abrogation of G2/M checkpoint arrest, was noted in FaDu cells at this timepoint. These results suggest that alterations of chk1 signaling are associated with the response to topoisomerase I poison SN-38. Furthermore, A253 cells possess higher levels of endogenous hMLH1, compared to FaDu cells. A deficiency in G2 arrest was observed in FaDu cells, suggesting endogenous hMLH1 protein expression is associated with the abrogation of G2/M arrest, subsequently with the response to topoisomerase I poison SN-38.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Head and Neck Neoplasms/drug therapy , Protein Kinases/metabolism , Serine/metabolism , Topoisomerase I Inhibitors , 14-3-3 Proteins , Adaptor Proteins, Signal Transducing , Carrier Proteins , Cell Cycle Proteins/analysis , Checkpoint Kinase 1 , DNA Damage , DNA Fragmentation/drug effects , G2 Phase/drug effects , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Irinotecan , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/analysis , Nuclear Proteins , Phosphorylation , Proto-Oncogene Proteins/analysis , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/analysis , cdc25 Phosphatases/analysis
5.
Biochem Biophys Res Commun ; 295(2): 435-44, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-12150968

ABSTRACT

A novel karenitecin, BNP1350, is a topoisomerase I-targeting anticancer agent with significant antitumor activity in vitro and in vivo. A BNP1350-resistant human head and neck carcinoma A253 cell line, denoted A253/BNPR, was developed. The A253/BNPR cell line was approximately 9-fold resistant to BNP1350 and 4-fold cross-resistant to another topoisomerase I inhibitor SN-38, the active metabolite of irinotecan. After drug treatment with equimolar concentrations of BNP1350 (0.7 microM) for 2h, activation of the DNA double-strand break repair protein complexes was similar in the two cell lines, suggesting that DNA dsb repair is not attributable to resistance to BNP1350 in the A253/BNPR cells. Cell cycle analysis indicates that the A253 cell line accumulated primarily in S phase, but G(2) phase accumulation was observed in the A253/BNPR cell line at 48 h after drug removal. Elevated chk1 phosphorylation at Ser(345) following DNA damage induced by BNP1350 was accompanied by G(2) accumulation in the A253/BNPR cell line, while exposure to equimolar concentrations of BNP1350 (0.7 microM) induced S-phase arrest and no increased phosphorylation of chk1 at Ser(345) in the A253 cell line. Under the same conditions, increased chk1 activity was observed in the A253/BNPR cell line, but not in the A253 cell line. Moreover, stimulated binding of 14-3-3 proteins to chk1 was observed in BNP1350-treated A253/BNPR cells. To confirm relationship between chk1 expression/phosphorylation and drug resistance to topo I poisons, we examined the effects of chk1 or chk2 antisense oligonucleotides on the cellular growth inhibition. Chk1 antisense oligonucleotide can sensitize the A253/BNPR cells to killing by topo I inhibitor BNP1350, but no significant sensitization of BNP1350-induced growth inhibition was observed in the drug-sensitive cell line. Chk2 antisense oligonucleotide has only a small sensitization effect on BNP1350-induced growth inhibition in both cell lines. The data indicate that the chk1 signaling pathways that mediate cell cycle checkpoint are associated with cellular resistance to BNP1350 in the A253/BNPR cell line.


Subject(s)
Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , G2 Phase , Mitosis , Protein Kinases/metabolism , Signal Transduction , Topoisomerase I Inhibitors , 14-3-3 Proteins , Cell Division/drug effects , Checkpoint Kinase 1 , DNA Damage , DNA Repair , Humans , Oligonucleotides, Antisense/metabolism , Phosphorylation , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/metabolism
6.
Mol Pharmacol ; 61(4): 742-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11901212

ABSTRACT

Camptothecins demonstrate a broad spectrum of antitumor activity. Although they are known to trap DNA topoisomerase I on DNA, form cleavable complexes, and generate DNA breaks upon collision with DNA or RNA polymerases, the precise mechanisms predictive for antitumor activity remain to be identified. Recent studies using panels of colorectal and breast cancer cell lines indicate that events downstream of cleavable complexes are more relevant. In this study, we chose SN-38, an active metabolite of irinotecan, to characterize DNA double strand breaks and repair mechanisms induced by this type of drugs using a human head and neck squamous cell carcinoma cell line A253. The results showed that 2-h exposure of cells to an IC(50) concentration of SN-38 induces biphasic DNA double-strand break (DSBs): an immediate phase, which was greatly reduced within 8 h, and a lagging phase, culminating 24 h after drug removal. Three DNA double-strand break repair protein complexes were activated: DNA-dependent protein kinase (DNA-PK), NBS1-MRE11-RAD50, and BRCA1. Aphidicolin, a DNA polymerase inhibitor, abolished both phase I DSBs and the activation of repair protein complexes, suggesting that they resulted from the collision between the cleavable complex and DNA polymerase of S-phase cells. This is in contrast to ionizing radiation-induced activation of DNA-PK and NBS1-MRE11-RAD50 complexes that occur predominantly among non-S-phase cells. The trigger for phase II DSBs cannot be abolished by aphidicolin. The data also indicate that DNA fragments in the size of 50 to 200 kilobases were detected in the lagging phase. This suggests that the late DNA DSBs were associated with apoptotic cell death.


Subject(s)
Camptothecin/pharmacology , DNA Ligases/metabolism , DNA Repair/physiology , DNA Topoisomerases, Type I/metabolism , Enzyme Inhibitors/pharmacology , Aphidicolin/pharmacology , Apoptosis , Camptothecin/analogs & derivatives , Cell Division/drug effects , DNA Damage , DNA Fragmentation/drug effects , Drug Interactions , Electrophoresis, Gel, Pulsed-Field , Enzyme Activation , Humans , Irinotecan , Nucleic Acid Synthesis Inhibitors , Phosphorylation , Topoisomerase I Inhibitors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...