Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
3.
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
4.
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
5.
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
6.
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
7.
PLoS One ; 2(3): e269, 2007 Mar 07.
Article in English | MEDLINE | ID: mdl-17342203

ABSTRACT

BACKGROUND: Chemo- and radiotherapeutic responses of leukemia cells are modified by integrin-mediated adhesion to extracellular matrix. To further characterize the molecular mechanisms by which beta1 integrins confer radiation and chemoresistance, HL60 human acute promyelocytic leukemia cells stably transfected with beta1 integrin and A3 Jurkat T-lymphoma cells deficient for Fas-associated death domain protein or procaspase-8 were examined. METHODOLOGY/PRINCIPAL FINDINGS: Upon exposure to X-rays, Ara-C or FasL, suspension and adhesion (fibronectin (FN), laminin, collagen-1; 5-100 microg/cm(2) coating concentration) cultures were processed for measurement of apoptosis, mitochondrial transmembrane potential (MTP), caspase activation, and protein analysis. Overexpression of beta1 integrins enhanced the cellular sensitivity to X-rays and Ara-C, which was counteracted by increasing concentrations of matrix proteins in association with reduced caspase-3 and -8 activation and MTP breakdown. Usage of stimulatory or inhibitory anti beta1 integrin antibodies, pharmacological caspase or phosphatidylinositol-3 kinase (PI3K) inhibitors, coprecipitation experiments and siRNA-mediated beta1 integrin silencing provided further data showing an interaction between FN-ligated beta1 integrin and PI3K/Akt for inhibiting procaspase-8 cleavage. CONCLUSIONS/SIGNIFICANCE: The presented data suggest that the ligand status of beta1 integrins is critical for their antiapoptotic effect in leukemia cells treated with Ara-C, FasL or ionizing radiation. The antiapoptotic actions involve formation of a beta1 integrin/Akt complex, which signals to prevent procaspase-8-mediated induction of apoptosis in a PI3K-dependent manner. Antagonizing agents targeting beta1 integrin and PI3K/Akt signaling in conjunction with conventional therapies might effectively reduce radiation- and drug-resistant tumor populations and treatment failure in hematological malignancies.


Subject(s)
Caspase Inhibitors , Cell Adhesion/physiology , Drug Resistance, Neoplasm , HL-60 Cells/physiology , Integrin beta Chains/physiology , Radiation Protection , Cell Adhesion/drug effects , Cell Survival/drug effects , Cell Survival/radiation effects , Collagen/pharmacology , Cytarabine/pharmacology , Fibronectins/pharmacology , HL-60 Cells/drug effects , HL-60 Cells/radiation effects , Humans , Integrin beta Chains/drug effects , Integrin beta Chains/radiation effects , Laminin/pharmacology , X-Rays
8.
Apoptosis ; 12(4): 743-51, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17219053

ABSTRACT

To study the inhibitory effects of caspase-3 mRNA antisense oligodeoxynucleotides (ASODNs) on apoptosis, we designed four ASODNs targeting different regions of caspase-3 mRNA and transfected them into human leukemia HL-60 cells. The transfected cells were given 10 Gy gamma-irradiation followed by incubation for 18 h and measurement of apoptosis and caspase-3 expression. Our results showed that ASODN-2 targeting the 5' non-coding region of sites -62 to -46, and ASODN-3 targeting the 5' coding region of sites -1 to 16, both reduced apoptosis measured by gel electrophoresis and flow cytometry. Hoechst 33258 staining and TUNEL assay revealed that apoptotic indexes in the ASODN-2 and ASODN-3 groups were significantly lower than those in the untransfected and mismatched oligodeoxynucleotide (MODN) groups. Immunocytochemistry, Western blotting and RT-PCR showed that expression levels of caspase-3 protein and mRNA in both ASODN-2 and ASODN-3 groups were decreased compared with those in the untransfected and MODN groups. In conclusion, caspase-3 mRNA ASODNs can inhibit gamma-radiation-induced apoptosis of HL-60 cells and reduce expression of caspase-3 protein and mRNA. The results suggest that antisense approach may be useful for therapeutic treatment of certain neurodegenerative diseases in which apoptosis is involved.


Subject(s)
Apoptosis/physiology , Caspase 3 , HL-60 Cells , Oligonucleotides, Antisense/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cell Shape , DNA Fragmentation , Gamma Rays , HL-60 Cells/physiology , HL-60 Cells/radiation effects , Humans , In Situ Nick-End Labeling , Oligonucleotides, Antisense/genetics
9.
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
10.
Bioorg Med Chem ; 14(24): 8712-28, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-16987666

ABSTRACT

A new class of compounds, the pyrrolo[2,3-h]quinolin-2-ones, nitrogen isosters of the angular furocoumarin Angelicin, was synthesized with the aim of obtaining new photochemotherapeutic agents with increased antiproliferative activity and lower undesired toxic effects than the lead compound. Two synthetic pathways were approached to allow the isolation both of the dihydroderivatives 10-17 and of the aromatic ring system 23. Compounds 10-17 showed a remarkable phototoxicity and a great UVA dose dependence reaching IC(50) values at submicromolar level. Intracellular localization of these compounds has been evaluated by means of fluorescence microscopy using tetramethylrhodamine methyl ester and acridine orange, which are specific fluorescent probes for mitochondria and lysosomes, respectively. A weak co-staining was observed with mitochondrial stain, whereas a specific localization in lysosomes was observed. Studies directed to elucidate the mode of action of this series of compounds revealed that they do not intercalate with DNA and do not induce photodamage to the macromolecule. On the contrary, they induce significative photodamage to lipids and proteins.


Subject(s)
Cell Proliferation/drug effects , Photochemotherapy , Photosensitizing Agents/pharmacology , Quinolones/pharmacology , Acridine Orange , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Chromatography, High Pressure Liquid , Cross-Linking Reagents/pharmacology , DNA/drug effects , DNA/radiation effects , DNA Damage , Erythrocyte Membrane/chemistry , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/radiation effects , Fibrosarcoma/drug therapy , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Fluorescent Dyes , HL-60 Cells/drug effects , HL-60 Cells/metabolism , HL-60 Cells/radiation effects , Humans , Intercalating Agents/pharmacology , Lipid Peroxidation/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Mass Spectrometry , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/metabolism , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Proteins/metabolism , Quinolones/chemical synthesis , Quinolones/chemistry , Rhodamines/metabolism , Ultraviolet Rays
11.
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
12.
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
13.
Leuk Lymphoma ; 44(10): 1767-73, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14692532

ABSTRACT

Two non-psychotropic cannabinoids, cannabidiol (CBD) and cannabidiol-dimethylheptyl (CBD-DMH), induced apoptosis in a human acute myeloid leukemia (AML) HL-60 cell line. Apoptosis was determined by staining with bisBenzimide and propidium iodide. A dose dependent increase of apoptosis was noted, reaching 61 and 43% with 8 microg/ml CBD and 15 microg/ml CBD-DMH, respectively, after a 24 h treatment. Prior exposure of the cells to gamma-irradiation (800 cGy) markedly enhanced apoptosis, reaching values of 93 and 95%, respectively. Human monocytes from normal individuals were resistant to either cannabinoids or gamma-irradiation. Caspase-3 activation was observed after the cannabinoid treatment, and may represent a mechanism for the apoptosis. Our data suggest a possible new approach to treatment of AML.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , Cannabidiol/analogs & derivatives , Cannabidiol/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Caspase 3 , Caspases/metabolism , Gamma Rays , HL-60 Cells/drug effects , HL-60 Cells/radiation effects , Humans , Monocytes/drug effects , Monocytes/radiation effects
14.
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
15.
Article in English | MEDLINE | ID: mdl-12973926

ABSTRACT

To understand the expression and effect of mismatch repair genes, hMSH2 and hMLH1, and to investigate anti-leukemic cell proliferation mechanism of curcumin, the levels of both genes were detected by multiple comparative RT-PCR. The protein of hMSH2 was determined by flow cytometry (FCM) and the gene mutation of hMSH2 and hMLH1 were detected by PCR-SSCP and microsatellite instability assay. After UV irradiation, the gene expression of hMSH2 and hMLH1 was not increased and showed no response. This phenomenon was not ascribed to gene mutation, because PCP-SSCP and microsatellite instability assay revealed no abnormal gel-shift band in both genes. After irradiation and addition of curcumin, the expression of hMSH2 mRNA increased and the cellular apoptotic rate also increased at the same time. The difference was statistically significant as compared with groups without addition of curcumin and control groups (P < 0.05). Our results suggested that when MMR system was inhibited by the same agents, curcumin can remove this suppression and switch to cellular apoptosis.


Subject(s)
Base Pair Mismatch/drug effects , Curcumin/pharmacology , DNA-Binding Proteins/drug effects , Proteins/drug effects , Proto-Oncogene Proteins/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Base Pair Mismatch/genetics , DNA Repair/drug effects , HL-60 Cells/radiation effects , Humans , MutS Homolog 2 Protein , Neoplasm Proteins/metabolism , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Ultraviolet Rays
16.
J Photochem Photobiol B ; 69(2): 71-85, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12633980

ABSTRACT

We studied the mechanism of the cytotoxic effects of 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT; induction with 1 mM ALA for 4 h followed by a blue light dose of 18 J/cm(2)) on the human promyelocytic leukemia cell line HL60 using biochemical and electron microscopy methods. The disruption of mitochondrial membrane potential, deltapsi(m), was paralleled by a decrease in ATP level, unmasking of the mitochondrial antigen 7A6, release of cytochrome c into the cytoplasm, activation of caspases 9 and 3 and cleavage of poly(ADP-ribose) polymerase (PARP). This was followed by DNA fragmentation. These data suggest that ALA-PDT activates the mitochondrial apoptotic pathway. The level of endoplasmic reticulum Ca(2+)-binding chaperones ERp57 and ERp72 and of anti-apoptotic proteins Bcl-2 and Bcl-x(L) was decreased whereas that of Ca(2+)-binding protein calmodulin and the stress protein HSP60 was elevated following ALA-PDT. Inhibition of the initiator caspase 9, execution caspase 3 and Ca(2+)-dependent protease m-calpain, did not prevent DNA fragmentation. We conclude that, in our in vitro model, ALA-based photodynamic treatment initiates several signaling processes in HL60 cells that lead to rapidly progressing apoptosis, which is followed by slow necrosis. Two apoptotic processes proceed in parallel, one representing the mitochondrial pathway, the other involving disruption of calcium homeostasis and activation of the endoplasmic reticulum stress-mediated pathway.


Subject(s)
Amino Acids, Neutral/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/radiation effects , HL-60 Cells/drug effects , Mitochondria/drug effects , Mitochondria/radiation effects , Photochemotherapy/methods , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Division/drug effects , Cell Division/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Endoplasmic Reticulum/ultrastructure , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , HL-60 Cells/physiology , HL-60 Cells/radiation effects , HL-60 Cells/ultrastructure , Humans , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Mitochondria/physiology , Neoplasm Proteins/metabolism , Photosensitizing Agents/pharmacology , Stress, Physiological/chemically induced , Stress, Physiological/metabolism , Stress, Physiological/pathology
17.
Blood ; 101(9): 3628-34, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12506040

ABSTRACT

The mevalonate pathway produces many critical substances in cells, including sterols essential for membrane structure and isoprenoids vital to the function of many membrane proteins. 3-Hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase is a rate-limiting enzyme in the mevalonate pathway. Because cholesterol is a product of this pathway, HMG-CoA reductase inhibitors (statins) are used to treat hypercholesterolemia. Statins are also toxic to several malignancies, including acute myeloid leukemia (AML). Although this toxicity has been attributed to the inhibition of Ras/Rho isoprenylation, we have previously shown that statin toxicity in primary AML cells (AMLs) does not correlate with Ras isoprenylation or with activating Ras mutations. In other studies, we have shown that hypoxic and oxidant injuries induce cholesterol increments in renal tubule cells and that statins sensitize these cells to injury by blocking protective cholesterol responses. We now demonstrate that exposing particular AMLs to radiochemotherapy induces much greater cellular cholesterol increments than those seen in similarly treated normal bone marrow. Treatment of these AMLs with mevastatin or zaragozic acid (which inhibits cholesterol synthesis but not isoprenoid synthesis) attenuates the cholesterol increments and sensitizes cells to radiochemotherapy. The extent of toxicity is affected by the availability of extracellular lipoproteins, further suggesting that cellular cholesterol is critical to cell survival in particular AMLs. Because zaragozic acid does not inhibit isoprenoid synthesis, these data suggest that cholesterol modulation is an important mechanism whereby statins exert toxic effects on some AMLs and that cholesterol modulators may improve therapeutic ratios in AML by impacting cholesterol-dependent cytoresistance.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cholesterol/biosynthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Leukemia, Myeloid/pathology , Lovastatin/analogs & derivatives , Lovastatin/pharmacology , Radiation-Sensitizing Agents/pharmacology , Tricarboxylic Acids/pharmacology , Acute Disease , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Cholesterol/physiology , Cytarabine/pharmacology , Daunorubicin/pharmacology , Drug Synergism , HL-60 Cells/drug effects , HL-60 Cells/metabolism , HL-60 Cells/radiation effects , Humans , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Membrane Lipids/physiology , Protein Prenylation/drug effects , Protein Processing, Post-Translational/drug effects , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects
18.
Cytometry ; 49(3): 113-8, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12442311

ABSTRACT

BACKGROUND: Much attention is being paid to the biologic effects of magnetic fields (MFs). Although MFs enhance tumorigenesis, they are neither mutagenic nor tumorigenic. The mechanism of their tumorigenic effect has not been elucidated. METHODS: To investigate the effect of MFs on apoptosis in HL-60 cells, we exposed the cells to static MFs of 6 mT generated by a magnetic disk of known intensity. Apoptosis was triggered by the DNA topoisomerase I inhibitor, camptothecin (CPT). Activation of caspases in situ using the fluorochrome-labeled inhibitor (FLICA) method and determination of plasma membrane integrity by excluding propidium iodide (PI) were measured by both laser scanning cytometry (LSC) and flow cytometry (FC). LSC and FC identified cells at three sequential stages of their demise: early apoptosis (cells with activated caspases and PI negative); late apoptosis (cells with activated caspases but unable to exclude PI); secondary necrosis (cells with apoptotic morphology no longer stained with FLICA, not excluding PI). RESULTS: MF alone did not induce any apoptogenic or necrogenic effect. CPT exposure led to the sequential appearance of apoptotic cells. In the presence of CPT and MF, the overall proportion of cells undergoing apoptosis was not significantly changed. However, we consistently observed a significant increase in the frequency of late apoptotic/necrotic cells when compared with samples treated with CPT alone (P < 0.001), as well as a decrease in the percentage of early apoptotic cells (P = 0.013). The data obtained by FC and LSC were consistent with each other, showing a similar phenomenon. CONCLUSION: Whereas MF alone or with CPT did not affect overall cell viability, it accelerated the rate of cell transition from apoptosis to secondary necrosis after induction of apoptosis by the DNA-damaging agent, CPT. Modulation of the kinetics of the transition from apoptosis to secondary necrosis by MF in vivo may play a role in inflammation and tumorigenesis.


Subject(s)
Apoptosis/radiation effects , Cell Membrane/radiation effects , HL-60 Cells/pathology , Magnetics/adverse effects , Apoptosis/physiology , Camptothecin/pharmacology , Cell Count , Cell Survival/radiation effects , Flow Cytometry , HL-60 Cells/radiation effects , Humans , Image Cytometry , Leukemia , Necrosis
19.
Radiat Med ; 20(3): 121-9, 2002.
Article in English | MEDLINE | ID: mdl-12126084

ABSTRACT

PURPOSE: A telomere consists of the short tandem DNA repeats of (T2AG3)n localized to the distal ends of chromosomes. Telomere repeat binding factor 2 (TRF2) has been implicated in the protection of chromosome ends. Recently, it has been reported that the loss of TRF2 induces apoptosis by various stimuli or genetic technique, however, the effects of radiation are not known. Therefore, this study investigated the interaction between TRF2 and radiation. MATERIALS AND METHODS: Western blot analysis, immunohistochemistry, and a DNA fragmentation assay for the detection of apoptosis were performed. The interaction between elastase and TRF2 was also investigated in vitro. RESULTS: Western blot analyses and immunohistochemistry showed that gamma-rays induce the temporary accumulation and subsequent loss of TRF2 protein in the nuclei of irradiated HL60 cells. Following DNA fragmentation, the loss of TRF2 could be detected. TRF2 was broken down by elastase, which was translocated into the nucleus before the loss of TRF2. CONCLUSIONS: The results of the study showed that irradiation first induces activation of TRF2, consequently protecting the end of the chromosome. Subsequently, translocation of elastase into the nucleus results in the breakdown of TRF2 after DNA fragmentation has occurred.


Subject(s)
Apoptosis/radiation effects , Gamma Rays/adverse effects , HL-60 Cells/radiation effects , Telomere/genetics , Telomeric Repeat Binding Protein 2/radiation effects , Blotting, Western , Humans , Leukocyte Elastase/metabolism , Telomere/radiation effects
20.
Radiats Biol Radioecol ; 42(2): 164-8, 2002.
Article in Russian | MEDLINE | ID: mdl-12004612

ABSTRACT

It was shown that the addition of synthetic six-membered peptide (HLDF-6) and its Tyr-analog (HLDF-Y) to cultural medium significantly increased the survival of cells HL-60, treated by cold shock. The prophylactic administration of HDLF-Y (1 mg/kg, 4 hours prior to applied actions) decreased the response of hypothalamushypophysis-adrenal glands system and sympathicoadrenal system of rat males on supercooling and also increased the resistance of mouse males to supercooling and X-irradiation. In the experiences with females HDLF-Y did not show the similar biological activity.


Subject(s)
Adrenal Cortex Hormones/analysis , Biogenic Monoamines/analysis , HL-60 Cells/radiation effects , Neoplasm Proteins , Peptide Fragments/pharmacology , Radiation Tolerance , Adaptation, Physiological , Adrenal Glands/chemistry , Animals , Apoptosis , Cell Differentiation , Cell Survival , Cold Temperature , Culture Media , Data Interpretation, Statistical , Dogs , Epinephrine/analysis , Female , Fluorometry , HL-60 Cells/drug effects , Humans , Male , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Norepinephrine/analysis , Radiation Dosage , Radiation, Ionizing , Radiation-Protective Agents , Rats , Rats, Wistar , Serotonin/analysis , Sex Factors , Spleen/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...