Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Publication year range
1.
Mutat Res ; 755(2): 163-6, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23817106

ABSTRACT

Conflicting results have been published regarding the induction of genotoxic effects by exposure to radiofrequency electromagnetic fields (RF-EMF). Various results indicating a genotoxic potential of RF-EMF were reported by the collaborative EU-funded REFLEX (Risk Evaluation of Potential Environmental Hazards From Low Energy Electromagnetic Field Exposure Using Sensitive in vitro Methods) project. There has been a long-lasting scientific debate about the reliability of the reported results and an attempt to reproduce parts of the results obtained with human fibroblasts failed. Another part of the REFLEX study was performed in Berlin with the human lymphoblastoid cell line HL-60; genotoxic effects of RF-EMF were measured by means of the comet assay and the micronucleus test. The plausibility and reliability of these results were also questioned. In order to contribute to a clarification of the biological significance of the reported findings, a repeat study was performed, involving scientists of the original study. Comet-assay experiments and micronucleus tests were performed under the same experimental conditions that had led to genotoxic effects in the REFLEX study. Here we report that the attempts to reproduce the induction of genotoxic effects by RF-EMF in HL-60 cells failed. No genotoxic effects of RF-EMF were measured in the repeat experiments. We could not find an explanation for the conflicting results. However, the negative repeat experiments suggest that the biological significance of genotoxic effects of RF-EMF reported by the REFLEX study should be re-assessed.


Subject(s)
HL-60 Cells/radiation effects , Radio Waves/adverse effects , Comet Assay , Humans , Micronucleus Tests , Multicenter Studies as Topic , Reproducibility of Results , Research Design
2.
Bioelectromagnetics ; 30(3): 213-21, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19025782

ABSTRACT

This study investigated whether glutathione depletion affected the sensitivity of HL-60 cells to static magnetic fields. The effect of Diethylmaleate (DEM) on static magnetic field induced changes in cytosolic free calcium concentration ([Ca(2+)](c)) was examined. Cells were loaded with a fluorescent dye and exposed to a uniform static magnetic field at a strength of 0 mT (sham) or 100 mT. [Ca(2+)](c) was monitored during field and sham exposure using a ratiometric fluorescence spectroscopy system. Cells were activated by the addition of ATP. Metrics extracted from the [Ca(2+)](c) time series included: average [Ca(2+)](c) during the Pre-Field and Field Conditions, peak [Ca(2+)](c) following ATP activation and the full width at half maximum (FWHM) of the peak ATP response. Comparison of each calcium metric between the sham and 100 mT experiments revealed the following results: average [Ca(2+)](c) measured during the Field condition was 53 +/- 2 nM and 58 +/- 2 nM for sham and 100 mT groups, respectively. Average FWHM was 51 +/- 3 s and 54 +/- 3 s for sham and 100 mT groups, respectively. An effect of experimental order on the peak [Ca(2+)](c) response to ATP in sham/sham experiments complicated the statistical analysis and did not allow pooling of the first and second order experiments. No statistically significant difference between the sham and 100 mT groups was observed for any of the calcium metrics. These data suggested that manipulation of free radical buffering capacity in HL-60 cells did not affect the sensitivity of the cells to a 100 mT static magnetic field.


Subject(s)
Adenosine Triphosphate/pharmacology , Calcium/metabolism , Cytosol/metabolism , Electromagnetic Fields , Free Radicals/metabolism , HL-60 Cells/radiation effects , Humans , Maleates/pharmacology
3.
Bioelectromagnetics ; 30(4): 322-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19204977

ABSTRACT

Calcium is an important molecule in a number of biological systems. Often these systems are signal transduction cascades involving molecules such as ATP. ATP activates second messengers which can interact with ion channels on the endoplasmic/sarcoplasmic reticulum resulting in the emptying of the intracellular calcium stores and an increase in cytosolic free calcium concentration ([Ca2+]c). Changes in [Ca2+]c can be influenced by external factors such as a static magnetic field (SMF). One hypothesis suggests that a SMF affects the cells through the radical pair mechanism. By reducing the number of antioxidant molecules like glutathione (GSH), the proportion of free radicals in the cells is increased and may lead to a greater probability of a biological response to a SMF. The purpose of this study was to determine if the [Ca2+]c response to ATP was affected by depletion of GSH by diethylmaleate (DEM) and the absence or presence of a 100 mT homogeneous SMF. Undifferentiated HL-60 cells were loaded with fura-2 AM. [Ca2+]c was measured in real time using a ratiometric fluorescence spectroscopy system. Various (DEM) ranging from 1 to 15 mM were added to deplete GSH. Cells were either exposed to sham or magnetic field (100 mT) for 13 min (780 s) and challenged with 1 microM ATP. The data show that [Ca2+]c was elevated following treatment with DEM with greater [Ca2+]c at higher [DEM]. The [Ca2+]c response to ATP was decreased as the DEM concentration increased. However, there was no effect of a 100 mT SMF on the average [Ca2+]c peak following ATP activation or the full width at half maximum (FWHM) of the [Ca2+]c response and recovery after ATP activation.


Subject(s)
Adenosine Triphosphate/pharmacology , Calcium/metabolism , Glutathione/metabolism , Magnetics , Calcium Signaling/drug effects , Calcium Signaling/radiation effects , Cytosol/metabolism , HL-60 Cells/radiation effects , Humans , Maleates/pharmacology
4.
Leuk Res ; 32(12): 1866-77, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18534676

ABSTRACT

The nuclear arrangement of promyelocytic leukaemia nuclear bodies (PML NBs) was studied in vitro after the cell treatment by clinically used agents such as all-trans retinoic acid (RA) in human leukaemia and cytostatics or gamma radiation in multiple myeloma cells. In addition, the influence of phorbol ester (PMA) on PML NBs formation was analyzed. A reduced number of PML bodies, which led to relocation of PML NBs closer to the nuclear interior, mostly accompanied RA- and PMA-induced differentiation. Centrally located PML NBs were associated with transcriptional protein RNAP II and SC35 regions, which support importance of PML NBs in RNA processing that mostly proceeds within the nuclear interior. Conversely, the quantity of PML NBs was increased after cytostatic treatment, which caused re-distribution of PML NBs closer to the nuclear envelope. Here we showed correlations between the number of PML NBs and average Centre-to-PML distances. Moreover, a number of cells in S phase, especially during differentiation, influenced number of PML NBs. Studying the proteins involved in PML compartment, such as c-MYC, cell-type specific association of c-MYC and the PML NBs was observed in selected leukaemic cells undergoing differentiation, which was accompanied by c-MYC down-regulation.


Subject(s)
Cell Cycle/physiology , Cell Nucleus/pathology , Intranuclear Inclusion Bodies/pathology , Leukemia, Promyelocytic, Acute/pathology , Multiple Myeloma/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Flow Cytometry , Gamma Rays , HL-60 Cells/drug effects , HL-60 Cells/pathology , HL-60 Cells/radiation effects , Humans , Intranuclear Inclusion Bodies/drug effects , Intranuclear Inclusion Bodies/radiation effects , K562 Cells/drug effects , K562 Cells/pathology , K562 Cells/radiation effects , Melphalan/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology , U937 Cells/pathology
5.
Mol Cell Biol ; 15(11): 5849-57, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7565737

ABSTRACT

The p53 tumor suppressor gene is thought to be required for the induction of programmed cell death (apoptosis) initiated by DNA damage. We show here, however, that the human promyelocytic leukemia cell line HL-60, which is known to be deficient in p53 because of large deletions in the p53 gene, can be induced to undergo apoptosis following X-irradiation. We demonstrate that the decision to undergo apoptosis in this cell line appears to be made at a G2 checkpoint. In addition, we characterize an HL-60 variant, HCW-2, which is radioresistant. HCW-2 cells display DNA damage induction and repair capabilities identical to those of the parental HL-60 cell line. Thus, the difference between the two cell lines appears to be that X-irradiation induces apoptosis in HL-60, but not in HCW-2, cells. Paradoxically, HCW-2 cells display high levels of expression of bax, which enhances apoptosis, and no longer express bcl-2, which blocks apoptosis. HCW-2 cells' resistance to apoptosis may be due to the acquisition of expression of bcl-XL, a bcl-2-related inhibitor of apoptosis. In summary, apoptosis can be induced in X-irradiated HL-60 cells by a p53-independent mechanism at a G2 checkpoint, despite the presence of endogenous bcl-2. The resistance shown by HCW-2 cells suggests that bcl-XL can block this process.


Subject(s)
Apoptosis/radiation effects , G2 Phase , HL-60 Cells/radiation effects , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p53/physiology , DNA Damage/radiation effects , DNA Repair/radiation effects , Dose-Response Relationship, Radiation , Gene Expression , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogenes , RNA, Messenger/genetics , X-Rays , bcl-2-Associated X Protein
6.
Anticancer Res ; 26(5A): 3373-80, 2006.
Article in English | MEDLINE | ID: mdl-17094455

ABSTRACT

Three antitumor antibiotics, mitomycin C, bleomycin sulfate and peplomycin sulfate, were compared for their tumor-specific cytotoxicity, using human oral squamous cell lines (HSC-2, HSC-3, HSC-4, Ca9-22 and NA), human promyelocytic leukemic cell line HL-60 and human normal oral cell types (gingival fibroblast HGF, pulp cell HPC and periodontal ligament fibroblast HPLF). Among these three compounds, mitomycin C showed the highest tumor-specificity, due to its higher cytotoxic activity against human oral tumor cell lines than bleomycin and peplomycin. However, there was considerable variation of drug sensitivity among the six tumor cell lines. Mitomycin C induced internucleosomal DNA fragmentation and caspase-3, -8 and -9 activation in HL-60 cells only after 24 h. On the other hand, mitomycin C induced no clear-cut DNA fragmentation in HCS-2 cells, although it activated caspase-3, -8 and -9 to a slightly higher extent. Western blot analysis demonstrated that mitomycin C did not induce any apparent change in the intracellular concentration of anti-apoptotic protein (Bcl-2) and pro-apoptotic proteins (Bax, Bad). Electron microscopy of mitomycin C-treated HL-60 cells showed intact mitochondria (as regards to integrity and size) and cell surface microvilli, without production of an apoptotic body or autophagosome, at an early stage after treatment. The present study suggests the incomplete induction of apoptosis or the induction of another type of cell death by mitomycin C treatment.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Carcinoma, Squamous Cell/pathology , Mitomycin/pharmacology , Mouth Neoplasms/pathology , Peplomycin/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Caspases/metabolism , Cell Death/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Fibroblasts/drug effects , HL-60 Cells/drug effects , HL-60 Cells/radiation effects , HL-60 Cells/ultrastructure , Humans , Mouth Neoplasms/metabolism , Ultraviolet Rays
7.
Cancer Res ; 58(3): 421-5, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9458083

ABSTRACT

There is strong clinical and epidemiological evidence that ionizing radiation can cause leukemia by inducing DNA damage. This crucial initiation event is believed to be the result of random DNA breakage and misrepair, whereas the subsequent steps, promotion and progression, must rely on mechanisms of selective pressure to provide the expanding leukemic population with its proliferative/renewal advantage. To investigate the susceptibility of human cells to external agents at the genetic recombination stage of leukemogenesis, we subjected two hematopoietic cell lines, KG1 and HL60, to high doses of gamma-irradiation. The irradiation induced the formation of fusion genes characteristic of leukemia in both cell lines, but at a much higher frequency in KG1 than in HL60. In KG1 cells, the AML1-ETO hybrid gene [associated with the t(8;21) translocation of acute myeloid leukemia] occurred significantly more often than the BCR-ABL [associated with t(9;22) chronic myeloid leukemia] or the DEK-CAN [associated with t(6;9) acute myeloid leukemia] fusion genes. These findings support the notion that ionizing radiation can directly generate leukemia-specific fusion genes but emphasize the differing susceptibility of different cell populations and the differing frequency with which the various fusion genes are formed. The selectivity observed at the primary level of gene fusion formation may explain at least in part the differential risk for development of some but not other forms of leukemia after high-dose radiation exposure.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Gamma Rays , HL-60 Cells/radiation effects , Hematopoietic Stem Cells/radiation effects , Leukemia, Radiation-Induced/genetics , Oncogene Proteins, Fusion , Oncogene Proteins/genetics , Transcription Factors/genetics , Translocation, Genetic/radiation effects , Core Binding Factor Alpha 2 Subunit , DNA Damage , DNA, Neoplasm/radiation effects , Fusion Proteins, bcr-abl/analysis , Humans , Oncogene Proteins/analysis , Polymerase Chain Reaction , RUNX1 Translocation Partner 1 Protein , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Transcription Factors/analysis
8.
Cancer Res ; 58(13): 2817-24, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9661896

ABSTRACT

We surveyed five human hematopoietic cell lines (HSB-2, MOLT-4, Reh, CEM, and HL-60) to determine whether any simple correlates with sensitivity to killing by gamma-irradiation might be revealed. The clonogenic survival gamma-ray dose-response curves for these cell lines cover a wide range of sensitivities. Consistent with previous results for murine hematopoietic cell lines, there was a clear correlation between the rapidity with which irradiation induced apoptosis and clonogenic radiosensitivity of a cell line, although the relationship between timing of apoptosis and radiosensitivity differed between human and murine cell lines. Flow cytometric determination of cell cycle distribution after irradiation showed that differences between human hematopoietic cell lines, in the rate of induction of apoptosis, were generally related to the functioning of cell cycle checkpoints. Whereas the rapidly dying and radiosensitive HSB-2 cell line underwent apoptosis from different points in the cell cycle, the more slowly dying cell lines showed a variety of cell cycle arrest profiles and initiated apoptosis after accumulation of cells in the G2 phase. The lag-phase between arrest in G2 and induction of apoptosis was comparable for MOLT-4, Reh, and CEM; however, HL-60 cells showed a markedly longer G2 arrest that correlated with their greater radioresistance. The results suggest that the total length of time available for DNA damage repair (irrespective of whether this time accrues as blockage in G1, S, or G2), prior to potential activation of apoptosis, is a critical determinant of radiosensitivity in human hematopoietic cell lines. Comparison of the p53 status of these cell lines suggested that mutations in the TP53 gene are contributing to the delay of induction of apoptosis seen in the more radioresistant cell lines. The sensitivity of MOLT-4 and HL-60 cells to killing by DNA-associated 125I decays was determined and was found to correlate with the relative sensitivity of these lines to gamma-irradiation. The highly localized deposition of energy by 125I decays argues that DNA damage is a potent initiator of apoptosis in these cell lines. The results presented suggest that differences in the radiosensitivity of the cell lines examined reflect differences in the rapidity of induction of apoptosis and that radiation-induced cell death in hematopoietic cells can be explained as a response to DNA damage.


Subject(s)
Apoptosis/radiation effects , Cell Cycle/radiation effects , DNA Damage , Gamma Rays , Cell Cycle/physiology , Cell Survival/radiation effects , DNA Repair , Dose-Response Relationship, Radiation , G2 Phase/physiology , G2 Phase/radiation effects , HL-60 Cells/radiation effects , Humans , Iodine Radioisotopes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Radiation Tolerance , Time Factors , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Protein p53/metabolism
9.
Cancer Res ; 57(17): 3640-3, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9288762

ABSTRACT

Mammalian cells respond to ionizing radiation (IR) with transient cell cycle arrest and induction of apoptosis. Here we show that IR increases the expression of the E2F-1 transcription factor and the entry of cells into S phase. E2F-1 transactivation function is inhibited by cyclin A-kinase to ensure orderly progression through S phase. However, in contrast to proliferating cells, IR treatment results in down-regulation of cyclin A-kinase. Expression of a dominant negative form of the E2F heterodimeric partner DP-1 confirmed the involvement of E2F in IR-induced S-phase entry. These findings also support opposing signals involving the induction of E2F and the down-regulation of cyclin A-kinase in the IR response.


Subject(s)
Carrier Proteins , Cell Cycle Proteins/radiation effects , DNA Damage , DNA-Binding Proteins/radiation effects , DNA/metabolism , Protein Kinases/radiation effects , S Phase/genetics , Transcription Factors/radiation effects , Apoptosis , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , E2F Transcription Factors , E2F1 Transcription Factor , Flow Cytometry , G1 Phase/genetics , HL-60 Cells/metabolism , HL-60 Cells/radiation effects , Humans , Protein Kinases/metabolism , RNA, Messenger/metabolism , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/metabolism
10.
Cancer Res ; 57(23): 5243-5, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9393742

ABSTRACT

An apoptosis-resistant mutant (VC-33) was selected from HL-60 by alternating exposure to camptothecin and etoposide. VC-33 cells demonstrated resistance to apoptosis as induced not only by camptothecin and etoposide but by a variety of other agents as well, including 1-beta-D-arabinofuranosylcytosine, hydroxyurea, calcium ionophore (A23187), cycloheximide, and UV irradiation. In an effort to identify the mechanism of such apoptosis resistance, a mRNA differential display analysis was used. Among a total of 12 bands with reduced expression in VC-33 cells, 1 cDNA clone was isolated that was hybridized to the wild-type transcript but not to the VC-33 transcript on Northern blotting. Partial sequence of this gene revealed 98% homology to mitochondrial NADH dehydrogenase subunit 5. When cell growth and intracellular ATP levels under glucose starvation were measured, VC-33 cells were found to be more sensitive than wild-type cells. Thus, NADH dehydrogenase deficiency may contribute, at least in part, to the mechanism of resistance to apoptosis in VC-33 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , HL-60 Cells/physiology , NADH Dehydrogenase/biosynthesis , NADH Dehydrogenase/deficiency , Apoptosis/drug effects , Apoptosis/radiation effects , Calcimycin/pharmacology , Camptothecin/pharmacology , Cell Division/drug effects , Clone Cells , Cloning, Molecular , Cytarabine/pharmacology , Deoxyglucose/pharmacology , Etoposide/pharmacology , Glucose/metabolism , HL-60 Cells/drug effects , HL-60 Cells/radiation effects , Humans , Hydroxyurea/pharmacology , Macromolecular Substances , Mitochondria/enzymology , Mutagenesis , NADH Dehydrogenase/genetics , Transcription, Genetic , Ultraviolet Rays
11.
Cancer Res ; 58(23): 5570-6, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9850096

ABSTRACT

Studies on the mechanism of apoptosis in this laboratory support a model in which signal transduction involving caspase 3 leads to activation of a serine protease called Mr 24,000 apoptotic protease (AP24), which then induces internucleosomal DNA fragmentation in the nucleus. This study examined the effect of Bcl-2 overexpression on activation of AP24 and the induction of DNA fragmentation by AP24 in isolated nuclei. It was demonstrated that overexpression of Bcl-2 in either HL-60 or PW leukemia cell lines suppressed activation of AP24 induced by either tumor necrosis factor or UV light and protected cells from apoptosis. Furthermore, nuclei isolated from Bcl-2-overexpressing cells were relatively resistant to internucleosomal DNA fragmentation induced by AP24 isolated from apoptotic cells. Bcl-2-overexpressing cells that were nutritionally depleted of glutathione (GSH) became sensitive to tumor necrosis factor- or UV light-induced activation of AP24 and underwent apoptotic cell death. Moreover, nuclei isolated from Bcl-2-overexpressing cells that were depleted of GSH became sensitive to AP24-induced DNA fragmentation. The addition of exogenous GSH blocked the proteolytic activity of AP24, as well as its ability to induce DNA fragmentation in normal isolated nuclei. These results indicate that Bcl-2 can attenuate at least two events in the AP24 apoptotic pathway: activation of AP24 and induction of DNA fragmentation by activated AP24. Furthermore, agents that deplete intracellular levels of GSH may have therapeutic use in the sensitization of Bcl-2-overexpressing cancer cells to apoptotic cell death.


Subject(s)
Apoptosis/physiology , DNA, Neoplasm/metabolism , Glutathione/pharmacology , Proto-Oncogene Proteins c-bcl-2/physiology , Serine Endopeptidases/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , DNA Damage , DNA, Neoplasm/drug effects , Enzyme Activation/drug effects , Glutathione/metabolism , Glutathione/physiology , HL-60 Cells/drug effects , HL-60 Cells/metabolism , HL-60 Cells/radiation effects , Humans , Nucleosomes/drug effects , Nucleosomes/metabolism , Nucleosomes/radiation effects , Oligopeptides/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Serine Endopeptidases/physiology , Tumor Necrosis Factor-alpha/pharmacology , Ultraviolet Rays
12.
Cancer Res ; 58(3): 413-20, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9458082

ABSTRACT

Recent investigations that showed that amiloride delayed or inhibited apoptosis indicated it might also attenuate cell cycle checkpoints activated by ionizing radiation. In this report, single- and dual-parameter flow cytometry were used to investigate the effects of amiloride on cell cycle progression, and the effectiveness of amiloride to attenuate the S and G2 phase checkpoint responses induced by 2.5, 5.0, and 7.5 Gy of gamma radiation. The late S-phase delay, noted at 8 h following irradiation, and a radiation-induced G2 block, which was maximum at 16 h after irradiation, were both significantly reduced in amiloride-treated samples. Attenuation of the radiation-induced late S phase and G2 blocks resulted in cell division without apparent apoptosis or necrosis over a 24-h period. Results presented indicate that amiloride reduces the radiation-induced G2 block in HL-60 cell populations almost equally well as caffeine and to a greater extent than staurosporine. Immunofluorescent detection and quantitation of cyclin B1 expression demonstrated that amiloride only significantly reduced cyclin B1 expression following 5.0 Gy, when there was a notable induction of a significant G2 delay, followed by a relatively rapid recovery in cycling potential. The results suggest that amiloride affects the radiation-triggered signaling cascades to alter the kinase activity of proteins associated with mitotic progression, particularly the cyclin B1-p34cdc2 complex. Alternatively, alterations in intracellular ion concentrations induced by amiloride may lead to changes in Ca2+-dependent signaling cascades and thereby decrease the radiation-mediated cell cycle perturbations.


Subject(s)
Amiloride/pharmacology , G2 Phase/drug effects , HL-60 Cells/drug effects , Radiation-Protective Agents/pharmacology , S Phase/drug effects , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Apoptosis/drug effects , Caffeine/pharmacology , Cyclin B/biosynthesis , Cyclin B/genetics , Cyclin B1 , DNA Fragmentation , DNA, Neoplasm/analysis , Enzyme Inhibitors/pharmacology , Flow Cytometry , G2 Phase/radiation effects , Gamma Rays , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/radiation effects , HL-60 Cells/radiation effects , Humans , S Phase/radiation effects , Staurosporine/pharmacology
13.
Oncogene ; 14(25): 3051-7, 1997 Jun 26.
Article in English | MEDLINE | ID: mdl-9223668

ABSTRACT

A novel polymerase chain reaction (PCR)-based method was used to identify candidate genes whose expression is altered in cancer cells by ionizing radiation. Transcriptional induction of randomly selected genes in control versus irradiated human HL60 cells was compared. Among several complementary DNA (cDNA) clones recovered by this approach, one cDNA clone (CL68-5) was downregulated in X-irradiated HL60 cells but unaffected by 12-O-tetradecanoyl phorbol-13-acetate, forskolin, or cyclosporin-A. DNA sequencing of the CL68-5 cDNA revealed 100% nucleotide sequence homology to the reported human Csa-19 gene. Northern blot analysis of RNA from control and irradiated cells revealed the expression of a single 0.7-kilobase (kb) messenger RNA (mRNA) transcript. This 0.7-kb Csa-19 mRNA transcript was also expressed in a variety of human adult and corresponding fetal normal tissues. Moreover, when the effect of X- or fission neutron-irradiation on Csa-19 mRNA was compared in cultured human cells differing in p53 gene status (p53-/- versus p53+/+), downregulation of Csa-19 by X-rays or fission neutrons was similar in p53-wild type and p53-null cell lines. Our results provide the first known example of a radiation-responsive gene in human cancer cells whose expression is not associated with p53, adenylate cyclase or protein kinase C.


Subject(s)
Breast Neoplasms/genetics , HL-60 Cells/radiation effects , Oncogenes/drug effects , Oncogenes/genetics , Oncogenes/radiation effects , Age Factors , Blotting, Northern , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Cloning, Molecular , Colforsin/pharmacology , Cyclosporine/pharmacology , DNA, Complementary , Dose-Response Relationship, Radiation , Down-Regulation , Gene Expression Regulation, Developmental , Gene Library , Genes, p53/drug effects , Genes, p53/radiation effects , HL-60 Cells/drug effects , HL-60 Cells/physiology , Humans , Molecular Sequence Data , Neutrons , Tetradecanoylphorbol Acetate/pharmacology , Tissue Distribution , Tumor Cells, Cultured , X-Rays
14.
FEBS Lett ; 579(21): 4829-36, 2005 Aug 29.
Article in English | MEDLINE | ID: mdl-16107253

ABSTRACT

The biological effect of radiofrequency (RF) fields remains controversial. We address this issue by examining whether RF fields can cause changes in gene expression. We used the pulsed RF fields at a frequency of 2.45 GHz that is commonly used in telecommunication to expose cultured human HL-60 cells. We used the serial analysis of gene expression (SAGE) method to measure the RF effect on gene expression at the genome level. We observed that 221 genes altered their expression after a 2-h exposure. The number of affected genes increased to 759 after a 6-h exposure. Functional classification of the affected genes reveals that apoptosis-related genes were among the upregulated ones and the cell cycle genes among the downregulated ones. We observed no significant increase in the expression of heat shock genes. These results indicate that the RF fields at 2.45 GHz can alter gene expression in cultured human cells through non-thermal mechanism.


Subject(s)
Electromagnetic Fields/adverse effects , Gene Expression/radiation effects , HL-60 Cells/radiation effects , Radio Waves/adverse effects , Dose-Response Relationship, Radiation , Gene Expression Profiling , Genome, Human , Humans , Telecommunications
15.
Leukemia ; 17(9): 1794-805, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12970779

ABSTRACT

It is now well established that the reduced capacity of tumor cells of undergoing cell death through apoptosis plays a key role both in the pathogenesis of cancer and in therapeutic treatment failure. Indeed, tumor cells frequently display multiple alterations in signal transduction pathways leading to either cell survival or apoptosis. In mammals, the pathway based on phosphoinositide 3-kinase (PI3K)/Akt conveys survival signals of extreme importance and its downregulation, by means of pharmacological inhibitors of PI3K, considerably lowers resistance to various types of therapy in solid tumors. We recently described an HL60 leukemia cell clone (HL60AR cells) with a constitutively active PI3K/Akt pathway. These cells were resistant to multiple chemotherapeutic drugs, all-trans-retinoic acid (ATRA), and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Treatment with two pharmacological inhibitors of PI3K, wortmannin and Ly294002, restored sensitivity of HL60AR cells to the aforementioned treatments. However, these inhibitors have some drawbacks that may severely limit or impede their clinical use. Here, we have tested whether or not a new selective Akt inhibitor, 1L-6-hydroxymethyl-chiro-inositol 2(R)-2-O-methyl-3-O-octadecylcarbonate (Akt inhibitor), was as effective as Ly294002 in lowering the sensitivity threshold of HL60 cells to chemotherapeutic drugs, TRAIL, ATRA, and ionizing radiation. Our findings demonstrate that, at a concentration which does not affect PI3K activity, the Akt inhibitor markedly reduced resistance of HL60AR cells to etoposide, cytarabine, TRAIL, ATRA, and ionizing radiation. This effect was likely achieved through downregulation of expression of antiapoptotic proteins such as c-IAP1, c-IAP2, cFLIP(L), and of Bad phosphorylation on Ser 136. The Akt inhibitor did not influence PTEN activity. At variance with Ly294002, the Akt inhibitor did not negatively affect phosphorylation of protein kinase C-zeta and it was less effective in downregulating p70S6 kinase (p70S6K) activity. The Akt inhibitor increased sensitivity to apoptotic inducers of K562 and U937, but not of MOLT-4, leukemia cells. Overall, our results indicate that selective Akt pharmacological inhibitors might be used in the future for enhancing the sensitivity of leukemia cells to therapeutic treatments that induce apoptosis or for overcoming resistance to these treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Inositol/pharmacology , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/antagonists & inhibitors , Tretinoin/pharmacology , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/metabolism , Caspases/metabolism , Chromones/pharmacology , Cytarabine/pharmacology , Cytochrome c Group/metabolism , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , HL-60 Cells/drug effects , HL-60 Cells/radiation effects , Humans , Inhibitor of Apoptosis Proteins , Inositol/analogs & derivatives , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Morpholines/pharmacology , PTEN Phosphohydrolase , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C-theta , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Radiation, Ionizing , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Transfection , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases , bcl-Associated Death Protein
16.
FEBS Lett ; 404(1): 27-33, 1997 Mar 03.
Article in English | MEDLINE | ID: mdl-9074631

ABSTRACT

Apoptosis or programmed cell death was induced in the human promyelocytic leukemia cell line HL-60 by UV irradiation or treatment with cytotoxic drugs (etoposide, camptothecin, melphalan or chlorambucil). These treatments caused a rapid increase in intracellular peroxide levels. Preincubation of HL-60 cells with the hydrogen peroxide-scavenging enzyme catalase (500 U/ml) inhibited apoptosis due to UV irradiation or low concentrations of camptothecin, etoposide or melphalan, but did not protect against higher concentrations. In contrast, superoxide anion levels in the cells remained unchanged upon treatment with cytotoxic drugs, while UV irradiation led to a transient doubling in superoxide levels. Exogenous superoxide dismutase (400 U/ml) provided modest protection against UV irradiation and had no effect on cytotoxic drug-induced apoptosis. The results suggest that both hydrogen peroxide and superoxide anion may be involved in the induction of apoptosis by UV irradiation. On the other hand, while exposure to cytotoxic drugs induces a large increase in intracellular peroxide levels, catalase is able to protect the cells from apoptosis only when low concentrations of these compounds are used, thus indicating the involvement of other factors in this process, particularly at higher drug concentrations.


Subject(s)
Apoptosis/drug effects , HL-60 Cells/metabolism , Peroxides/metabolism , Superoxides/metabolism , Anions , Apoptosis/radiation effects , Catalase/pharmacology , HL-60 Cells/drug effects , HL-60 Cells/radiation effects , Humans , Peroxides/pharmacology , Peroxides/radiation effects , Superoxides/pharmacology , Superoxides/radiation effects , Ultraviolet Rays
17.
Int J Radiat Oncol Biol Phys ; 43(2): 423-30, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-10030271

ABSTRACT

PURPOSE/OBJECTIVE: To investigate the effect of the enforced expression of p29Bcl-xL or its loop deletional mutant, p18Bcl-xLdelta, on irradiation-induced apoptosis and cell-cycle distribution of HL-60 cells. MATERIALS & METHODS: We compared the irradiation-induced molecular cascade of apoptosis in control human AML HL-60/neo versus Bcl-xL overexpressing (approximately 8-fold) (HL-60/Bcl-xL) and HL-60/Bcl-XLdelta cells that express the loop domain deletional mutant construct (delta26-83 AA) of Bcl-xL. The three cell lines were irradiated with 6MV photons to varying doses up to 20 Gy. Following this, cytosolic cyt c levels, caspase-3 activity, and the Bcl-2 family of proteins were evaluated utilizing Western blot analysis (whole cell lysate or cytosolic S-100 fraction). Apoptosis was assessed by internucleosomal DNA fragmentation, Annexin-V staining and FACS analysis, as well as by morphologic criteria. The cell-cycle effects of radiation were analyzed by flow cytometry. RESULTS: Eight hours following irradiation (12 Gy) of HL-60/neo cells, a marked increase (approximately 8-fold) in the cytosolic accumulation of cyt c in the S-100 fraction was observed. This was associated with the cleavage of caspase-3, as well as the generation of its poly (ADP-ribose) polymerase (PARP) and DFF (DNA fragmentation factor)-45 cleavage activity. Twenty-four to forty-eight hours after irradiation, internucleosomal DNA fragmentation and positive Annexin-V staining (32.3+/-3.3%) was detected in HL-60/neo cells. In contrast, in both HL-60/Bcl-xL and HL-60/Bcl-xLdelta cells, a significantly lower percentage of apoptotic cells (p<0.05) were detected and internucleosomal DNA fragmentation was not induced. Following irradiation, Western analysis neither demonstrated any significant alteration in Bcl-2, p29Bcl-xL, p18Bcl-xLdelta, or Bax; nor induced CD95 (Fas receptor) or Fas ligand expression in any cell type. However, in all cell types, irradiation produced approximately a 2-fold increase in the percentage of cells in the G2/M phase of the cell cycle. CONCLUSION: These results demonstrate that an intact loop domain is not necessary for the full antiapoptotic function of Bcl-xL against irradiation-induced cytosolic accumulation of cyt c, caspase activation, and apoptosis of HL-60 cells. Additionally, the cell-cycle effects of ionizing radiation in HL-60 cells are not affected by enforced expression of Bcl-xL or Bcl-xLdelta.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Cytochrome c Group/metabolism , Cytosol/enzymology , Proto-Oncogene Proteins c-bcl-2/physiology , Proto-Oncogene Proteins/metabolism , Caspase 3 , G2 Phase , HL-60 Cells/radiation effects , Helix-Loop-Helix Motifs , Humans , Mitosis , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-2-Associated X Protein , bcl-X Protein
18.
Int J Radiat Oncol Biol Phys ; 59(3): 844-51, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15183488

ABSTRACT

PURPOSE: Developing myeloid cells are particularly sensitive to chemotherapy and ionizing radiation. Mature cells of the hematopoietic lineages, such as are found in the peripheral blood mononuclear cells (PBMCs), are much less sensitive for reasons that are not yet understood. Protecting the myeloid precursors from radiation or chemotherapy is an important goal. METHODS: We have used fluorescence microscopy to assess the ability of WR-1065, the active metabolite of amifostine (Ethyol), to protect cultured myeloid leukemic HL-60 cells or freshly isolated PBMCs from the induction of apoptosis by ionizing radiation or etoposide. RESULTS: WR-1065 greatly reduced the percentage of radiation-induced apoptosis in the p53 negative HL-60 cells 24 h after exposure to 8 Gy. WR-1065 also greatly reduced the percentage of HL-60 cells undergoing apoptosis 24 h after a 1-h exposure to 1 microM etoposide. The pan-caspase inhibitor ZVAD-fmk completely inhibited radiation-induced apoptosis in HL-60 cells when present for the first hour after exposure to radiation, but had no effect on cell survival. In contrast, neither WR-1065 nor ZVAD-fmk reduced the level of radiation-induced apoptosis in normal human PBMCs. CONCLUSION: These results suggest that pro-apoptotic pathways are present in immature myeloid cells that can be selectively protected from radiation or chemotherapy-induced apoptosis.


Subject(s)
Apoptosis/drug effects , HL-60 Cells/drug effects , HL-60 Cells/radiation effects , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/radiation effects , Mercaptoethylamines/pharmacology , Radiation-Protective Agents/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Caspase Inhibitors , Cell Survival , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Humans
19.
Biochem Pharmacol ; 50(7): 1021-9, 1995 Sep 28.
Article in English | MEDLINE | ID: mdl-7575657

ABSTRACT

Cell death via apoptosis is an important event involved in a number of immunological processes. Recently, apoptosis has been associated with oxidative stress in a number of cell systems. Here we assessed the inhibitory capacity of different antioxidants on UV- and drug-induced apoptosis in the human leukemic cell line, HL-60. We found that the oxygen radical scavenger, BHA, the radioprotector cysteamine and the metal chelators, pyrrolidinedithiocarbamate (PDTC), diethyldithiocarbamate (DEDTC), and dimethyldithiocarbamate (DMDTC), were able to significantly inhibit nuclear fragmentation and reduce the formation of apoptotic bodies in UV-irradiated human leukemic cells. Both BHA and PDTC were found to reduce DNA fragmentation as assessed by in situ DNA nick-end labelling and quantification thereof using fluorescence flow cytometry. In addition to inhibiting UV-induced apoptosis, PDTC was also capable of reducing the amount of apoptosis induced by a range of cytotoxic drugs, such as actinomycin-D, camptothecin, etoposide, and melphalan, whereas BHA and cysteamine were not as effective in these cases after more than four hours in culture when compared to PDTC. To further elucidate the working mechanism of PDTC, we have looked at the effect of PDTC on DNA fragmentation in isolated nuclei, under conditions that promote activation of endogenous endonuclease involved in apoptosis. In contrast to ZnCl2, a potent inhibitor of endonuclease activity, PDTC was unable to inhibit DNA-ladder formation in this assay. Taken together, these results indicate that oxygen radicals may have a central role to play in the induction of apoptosis and that dithiocarbamates can serve as potent inhibitors of apoptosis induced by a wide variety of stimuli.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , HL-60 Cells/drug effects , Butylated Hydroxyanisole/pharmacology , Cell Size/drug effects , Cysteamine/pharmacology , DNA/analysis , Dose-Response Relationship, Drug , Endonucleases/metabolism , HL-60 Cells/metabolism , HL-60 Cells/radiation effects , Humans , Hydrogen Peroxide/antagonists & inhibitors , Oxidative Stress , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Ultraviolet Rays
20.
Environ Health Perspect ; 104(11): 1188-98, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8959408

ABSTRACT

Some epidemiological studies suggest that exposure to power frequency magnetic fields (MFs) may be associated with an elevated risk of human cancer, but the experimental database remains limited and controversial. We investigated the hypothesis that 60-Hz MF action at the cellular level produces changes in gene expression that can result in neoplastic transformation. Twenty-four hour 200 microT continuous MF exposure produced negative results in two standard transformation systems (Syrian hamster embryo cells and C3H/10T1/2 murine fibroblasts) with or without postexposure to a chemical promoter. This prompted a reexamination of previously reported MF-induced changes in gene expression in human HL60 cells. Extensive testing using both coded and uncoded analyses was negative for an MF effect. Using the same exposure conditions as in the transformation studies, no MF-induced changes in ornithine decarboxylase expression were observed in C3H/10T1/2 cells, casting doubt on a promotional role of MF for the tested cells and experimental conditions.


Subject(s)
Cell Transformation, Neoplastic , Electromagnetic Fields/adverse effects , Gene Expression/radiation effects , Animals , Cell Line/radiation effects , Cricetinae , HL-60 Cells/radiation effects , Humans , Mice , Ornithine Decarboxylase/genetics , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL