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










Publication year range
1.
Int J Hyperthermia ; 20(2): 131-9, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15195507

ABSTRACT

All biochemical and cytogenetic data on radiosensitization by heat treatment at and above 43 degrees C indicate that inhibition of DNA repair plays a central role. There are several DNA repair pathways involved in restoration of damage after ionising irradiation and the kinetics of all of them are affected by heat shock. This, however, does not imply that the inhibition of each of these pathways is relevant to the effect of heat on cellular radiosensitivity. The current review evaluates the available data on heat radiosensitization in mutant or knockout cell lines defective in various DNA repair proteins and/or pathways. The data show that thermal inhibition of the non-homologous end-joining pathway (NHEJ) plays no role in heat radiosensitization. Furthermore, limited data suggest that the homologous recombination pathway may also not be a major heat target. By deduction, it is suggested that inhibition of base damage repair (BER) could be the crucial step in radiosensitization by heat. While a lack of mutant cell lines and redundancy of the BER pathway have hampered efforts toward a conclusive study, biochemical and correlative evidence support this hypothesis.


Subject(s)
DNA Repair/genetics , Hyperthermia, Induced , Radiation Tolerance/physiology , Animals , Cell Line , Humans , Mutation
2.
Int J Hyperthermia ; 20(2): 144-56, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15195509

ABSTRACT

The mechanism by which hyperthermia sensitizes mammalian cells to ionizing radiation remains to be elucidated, but an overwhelming amount of circumstantial evidence suggests that heat radiosensitization might be mediated by inhibition of double-strand break repair, particularly after exposure of irradiated cells to heat treatments in excess of about 43 degrees C. In mammalian cells, double-strand break repair usually occurs via two pathways, non-homologous end-joining and homologous recombination. Several reports suggest a role for non-homologous end-joining in heat radiosensitization, while others implicate homologous recombination as a target. However, cell lines that are compromised in either the non-homologous end-joining or homologous recombination pathway are still capable of being radiosensitized, suggesting that heat affects both pathways. Indeed, several of the proteins involved in one or both of these pathways have been observed to undergo alterations or translocation after unirradiated or irradiated cells are exposed to heat shock. The work summarized in this review implicates proteins of the Mre11/Rad50/Nbs1 complex as targets for heat radiosensitization.


Subject(s)
Cell Cycle Proteins/physiology , DNA Repair Enzymes/physiology , DNA-Binding Proteins/physiology , Heat-Shock Response/physiology , Nuclear Proteins/physiology , Radiation Tolerance/physiology , Acid Anhydride Hydrolases , Animals , Cell Line/physiology , Cell Line/radiation effects , Humans , Hyperthermia, Induced , MRE11 Homologue Protein
3.
Radiat Res ; 156(6): 767-74, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11741501

ABSTRACT

XRCC5 (also known as Ku80) is a component of the DNA-dependent protein kinase (DNA-PK), existing as a heterodimer with G22P1 (also known as Ku70). DNA-PK is involved in the nonhomologous end-joining (NHEJ) pathway of DNA double-strand break (DSB) repair, and kinase activity is dependent upon interaction of the Ku subunits with the resultant DNA ends. Nuclear XRCC5 is normally extractable with non-ionic detergent; it is found in the soluble cytoplasmic fraction after nuclear isolation with Triton X-100. In this study, we found that heating at 45.5 degrees C causes a decreased extractability of XRCC5 from the nuclei of human U-1 melanoma or HeLa cells. Such decreases in extractability are indicative of protein aggregation within nuclei. Recovery of extractability of XRCC5 to that of unheated control cells was observed after incubation at 37 degrees C after heat shock. The decrease in extractability and the kinetics of recovery were dependent on dose, although the decrease in extractability reached a plateau after heating for 15 min or more. Thermotolerant U-1 cells also showed decreased extractability of XRCC5, but to a lesser degree compared to nontolerant cells. When a comparable initial reduction of extractability of XRCC5 was induced in both thermotolerant and nontolerant cells, the kinetics of recovery was nearly identical. The kinetics of recovery of the extractability of XRCC5 was different from that of total nuclear protein in nontolerant cells; recovery of extractability of XRCC5 occurred faster initially and returned to the level in unheated cells faster than total nuclear protein. Similar results were obtained for thermotolerant cells, with differences between the initial recovery of the extractability of XRCC5 and total protein being particularly evident after longer heating times. Heat has been shown to inactivate XRCC5. We speculate that inactivation of XRCC5 after heat shock results from protein aggregation, and that changes in XRCC5 may, in part, lead to inhibition of DSB repair through inactivation of the NHEJ pathway.


Subject(s)
Adaptation, Physiological , Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/metabolism , Hot Temperature , Nuclear Proteins/metabolism , DNA Repair , Humans , Kinetics , Ku Autoantigen , Microscopy, Confocal , Microscopy, Fluorescence , Tumor Cells, Cultured
4.
Radiat Res ; 156(1): 95-102, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11418077

ABSTRACT

Zhu, W-G., Seno, J. D., Beck, B. D. and Dynlacht, J. R. Translocation of MRE11 from the Nucleus to the Cytoplasm as a Mechanism of Radiosensitization by Heat. Radiat. Res. 156, 95-102 (2001).Hyperthermia sensitizes mammalian cells to ionizing radiation, presumably by inhibiting the repair of radiation-induced double-strand breaks (DSBs). However, the mechanism by which heat inhibits DSB repair is unclear. The nuclear protein MRE11 is a component of a multi-protein complex involved in nonhomologous end joining (NHEJ) of radiation-induced DSBs. Using one-dimensional sodium dodecylsulfate polyacrylamide gel electrophoresis and Western blotting, we found that MRE11 is translocated from the nucleus to the cytoplasm when human U-1 melanoma or HeLa cells are heated for 15 min at 45.5 degrees C or when cells are heated after irradiation with 12 Gy of X rays. No such translocation is observed in unheated irradiated cells. The kinetics of migration of MRE11 to the cytoplasm was dependent upon whether the heated cells were irradiated, while the magnitude of redistribution of MRE11 was dependent upon post-treatment incubation time at 37 degrees C. Cytoplasmic MRE11 content reached a maximum 2-4 h after heating; the increase was not due to new protein synthesis. Partial recovery of nuclear MRE11 content was observed when heated cells or heated irradiated cells were incubated for up to 7 h at 37 degrees C after treatment. Western blotting results showing translocation of MRE11 from the nucleus to the cytoplasm after heating and irradiation were confirmed using confocal microscopy and immunofluorescence staining of fixed cells. Our data suggest that radiosensitization by heat may be caused, at least in part, by translocation of the DNA repair protein MRE11 from the nucleus to the cytoplasm.


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Hot Temperature , Radiation Tolerance/physiology , Active Transport, Cell Nucleus/physiology , Active Transport, Cell Nucleus/radiation effects , Blotting, Western , Chromosome Breakage , DNA Repair/physiology , DNA Repair/radiation effects , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , MRE11 Homologue Protein , Melanoma/metabolism , Reproducibility of Results , Time Factors , Tumor Cells, Cultured , X-Rays
5.
Radiat Res ; 154(5): 515-30, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11025648

ABSTRACT

Several nuclear matrix proteins are substrates for proteolytic cleavage during apoptosis. Using Western blotting, the temporal patterns of cleavage of three nuclear matrix proteins (lamin B, NUMA and the nucleoporin TPR) were compared in HL60 cells induced to undergo apoptosis after irradiation, heat shock or treatment with etoposide. Flow cytometry was used to compare the kinetics of post-cleavage degradation of lamin B, NUMA and TPR after irradiation, and to correlate DNA fragmentation with protein degradation in cells induced to undergo apoptosis with different agents. During radiation-induced apoptosis, cleavage and subsequent degradation of lamin B, NUMA and TPR occurred with different kinetics. Low-molecular-weight DNA fragmentation occurred subsequent to the initiation of NUMA cleavage, coincided with lamin B cleavage, but occurred before more extensive degradation of lamin B and NUMA. A similar sequence was observed for cells treated with etoposide. However, during heat-induced apoptosis, cleavage of lamin B and NUMA occurred much sooner compared to other agents, with NUMA cleaved into multiple fragments within 15 min after heating. We conclude that the hierarchical sequence and kinetics of degradative events contributing to nuclear disassembly during apoptosis are highly dependent on the inducing agent. Furthermore, the nuclear pore complex, like the nuclear lamina and internal nuclear matrix, is a target for proteolytic cleavage.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , DNA Fragmentation/radiation effects , Etoposide/pharmacology , Nuclear Proteins/metabolism , Antigens, Nuclear , DNA Fragmentation/drug effects , HL-60 Cells , Hot Temperature , Humans , Nuclear Proteins/drug effects , Nuclear Proteins/radiation effects
6.
Radiat Res ; 152(6): 590-603, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10581529

ABSTRACT

Human promyelocytic leukemia (HL60) cells were irradiated with 10 or 50 Gy of X rays and studied for up to 72 h postirradiation to determine the mode of death and assess changes in the nuclear matrix. After 50 Gy irradiation, cells were found to die early, primarily by apoptosis, while cells irradiated with 10 Gy died predominantly by necrosis. Disassembly of the nuclear lamina and degradation of the nuclear matrix protein lamin B occurred in cells undergoing radiation-induced apoptosis or necrosis. However, using Western blotting and a recently developed flow cytometry assay to detect changes in nuclear matrix protein content, we found that the kinetics and mechanisms of disassembly of the nuclear lamina are different for each mode of cell death. During radiation-induced apoptosis, cleavage and degradation of lamin B to a approximately 28-kDa fragment was detected in most cells within 4-12 h after irradiation. Measurements of dual-labeled apoptotic cells revealed that nonrandom DNA fragmentation was evident prior to or concomitant with breakdown of the nuclear lamina. Disassembly of the nuclear lamina during radiation-induced necrosis occurred much later (between 30-60 h after irradiation), and a different cleavage pattern of lamin B was observed. Degradation of the nuclear lamina was also inhibited in apoptosis-resistant BCL2-overexpressing HL60 cells exposed to 50 Gy until approximately 48 h after irradiation. These data indicate that breakdown of the nuclear matrix may be a common element in radiation-induced apoptosis and necrosis, but that the mechanisms and temporal patterns of breakdown of the nuclear lamina during apoptosis are distinct from those of necrosis.


Subject(s)
Apoptosis/radiation effects , DNA, Neoplasm/radiation effects , Nuclear Matrix/radiation effects , Cell Death/radiation effects , DNA, Neoplasm/isolation & purification , Dose-Response Relationship, Radiation , Electrophoresis, Agar Gel , HL-60 Cells , Humans , Kinetics , Necrosis , Nuclear Matrix/ultrastructure , Time Factors , X-Rays
7.
J Cell Biochem ; 75(4): 620-8, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10572245

ABSTRACT

Previous work in our laboratory indicates that the nuclear matrix protein lamin B is a "prompt" heat shock protein, which increases significantly when human U-1 melanoma and HeLa cells are exposed to 45.5 degrees C for 5-40 min. Using Western blotting, we found that the lamin B content in U-1 and HeLa cells increased to a greater extent during post-heat incubation at 37 degrees C than during the heat dose itself. When HeLa cells were heated at 45.5 degrees C for 30 min, and then incubated at 37 degrees C for up to 7 h, lamin B content was increased significantly (1.69-fold maximum increase at 3 h) compared to unincubated heated cells. Also, thermotolerant HeLa cells showed a greater increase (up to 1.72-fold) in lamin B content during subsequent heating compared to nontolerant cells. The increase in lamin B content in thermotolerant cells, or when heated cells were incubated at 37 degrees C, was also observed in U-1 cells. HeLa cells heated in the presence of glycerol (a heat protector) showed a 1.21-1.72-fold increase in lamin B content compared to cells heated for 10-30 min without glycerol. In contrast, lamin B content decreased 1.23-1.85-fold when cells were heated for 10-30 min in the presence of procaine (a heat sensitizer) compared to cells heated without procaine. These data suggest that lamin B may play an important role in the heat shock response, and that modulation of lamin B content by heat sensitizers or protectors may play a role in regulation of heat sensitivity.


Subject(s)
HeLa Cells/metabolism , Melanoma/metabolism , Nuclear Proteins/metabolism , Anesthetics, Local/pharmacology , Blotting, Western , Cell Survival/drug effects , Glycerol/pharmacology , HeLa Cells/drug effects , Heat-Shock Response/drug effects , Humans , Lamin Type B , Lamins , Procaine/pharmacology , Time Factors , Tumor Cells, Cultured
8.
Int J Cancer ; 81(5): 808-12, 1999 May 31.
Article in English | MEDLINE | ID: mdl-10328237

ABSTRACT

An ideal treatment modality for metastasizing tumors should eradicate the primary tumor and elicit a systemic, tumor-selective response leading to elimination of metastases and long-term tumor resistance. Also, it should be induced by local treatment at the primary site, to limit adverse systemic effects. A new method for treating metastatic tumors which utilizes a combination of a near-infrared laser, a photosensitizer and an immunoadjuvant has been developed. It involves intra-tumor injection of the sensitizer/adjuvant solution, followed by local non-invasive laser irradiation. It has produced regression and total eradication of treated primary tumors and untreated metastases at remote sites against mammary tumors in rats. Successfully treated tumor-bearing rats showed total tumor resistance to subsequent tumor rechallenge. Our histochemical results showed that sera from cured tumor-bearing rats contained antibodies that bound strongly to the plasma membrane of both living and preserved tumor cells. Western blot analysis of tumor cell proteins using sera from successfully treated rats as the source of primary antibodies also showed distinct bands, indicating induction of tumor-selective antibodies. Our findings indicate that a systemic, long-term effect on metastatic tumors can be induced by local application of laser photo-immunotherapy.


Subject(s)
Immunotherapy , Lasers , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , Photochemotherapy , Adjuvants, Immunologic/therapeutic use , Animals , Antibodies, Neoplasm/metabolism , Blotting, Western , Chitin/analogs & derivatives , Chitin/therapeutic use , Chitosan , Disease-Free Survival , Female , Indocyanine Green/therapeutic use , Mammary Neoplasms, Experimental/mortality , Neoplasm Transplantation , Rats , Rats, Wistar , Remission Induction , Survival Rate , Time , Tumor Cells, Cultured
9.
J Cell Physiol ; 178(1): 28-34, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9886487

ABSTRACT

The cellular response to hyperthermia involves the increased synthesis of heat shock proteins (HSPs) within several hours after treatment. In addition, a subset of proteins has been shown to be increased immediately after heating. These "prompt" HSPs are predominantly found in the nuclear matrix-intermediate filament fraction and are not present or detectable in unheated cells. Since the nuclear matrix has been suggested to be a target for heat-induced cell killing, prompt HSPs may play a prominent role in the heat shock response. Using Western blotting and flow cytometry, we found that an increase in the synthesis of lamin B, one of the major proteins of the nuclear lamina, is induced during heating at 45.5 degrees C but not during heating at 42 degrees C. Since it is an abundant protein which is constitutively expressed in mammalian cells, lamin B appears to be a unique member of the prompt HSP family. The kinetics of induction of lamin B during 45.5 degrees C heating did not correlate with the dose-dependent reduction in cell survival. While increased levels of lamin B during 45.5 degrees C heating do not appear to confer a survival advantage directly, a possible role for lamin B in cellular recovery after heat shock cannot be discounted.


Subject(s)
Heat-Shock Proteins/metabolism , Nuclear Proteins/metabolism , Cell Survival/physiology , Flow Cytometry , Fluorescent Antibody Technique , HeLa Cells , Heat-Shock Proteins/analysis , Hot Temperature , Humans , Kinetics , Lamin Type B , Lamins , Melanoma , Nuclear Proteins/analysis , Time Factors
10.
Anticancer Res ; 18(6A): 4043-9, 1998.
Article in English | MEDLINE | ID: mdl-9891443

ABSTRACT

Many types of mammalian cells can repair potentially lethal damage (PLD) when incubated in conditioned medium (C-med) or Hank.'s balanced salt solution (HBSS) after irradiation. In this study, we found that there is an increase in clonogenic survival in FM3A cells, mouse mammary carcinoma cells when cells are incubated in C-med, but not in HBSS after irradiation. HBSS was lethal to unirradiated FM3A cells, and apoptosis was observed when cells were exposed to HBSS for more than 10 hours. However, irradiated cells underwent rapid apoptotic cell death and there was a decrease in clonogenic survival when cells were incubated in HBSS for a short time (3 h). Apoptotic cell death, was decreased significantly when irradiated cells were incubated in HBSS with 2% fetal calf serum. In addition, clonogenic survival was decreased significantly when irradiated cells were incubated in C-med with cycloheximide (CHX), an inhibitor of protein synthesis. Thus, the rapid apoptotic death of irradiated FM3A cells induced by HBSS might be due to the absence of serum or inhibition of protein synthesis. Some newly synthesized proteins, or proteins normally found in serum, may be required for irradiated cells to recover from X-ray induced damage.


Subject(s)
Apoptosis/physiology , Isotonic Solutions/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cattle , Cell Cycle/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Culture Media , Culture Media, Conditioned , Female , Mammary Neoplasms, Experimental , Mice , Tissue Preservation , Tumor Cells, Cultured , Tumor Stem Cell Assay , X-Rays
11.
Clin Cancer Res ; 3(6): 939-45, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9815769

ABSTRACT

The potential of retinoic acid as a radiosensitizer was investigated using SiHa and CC-1 human uterine cervical carcinoma cell lines, representative of high- and low-grade lesions, respectively. SiHa was significantly (P < 0.05) radiosensitized, whereas CC-1 was not. Although 48 h of treatment with 5 microM 13-cis-retinoic acid prior to irradiation was sufficient to induce radiosensitization, continuation of treatment after irradiation significantly increased the effect (P < 0.05). Three hypotheses were tested to explain the different responses of the two lines. One hypothesis was that SiHa is more sensitive to retinoic acid than CC-1. Measurement of growth revealed that SiHa was more sensitive to growth inhibition by retinoic acid than CC-1. The second hypothesis was that retinoic acid increases the proportion of G1-phase cells in SiHa but not in CC-1. This was found not to be true, because a retinoic acid treatment schedule that induced radiosensitization did not alter cell cycle distribution profiles in the absence of radiation. The third hypothesis was that retinoic acid alters the cell cycle response of SiHa but not CC-1 to radiation. Postirradiation cell cycle profiles revealed that retinoic acid increased G1 delay in SiHa, whereas CC-1 exhibited no significant G1 delay. Both lines exhibited G2 delays that were unaffected by retinoic acid. In conclusion, radiosensitization of SiHa but not CC-1 may be explained by different sensitivities to retinoic acid and differences in postirradiation cell cycle responses. Radiosensitization at radiation doses used clinically was observed when retinoic acid was administered both before and after irradiation.


Subject(s)
Cell Cycle/radiation effects , Cell Survival/radiation effects , Radiation-Sensitizing Agents/toxicity , Tretinoin/toxicity , Cell Cycle/drug effects , Cell Division/drug effects , Cell Division/radiation effects , Cell Survival/drug effects , Cobalt Radioisotopes , Dose-Response Relationship, Radiation , Female , Humans , Radiation Tolerance , Tumor Cells, Cultured , Uterine Cervical Neoplasms
12.
Cytometry ; 24(4): 348-59, 1996 Aug 01.
Article in English | MEDLINE | ID: mdl-8866219

ABSTRACT

The nuclear matrix (NM) is an important structural component of the nucleus that participates in the regulation of several diverse metabolic processes. Immunometric assays have shown that alterations in NM-associated functions and morphological characteristics may occur as a result of changes in NM composition. Recent evidence suggests that detection of quantitative or qualitative changes in nuclear matrix protein (NMP) composition may be useful in the diagnosis of cancer and as a reliable indicator of cell death. We have developed an in situ flow cytometric technique for the simultaneous detection of specific NMPs and DNA content in fixed, permeabilized cells. Illustrative results from two different applications of these methods involving two different cell lines (human melanoma and promyelocytic leukemia) are presented, including: 1) measurements of NM breakdown in necrotic and apoptotic cells after treatment with the cytotoxic agents camptothecin, etoposide, or hyperthermia; and 2) detection of changes in NMP content immediately after heat shock. We demonstrate that the technique is useful for the identification of cell-cycle specificity of NM breakdown and allows correlations to be made between the kinetics of DNA fragmentation and NMP solubilization. Furthermore, our studies indicate that flow cytometric detection of changes in NM composition may be useful for identifying different modes and temporal patterns of cell death. We discuss other potential applications of the technique and advantages over standard biochemical assays.


Subject(s)
Flow Cytometry/methods , Nuclear Proteins/analysis , Antibodies, Monoclonal , Antigens, Nuclear , Cell Cycle/immunology , DNA/analysis , DNA Fragmentation/immunology , Fluorescence , Hot Temperature , Staining and Labeling/methods , Tissue Fixation/methods , Tumor Cells, Cultured
13.
Radiat Res ; 146(2): 227-31, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8693073

ABSTRACT

The focus of this investigation was to determine whether the nitrone spin-trapping compounds alpha-phenyl-N-tert-butylnitrone (PBN), 5,5-dimethyl-1-pyrroline-N-oxide (DMPO) and alpha-(4-pyridyl-1-oxide)-N-tert-butylnitrone (POBN) are radioprotectors. Two methods were used to assess for radioprotection: measurement of oxidative damage to DNA bases and mammalian cell survival assays. Oxidative damage to DNA was quantified by measuring the relative amounts of 8-hydroxy-2'-deoxyguanosine (8-OH-dG) produced by the reaction of hydroxyl radicals (OH.) with 2-deoxyguanosine (dG) after irradiation. PBN, DMPO and POBN, when dissolved in aqueous solutions of either dG or naked salmon sperm DNA, reduced the formation of 8-OH-dG by 137Cs gamma irradiation significantly. The spin-trapping agents, especially PBN at lower concentrations, were more effective in preventing radiation-induced formation of 8-OH-dG in naked DNA than in free dG. These data suggest that PBN, DMPO and POBN act as free radical scavengers which may associate with DNA and afford protection against gamma rays. However, no enhancement of survival was observed when Chinese hamster ovary (CHO) cells were exposed to high non-toxic concentrations of PBN or POBN prior to and during irradiation with 60Co gamma rays and scored for clonogenic survival. DMPO provided only minimal protection from radiation-induced cell killing.


Subject(s)
Cyclic N-Oxides/pharmacology , Nitrogen Oxides/pharmacology , Radiation-Protective Agents/pharmacology , Spin Labels , 8-Hydroxy-2'-Deoxyguanosine , Animals , CHO Cells , Cell Survival/radiation effects , Cricetinae , DNA Damage , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/analysis , Pyridines
14.
Int J Radiat Biol ; 68(2): 141-52, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7658139

ABSTRACT

For a given amount of cell killing, heat alone (10-80 min, 45.5 degrees C) induced very few double-strand breaks (dsbs) compared with X-rays. Furthermore, 10 min at 45.5 degrees C immediately prior to X-rays caused only a 1.3-fold increase in the slope of the X-ray-induced dsb dose-response curve, i.e. 0.67 +/- 0.006 (95% confidence) dsbs/100Mbp/Gy for heated cells compared with 0.53 +/- 0.005 for unheated control cells. However, this same heat treatment caused > 5-fold inhibition in the rate of repair of dsbs induced by 60-Gy X-rays, with the degree of inhibition being much less in thermotolerant (TT) cells than in non-tolerant (NT) cells. This reduced inhibition of repair in TT cells correlated with the more rapid removal of excess nuclear protein from nuclei isolated from TT cells than from NT cells. These results plus a TT ratio of 2-3 for both heat-induced radiosensitization and heat-inhibition of repairing dsbs are consistent with the hypothesis that heat radiosensitization results primarily from heat aggregation of nuclear protein interfering with access of repair enzymes to DNA dsbs. The selective heat-radiosensitization of S-phase cells, however, may result from an increase in radiation-induced dsbs in or near replicating regions. For example, a preferential increase in dsbs in replicating DNA compared with bulk DNA was found following either hyperthermia alone (10-30 min, 45.5 degrees C) or a combined treatment (10 min, 45.5 degrees C before 60 Gy). A 30-min treatment at 45.5 degrees C induced dsbs equivalent to approximately 10 Gy in replicating DNA compared with 3-5 Gy in bulk DNA. When cells were heated immediately before irradiation, the increase in dsbs induced in the replicating DNA by 60 Gy was equivalent to 200 Gy. We hypothesize that the observed 2-fold increase in single-stranded regions in replicating DNA after heat resulted in radiation selectively inducing dsbs at or near the replication fork where the heat-induced increase in single-stranded DNA should occur. Thus, this preferential increase in dsbs in the replicating DNA by heat alone and especially when heat was combined with radiation may explain at least in part, the high sensitivity of S-phase cells to heat killing and heat radiosensitization.


Subject(s)
DNA Damage , Electrophoresis, Gel, Pulsed-Field , Hot Temperature , Animals , CHO Cells , Cricetinae , DNA Damage/radiation effects , DNA Repair , DNA Replication , In Vitro Techniques , Nuclear Proteins/metabolism , Time Factors , X-Rays
15.
Cancer Res ; 54(15): 4129-37, 1994 Aug 01.
Article in English | MEDLINE | ID: mdl-8033146

ABSTRACT

The chemotherapeutic drug etoposide (VP-16) causes the equilibrium reaction between noncleavable and cleavable topoisomerase II-DNA complexes to shift in favor of the cleavabel complex [H. Zang, P. D'Arpa, and L.F. Liu, Cancer Cells (Cold Spring Harbor), 2:23-27, 1990]. Pulsed-field gel electrophoresis was used to study induction and removal of cleavable complexes in cells heated before, during, or after VP-16 treatment. Pulsed-field gel electrophoresis results were evaluated both as the fraction of activity (DNA) released from the plug and as the number of double-strand breaks (DSBs) calculated from molecular weight distributions; both end points led to the same conclusions. When cells were heated at 42 degrees C during treatment with VP-16 (12 micrograms/ml up to 60 min), a slight decrease in cleavable complexes (from 30 to 20 DSBs/100 megabase pairs) was detected immediately after treatment when compared with cells treated with the drug at 37 degree C. Furthermore, heating at 42 degrees C caused a slight decrease in drug cytotoxicity as measured by less than a 2-fold increase in clonogenic survival. When cells were heated for 10 min at 45.5 degrees C prior to or after treatment with the drug, there was a reduction (approximately 50%) immediately after treatment in the number of DSBs/100 megabase pairs compared with unheated cells. The rate of removal of cleavable complexes was decreased slightly by heat. After 120 min at 37 degrees C, the number of DSB/100 megabase pairs decreased to approximately 6 for both unheated cells and those heated prior to drug treatment and to approximately 8 for cells heated after drug treatment. In agreement with a low effect of heat on the number of cleavable complexes after drug treatment, there was no significant effect of this heating protocol on drug cytotoxicity. However, heating at 45.5 degrees C prior to drug treatment at 37 degrees C protected cells from drug cytotoxicity (e.g., increased survival after 12 micrograms/ml for 60 min by approximately 100-fold) despite the similarity in the induction and rate of removal of cleavable complexes when compared with nonheated cells. Thus, when cells are heated prior to administration of VP-16, drug cytotoxicity does not correlate with the number of cleavable complexes measured either immediately after treatment or 180 min later when approximately 75% of the initial number have been removed. Finally, since hyperthermia can actually decrease drug cytotoxicity, the use of hyperthermia as an adjuvant to chemotherapy involving topoisomerase II poisons, such as VP-16, should be approached with caution.


Subject(s)
CHO Cells/drug effects , DNA Damage , DNA Topoisomerases, Type II/drug effects , DNA, Neoplasm/drug effects , Etoposide/pharmacology , Hyperthermia, Induced , Animals , CHO Cells/pathology , Cell Survival/drug effects , Cricetinae , Dose-Response Relationship, Drug , Electrophoresis, Gel, Pulsed-Field , Temperature
16.
Cytometry ; 14(2): 223-9, 1993.
Article in English | MEDLINE | ID: mdl-8440156

ABSTRACT

We wished to determine if local anesthetics (LAs) induced changes in intracellular free calcium ([fCa2+i]) that could have an effect on cell killing by hyperthermia. Flow cytometry was used to measure [fCa2+i] of mouse NIH 3T3 cells during heating at 45.5 degrees C. In both non-tolerant and thermotolerant cells, heating caused a rapid increase (within 1 min) in [fCa2+i] of approximately 100 nM, which remained relatively constant during 25 min of continued heating; however, survival was higher in thermotolerant cells. Procaine, lidocaine, and tetracaine had no effect on survival or [fCa2+i] of cells kept at 37 degrees C up to 25 min. Cells heated with procaine and lidocaine showed no difference in [fCa2+i] compared to cells heated without LAs but were greatly sensitized to killing. Cells heated with tetracaine became permeable to trypan blue within 10-15 min of heating. We conclude that heat sensitization by LAs does not involve changes in [fCa2+i]. Furthermore, these studies reject the hypothesis that changes in [fCa2+i] are involved in heat-induced cell killing.


Subject(s)
Calcium/analysis , Hyperthermia, Induced , 3T3 Cells/chemistry , 3T3 Cells/drug effects , Animals , Flow Cytometry , Lidocaine/pharmacology , Mice , Procaine/pharmacology , Tetracaine/pharmacology
17.
Int J Hyperthermia ; 8(3): 351-62, 1992.
Article in English | MEDLINE | ID: mdl-1607740

ABSTRACT

Chinese hamster ovary (CHO) cells heated in the presence of the membrane-active agents procaine, ethanol, butylated hydroxytoluene (BHT) and glycerol were analysed for changes in fluorescence polarization of the lipid probe (1-[4(trimethylamino)phenyl]-6-phenyl-1,3,5-hexatriene, an indicator of plasma membrane fluidity). Cells were heated in normal and acid (pH 6.6) medium. Procaine, ethanol, BHT and low pH sensitized cells to heat-killing. Procaine, ethanol and BHT decreased fluorescence polarization (increased plasma membrane fluidity) significantly. Polarization distributions for cells heated with these sensitizers were broadened substantially and were skewed toward lower polarization values. Glycerol, a heat-protector, inhibited changes in fluorescence polarization due to heating at temperatures up to 45.0 degrees C. Heating cells at either 42 or 45 degrees C in pH 6.6 medium had no significant effect on the fluorescence polarization compared with controls, while survival was reduced substantially. Thus, heat-sensitization of low pH cannot be ascribed to changes in membrane fluidity. The changes in membrane fluidity caused by other sensitizers and protector indicate that membrane fluidity changes may be a contributing cause of cell killing by hyperthermia.


Subject(s)
Hot Temperature , Membrane Fluidity , Animals , Butylated Hydroxytoluene/pharmacology , CHO Cells/drug effects , Cell Membrane/drug effects , Cell Survival/drug effects , Cricetinae , Ethanol/pharmacology , Flow Cytometry , Fluorescence Polarization , Glycerol/pharmacology , Hydrogen-Ion Concentration , Membrane Fluidity/drug effects , Procaine/pharmacology
18.
Radiat Res ; 130(1): 48-54, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1561317

ABSTRACT

Flow cytometry was used to measure the fluorescence polarization of the lipid probe trimethylammonium-diphenylhexatriene as an indicator of plasma membrane fluidity of Chinese hamster ovary (CHO) cells heated under various conditions. Fluorescence polarization was measured at room temperature about 25 min after heating. When cells were heated for 45 min at temperatures above 42 degrees C, fluorescence polarization decreased progressively, signifying an increase in plasma membrane fluidity. The fluorescence polarization of cells heated at 42 degrees C for up to 55 h was nearly the same as for unheated control populations, despite a reduction in survival. The fluorescence polarization of cells heated at 45 degrees C decreased progressively with heating time, which indicated a progressive increase in membrane fluidity. The fluorescence polarization distributions broadened and skewed toward lower polarization values for long heating times at 45 degrees C. Thermotolerant cells resisted changes in plasma membrane fluidity when challenged with subsequent 45 degrees C exposures. Heated cells were sorted on the basis of their position in the fluorescence polarization distribution and plated to determine survival. The survival of cells which were subjected to various heat treatments and then sorted from high or low tails of the fluorescence polarization histograms was not significantly different. These results show that hyperthermia causes persistent changes in the membrane fluidity of CHO cells but that membrane fluidity is not directly correlated with cell survival.


Subject(s)
CHO Cells/physiology , Hot Temperature , Membrane Fluidity/physiology , Animals , Cell Survival , Cricetinae , Flow Cytometry , Fluorescence Polarization
19.
Radiat Res ; 130(1): 55-60, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1561318

ABSTRACT

The membrane fluidity of cells of human (AG1522 human foreskin fibroblasts), rodent [Chinese hamster ovary (CHO) and radiation-induced mouse fibrosarcoma], and feline (Crandall feline kidney) cell lines after heating at 45 degrees C was measured by flow cytometry. In addition, a heat-resistant variant of radiation-induced mouse fibrosarcoma cells and two heat-sensitive CHO strains were studied. Fluorescence polarization of the plasma membrane probe trimethylammonium-diphenylhexatriene was used as a measure of membrane fluidity. The sensitivity of all cell lines to 45 degrees C hyperthermia was compared. The baseline membrane fluidity varied among the cell lines, but did not correlate with sensitivity to hyperthermia. However, CHO cells, especially the heat-sensitive mutants, had the largest increase in membrane fluidity after heating at 45 degrees C, while the heat-resistant mouse fibrosarcoma variants and Crandall feline kidney cells resisted changes in fluidity. In general, the more resistant the cell line was to killing by heat, the more resistant it was to changes in membrane fluidity.


Subject(s)
Hot Temperature , Membrane Fluidity/physiology , Animals , Cell Line/physiology , Cell Survival , Flow Cytometry , Fluorescence Polarization , Humans
20.
Radiat Res ; 128(1): 14-8, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1924724

ABSTRACT

The local anesthetic procaine greatly sensitizes cells to hyperthermia. Though it is generally accepted that procaine is a membrane-active agent that increases membrane fluidity in cells, the mechanism by which it potentiates heat killing is unknown. In this paper we report changes in intracellular pH (pHi) of Chinese hamster ovary (CHO) cells heated at 42.0 or 45.0 degrees C in the presence of procaine. The pHi was measured with flow cytometry using the dye 1,4-diacetoxy-2,3-dicyanobenzene (ADB). Studies were carried out using cells grown at normal pH (7.3) or cells placed in low-pH (6.6) medium 4 h prior to and during heating (acute low-pH treatment). Low-pH-adapted cells (PHV2), which were obtained previously by continuous culture in pH 6.6 medium, were also used. Normal cells heated in the presence of procaine at pH 7.3 underwent a large decrease in pHi compared to cells heated without procaine. Procaine had little additional effect on the intracellular pH of cells in medium with a pH of 6.6 for 4 h before and during 30 min of heating. PHV2 cells exposed to chronic low-pH conditions were resistant to acidification when heated with or without procaine. The surviving fraction of cells heated with procaine was significantly lower under all pH conditions than that of cells heated without procaine. Cells heated at 42.0 degrees C with procaine also became greatly acidified and their survival was reduced. These data suggest that the reduction in pHi caused by procaine may be part of the mechanism of heat sensitization, but cannot account for it entirely. Furthermore, the degree of procaine sensitization and intracellular acidification is dependent on the extracellular pH, with a larger effect occurring at pH 7.3 than at pH 6.6.


Subject(s)
Hot Temperature , Procaine/pharmacology , Adaptation, Physiological/drug effects , Adaptation, Physiological/physiology , Animals , CHO Cells , Cell Death/drug effects , Cell Death/physiology , Cricetinae , Hydrogen-Ion Concentration
SELECTION OF CITATIONS
SEARCH DETAIL
...