Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Arch Virol ; 149(5): 891-901, 2004 May.
Article in English | MEDLINE | ID: mdl-15098105

ABSTRACT

Cellular factors have been indicated to be essential for the replication of Measles virus (MV), but the exact roles of these components are, however, not understood in detail. In this study, we investigated the role of actin and tubulin in productive MV infection by inducing disassembly of the microfilaments and microtubules. Vero cells were treated with latrunculin-A, which sequesters actin monomers, or nocodazole, which breaks the microtubules. The disruption of either of the structures efficiently inhibited the maturation of new infectious virus. Interestingly, virus spreading to neighboring cells still occurred by fusion and large syncytia typical for MV infection appeared. We also investigated a possible role for proteins of the ERM-family. Our results support an important role for actin filaments and microtubules for efficient MV replication.


Subject(s)
Actin Cytoskeleton/virology , Cytoskeletal Proteins/physiology , Measles virus/physiology , Microtubules/virology , Actin Cytoskeleton/drug effects , Actins/analysis , Actins/physiology , Animals , Bridged Bicyclo Compounds, Heterocyclic , Chlorocebus aethiops , Fluorescent Antibody Technique , Giant Cells/virology , Microtubules/drug effects , Nocodazole , Thiazoles , Thiazolidines , Tubulin/analysis , Tubulin/physiology , Vero Cells , Viral Proteins/analysis , Viral Proteins/biosynthesis , Virus Replication
2.
Acta Vet Scand ; 44(1-2): 43-52, 2003.
Article in English | MEDLINE | ID: mdl-14650543

ABSTRACT

In the autumn of 1995 the first outbreaks of enterohemorrhagic Escherichia coli O157:H7 including ca 100 human cases were reported in Sweden. From outbreaks in other countries it is known that cattle may carry these bacteria and in many cases is the source of infection. Therefore, the present study was performed to survey the Swedish bovine population for the presence of verotoxin-producing E. coli (VTEC) of serotype O157:H7. Individual faecal samples were collected at the 16 main Swedish abattoirs from April 1996 to August 1997. Of 3071 faecal samples, VTEC O157 were found in 37 samples indicating a prevalence of 1.2% (CI95% 0.8-1.6). All 37 isolates carried genes encoding for verotoxin (VTI and/or VT2), intimin, EHEC-haemolysin and flagellin H7 as determined by PCR. Another 3 strains were of serotype O157:H7 but did not produce verotoxins. The 37 VTEC O157:H7 strains were further characterised by phage typing and pulsed-field gel electrophoresis. The results clearly show that VTEC O157:H7 is established in the Swedish bovine population and indicate that the prevalence of cattle carrying VTEC O157:H7 is correlated to the overall geographical distribution of cattle in Sweden. Results of this study have formed the basis for specific measures recommended to Swedish cattle farmers, and furthermore, a permanent monitoring programme was launched for VTEC O157:H7 in Swedish cattle at slaughter.


Subject(s)
Bacteriophage Typing/veterinary , Escherichia coli O157/isolation & purification , Feces/microbiology , Abattoirs , Animals , Cattle , Electrophoresis, Gel, Pulsed-Field , Escherichia coli O157/classification , Escherichia coli O157/genetics , Prevalence , Sweden
3.
Nucl Med Biol ; 28(4): 451-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11395319

ABSTRACT

A series of biodistribution studies were conducted with the radiotracer, [(18)F]N-(4'-fluorobenzyl)-4-(3-bromophenyl)acetamide, [(18)F]1 in nude mice bearing tumor xenografts of the mouse mammary adenocarcinoma, line 66. This radiotracer has a high affinity for both sigma(1) and sigma(2) receptors. In vivo studies were also conducted in order to assess the effect of blocking sigma(1) receptors on tumor uptake and the tumor:background ratio of this radiotracer. The results of these studies revealed that blocking the sigma(1) receptor so that only the sigma(2) receptors are labeled in vivo, results in a higher tumor:background ratio with only a small reduction in the tumor uptake of the radiotracer relative to the no-carrier-added (i.e., nonselective) conditions. Comparative in vivo studies were also conducted with the anatomic and metabolic imaging agent, [(18)F]FDG, and a radiolabeled DNA precursor, [(125)I]IUdR. Both of these radiolabeled compounds represent classes of agents that have been proposed for imaging the proliferative status of solid tumors. The results of these studies indicated that a sigma(2)-selective imaging agent may be, 1) a better anatomic imaging agent for breast cancer than [(18)F]FDG, and 2) a better functional imaging agent than the radiolabeled DNA precursors, [(123/124)I]IUdR and [(11)C]thymidine, for measuring the proliferative status of breast tumors with PET and SPECT. However, additional studies will be needed to compare sigma(2)-selective imaging agents with [(18)F]FLT in order to determine which is the more appropriate imaging agent for measuring the proliferative status of breast tumors with PET.


Subject(s)
Acetamides/chemical synthesis , Neoplasms/diagnostic imaging , Radiopharmaceuticals , Receptors, sigma/metabolism , Adenocarcinoma/diagnostic imaging , Animals , Female , Fluorodeoxyglucose F18 , Idoxuridine , Mammary Neoplasms, Experimental/diagnostic imaging , Mice , Mice, Nude , Neoplasms/metabolism , Radionuclide Imaging , Receptors, sigma/antagonists & inhibitors , Tissue Distribution
4.
Radiat Res ; 153(5 Pt 1): 548-56, 2000 May.
Article in English | MEDLINE | ID: mdl-10790276

ABSTRACT

For several years, we have concentrated our efforts on validating the use of radiation-induced DNA strand breaks and DNA-protein crosslinks to assess the oxygenation status of tumors and normal tissues. We have demonstrated that (1) the oxygen dependence of strand break formation is identical to that of radiation-induced cell killing; (2) the oxygen dependence of DNA-protein crosslink formation is the mirror image of that of radiation-induced cell killing; and (3) the formation of these radiation-induced DNA lesions is predominantly dependent on the oxygen concentration near the DNA and is independent of the cell type, metabolic status, proliferative status, pH of the surrounding environment, and composition or properties of the proteins tightly associated with the DNA. In the present study, the hypoxic fraction of three experimental tumors was estimated using our assay of radiation-induced DNA damage. The average hypoxic fraction of a large number of tumors estimated with this assay of radiation-induced DNA damage for (1) WiDR human colorectal carcinoma xenografts (40.8 +/- 4.2%), (2) 66 mouse mammary adenocarcinoma tumors (41.8 +/- 3.1%), and (3) subcutaneous tumors grown from 9L rat brain tumor cells (95% CI =-8.2-4.2%) was not statistically different from that of a large number of tumors measured for each of these tumor models by the paired survival curve method (38.3 +/- 6. 3%, 28.9 +/- 5.5%, 95% CI = 2.2-4.4%, respectively). When the hypoxic fraction measured by the alkaline elution method on one half of an individual tumor was compared to that measured by the paired survival curve method on the other half of the same tumor, no statistical correlation was found for either 66 or WiDR tumors. Although this assay of radiation-induced DNA damage can be used effectively in the laboratory to answer a number of important questions about the oxygenation status of animal tumors and normal tissues, failure to reliably estimate the hypoxic fraction of individual tumors and technical considerations make it unlikely that the assay can be used in the clinic to estimate the hypoxic fraction of human tumors.


Subject(s)
Cell Hypoxia , DNA Damage , DNA, Neoplasm/radiation effects , DNA/radiation effects , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Calibration , Cell Survival , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Repair , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Rats
5.
Br J Cancer ; 82(6): 1223-32, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10735510

ABSTRACT

Over the past several years, our group has provided considerable evidence that the expression of sigma-2 (sigma2) receptors may serve as a biomarker of tumour cell proliferation. In these in vitro studies, sigma2 receptors were expressed 8-10 times more in proliferative (P) tumour cells than in quiescent (Q) tumour cells, and the extent and kinetics of their expression were independent of a number of biological, physiological and environmental factors often found in solid tumours. Moreover, the expression of sigma2 receptors followed both the population growth kinetics when Q-cells were recruited into the P-cell compartment and the proliferative status of human breast tumour cells treated with cytostatic concentrations of tamoxifen. However, these in vitro studies may or may not be indicative of what might occur in solid tumours. In the present study, the sigma2 receptor P:Q ratio was determined for the cells from subcutaneous 66 (diploid) and 67 (aneuploid) tumours grown in female nude mice. The sigma2 receptor P:Q ratio of the 66 tumours was 10.6 compared to the sigma2 receptor P:Q ratio of 9.5 measured for the 66 tissue culture model. The sigma2 receptor P:Q ratio of the 67 tumours was 4.5 compared to the sigma2 receptor P:Q ratio of approximately equal 8 measured for the 67 tissue culture model. The agreement between the solid tumour and tissue culture data indicates that: (1) the expression of sigma2 receptors may be a reliable biomarker of the proliferative status of solid tumours and (2) radioligands with both high affinity and high selectivity for sigma2 receptors may have the potential to non-invasively assess the proliferative status of human solid tumours using imaging techniques such as positron emission tomography or single-photon emission computerized tomography.


Subject(s)
Biomarkers, Tumor/analysis , Mammary Neoplasms, Animal/genetics , Receptors, sigma/genetics , Animals , Cell Division , Female , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/physiopathology , Mice , Neoplasm Invasiveness , Prognosis , Receptors, sigma/physiology , Tumor Cells, Cultured
6.
Br J Cancer ; 81(6): 925-33, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10576647

ABSTRACT

Recently, we demonstrated that sigma-2 receptors may have the potential to be a biomarker of tumour cell proliferation (Mach et al (1997) Cancer Res 57: 156-161). If sigma-2 receptors were a biomarker of tumour cell proliferation, they would be amenable to detection by non-invasive imaging procedures, thus eliminating many of the problems associated with the flow cytometric measures of tumour cell proliferation presently used in the clinic. To be a good biomarker of tumour cell proliferation, the expression of sigma-2 receptors must be essentially independent of many of the biological, physiological, and/or environmental properties that are found in solid tumours. In the investigation reported here, the mouse mammary adenocarcinoma lines, 66 (diploid) and 67 (aneuploid), 9L rat brain tumour cells, and MCF-7 human breast tumour cells were used to study the extent and kinetics of expression of sigma-2 receptors in proliferative (P) and quiescent (Q) tumour cells as a function of species, cell type, ploidy, pH, nutrient depletion, metabolic state, recruitment from the Q-cell compartment to the P-cell compartment, and treatment with tamoxifen. In these experiments, the expression of sigma-2 receptors solely reflected the proliferative status of the tumour cells. None of the biological, physiological, or environmental properties that were investigated had a measurable effect on the expression of sigma-2 receptors in these model systems. Consequently, these data suggest that the proliferative status of tumours and normal tissues can be non-invasively assessed using radiolabelled ligands that selectively bind sigma-2 receptors.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Mammary Neoplasms, Experimental/metabolism , Receptors, sigma/biosynthesis , Tamoxifen/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Biomarkers, Tumor/analysis , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Division , Female , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice , Ploidies , Rats , Receptors, sigma/analysis , Tumor Cells, Cultured
7.
Anticancer Res ; 15(4): 1333-8, 1995.
Article in English | MEDLINE | ID: mdl-7654017

ABSTRACT

BACKGROUND: The ether lipid analogue 1-octadecyl-2-methyl-rac-glycero-3-phosphocholine (ET-18-OCH3) is known to have plasma membrane interacting antitumor activity. However, the mechanism and the mode of action of its activity remain to be elucidated fully. MATERIALS AND METHODS: In this study, the proportion of the inhibitory effects resulting either from direct cell kill or from cytostasis induced by (ET-18-OCH3) alone or in combination with heat in BG-1 human ovarian carcinoma cells was quantitated. RESULTS: The magnitude of both cell kill and cytostasis in BG-1 cells was dependent on the concentration of ET-18-OCH3 and the duration of exposure. Cell kill induced by exposure to 2 or 8 microM ET-18-OCH3 for 1-14 days could be expressed as a simple exponential function of exposure dose (concentration x time). As assayed by colony formation, cell survival after continuous exposure to the drug was significantly reduced from that measured when the drug was removed and the cells were incubated for 14 days in drug-free medium. CONCLUSIONS: We conclude that ET-18-OCH3 induces substantial cytostasis as well as cell kill in BG-1 cells and that this cytostasis is directly proportional to the amount of cell kill induced. These data are important in determining appropriate treatment regimens using ET-18-OCH3, either alone or in combination with other anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Hot Temperature , Phospholipid Ethers/pharmacology , Cell Survival/drug effects , Female , Humans , Tumor Cells, Cultured
8.
Radiat Res ; 142(2): 163-8, 1995 May.
Article in English | MEDLINE | ID: mdl-7724730

ABSTRACT

Results from several laboratories, including ours, have suggested that measurements of radiation-induced DNA strand breaks and DNA-protein crosslinks (DPCs) may be used to estimate the hypoxic fraction or fractional hypoxic volume of tumors and normal tissues. This suggestion has been predicted on both published and unpublished information that (1) the oxygen dependence of the formation of strand breaks in irradiated mammalian cells is similar to the oxygen dependence of radiation-induced cell killing, and (2) the oxygen dependence of the formation of DPCs in irradiated mammalian cells is the mirror image of the oxygen dependence of radiation-induced cell killing. However, the published studies that attempted to determine the relationship between the oxygen dependence of the formation of strand breaks and the radiation sensitivity of mammalian cells were not performed at 37 degrees C, the exact oxygen concentrations were not always known, and the results were conflicting. In addition, most of the data on the oxygen dependence of the formation of DPCs are unpublished. Consequently, we have undertaken a comprehensive investigation of one cell line, 9L/Ro rat brain tumor cells, to determine if the shape of the oxygen dependence curve and the Km value for radiation-induced strand breaks and DPCs were similar when 9L cells were irradiated under both ideal gas-liquid equilibrium conditions at 4 degrees C and nonideal gas-liquid equilibrium conditions at 37 degrees C. At 4 degrees C under ideal gas-liquid equilibrium conditions, the Km for the formation of strand breaks was approximately 0.0045 mM, and the Km for radiation sensitivity was approximately 0.005 mM. A similar comparison for the formation of DPCs at 4 degrees C could not be made, because the efficiency of the formation of DPCs was much lower at 4 degrees C than at 37 degrees C. At 37 degrees C under nonideal gas-liquid equilibrium conditions, the apparent Km for the formation of strand breaks and radiation sensitivity was approximately 0.032 mM, and the Km for the formation of DPCs was approximately 0.02 mM. The data for strand breaks are in agreement with the published data of Chapman et al. (Int. J. Radiat. Biol. 26, 383-389, 1974), and the data for DPCs are in agreement with the unpublished data of Meyn (personal communication). These results support the suggestion that measurements of radiation-induced strand breaks and/or DPCs may be used to detect hypoxic cells and estimate the hypoxic fraction or fractional hypoxic volume of tumors and normal tissues.


Subject(s)
DNA Damage , DNA/radiation effects , Oxygen/pharmacology , Proteins/metabolism , Animals , DNA/metabolism , Oxygen Consumption , Rats , Tumor Cells, Cultured
9.
Cancer Chemother Pharmacol ; 34(6): 472-6, 1994.
Article in English | MEDLINE | ID: mdl-7923557

ABSTRACT

The mechanisms that govern the activity and the factors that control the anticancer activity of synthetic ether lipids have not been fully elucidated. In this study, three factors were studied in relationship to cell survival after treatment with 1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine (ET-18-OCH3): (1) exposure dose, (2) drug uptake, and (3) cell density and cell-cycle distribution. In BG-1 human ovarian carcinoma cells, cell survival was an exponential function of exposure dose and was dependent on drug concentration. Drug uptake was dependent on the concentration of ET-18-OCH3, whereas the reduction in cell survival was directly related to the uptake of drug only in the first decade of cell kill. When the quantity of cells per flask was tripled from 4 to 12 x 10(6) cells, ET-18-OCH3 failed to induce a G2 block. Furthermore, the cell kill induced by a 72-h exposure to 2 microM ET-18-OCH3 was decreased by a factor of 2 when the cell density increased. Therefore, exposure dose and cell density are important parameters in determining the cell kill induced by ET-18-OCH3.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Phospholipid Ethers/pharmacology , Cell Cycle , Female , Humans , Ovarian Neoplasms , Phospholipid Ethers/metabolism , Time Factors , Tumor Cells, Cultured
10.
Int J Radiat Oncol Biol Phys ; 22(4): 727-30, 1992.
Article in English | MEDLINE | ID: mdl-1531976

ABSTRACT

Enhanced cell kill has been observed when experimental tumors were treated with alkylating agents in combination with 2-nitroimidazoles (2-NI). In this study, modification of the cell kill induced by cyclophosphamide (CY) and an analog, ifosfamide (IFO), by two radiation sensitizers, misonidazole (MISO) and etanidazole (SR-2508), was measured. Three important parameters were determined: (a) the necessity for hypoxic reduction of the 2-NI to achieve an increase in tumor cell kill, (b) the optimal timing for administration of the alkylating agents and the 2-NI, and (c) the degree of enhancement of the CY- and IFO-induced cell kill. The subcutaneous (sc) 9L tumor model in male Fisher 344 rats was used in these experiments, and the endpoint measured was clonogenic cell survival 18-20 hr after treatment. Under hypoxic conditions, MISO potentiated both CY- and IFO-induced cell kill with a sensitizer enhancement ratio of approximately 1.3 and 1.5, respectively, at the 10(-3) survival level. This enhancement was seen when CY was administered simultaneously or 1.5 hr prior to MISO administration. A similar enhancement of CY-induced cell kill was measured under hypoxic conditions when SR-2508 was used. Enhanced IFO-induced cell kill was measured under hypoxic conditions only when the IFO was given 1 hr before MISO administration. No enhancement of the IFO-induced cell kill was observed when SR-2508 was used instead of MISO. Increased normal tissue damage (i.e., hemorrhagic cystitis) was observed when the MISO was administered along with CY or IFO. Four conclusions can be drawn from these data. Metabolism of the 2-NI by hypoxic cells is necessary for potentiation of CY- or IFO-induced cell kill. Only MISO can potentiate the cell kill induced by IFO. The timing of administration of the alkylating agents and the 2-NI is a critical determinant of the extent of the cell kill obtained. Cell kill induced by IFO appears to be enhanced by MISO to a greater extent than the cell kill induced by CY.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms, Experimental/drug therapy , Animals , Cell Hypoxia/drug effects , Cell Survival/drug effects , Cyclophosphamide/administration & dosage , Etanidazole , Ifosfamide/administration & dosage , Male , Misonidazole/administration & dosage , Neoplasm Transplantation , Nitroimidazoles/administration & dosage , Radiation-Sensitizing Agents/therapeutic use , Rats , Rats, Inbred F344
11.
Int J Radiat Oncol Biol Phys ; 23(5): 1009-19, 1992.
Article in English | MEDLINE | ID: mdl-1639634

ABSTRACT

The feasibility of using a 2-dimensional (2D) modeling approach for retrospectively describing complete temperature distributions in the midplane of a tumor during a clinical hyperthermia treatment was tested. An experimental treatment, using a 915-MHz waveguide applicator to heat a large melanoma in a dog, was modeled. Detailed measurements of temperatures were made during the treatment. The steady-state blood flow distribution at the midplane was imaged by positron emission tomography (PET), and these data were used to prescribe the modeled perfusion pattern. A 2D finite element method (FEM) was used to approximate the solution to Maxwell's Equations to obtain the specific absorption rate (SAR) distribution. The blood-flow estimates, assumed material properties, SAR distribution, and temperature boundary conditions were then used with the same mesh in a second FEM program to obtain a solution to the bioheat transfer equation. This latter routine was embedded in a state-and-parameter-estimation program that systematically varied selected parameters until the differences between computed and measured temperatures were minimized. Optimizations were performed independently for three subsets of the measured temperature data to assess the sensitivity of the predicted temperature field to the number of measurements. The calculated temperature distributions that resulted were similar to each other, and the predicted temperatures at the sensor points excluded from these optimizations were in reasonable agreement with the measurements. However, lack of unique blood flow values following optimization indicates that the methods of estimating blood flow will need to be improved or that there are problems with model mismatch. This work is a clinical case study of an evolving 2D system of thermal dosimetry which relies on both empirical and theoretical concepts. The methodology is being evaluated for its ability to generate prognostically significant descriptors of the treatment temperature field.


Subject(s)
Blood Circulation/physiology , Body Temperature/physiology , Hyperthermia, Induced/instrumentation , Melanoma, Experimental/therapy , Animals , Dogs , Feasibility Studies , Melanoma, Experimental/blood supply , Melanoma, Experimental/physiopathology , Tomography, Emission-Computed
12.
Int J Radiat Oncol Biol Phys ; 23(4): 831-9, 1992.
Article in English | MEDLINE | ID: mdl-1618675

ABSTRACT

Our previous studies demonstrated that metabolism of misonidazole (MISO) by hypoxic cells is required to potentiate the cytotoxicity of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) in sc 9L tumors. To determine the influence of administration schedules on this chemosensitization, tumors were either clamped to produce a reversible hypoxia or left unclamped. MISO (2.5 mmoles kg-1) was administered to rats with unclamped tumors simultaneously with BCNU (9 or 12 mg kg-1), 20 min before BCNU, or 2.5 hr before BCNU, and the drug pharmacokinetics and BCNU cytotoxicity were measured. MISO administered 20 min or 2.5 hr before BCNU increased the plasma elimination half-time (t1/2) of BCNU, but MISO administered simultaneously with BCNU did not change the plasma elimination t1/2 of BCNU. In unclamped sc 9L tumors, all administration schedules decreased the peak BCNU concentration and increased the initial BCNU elimination t1/2; however, the BCNU exposure dose (AUC0-infinity) calculated from these data did not change significantly. In agreement with the AUC calculations, none of the administration schedules altered the BCNU cytotoxicity in unclamped tumors. If the tumors were clamped for 5-120 min after the peak MISO concentration was reached, BCNU-induced cell kill was increased by a constant factor of 3 over the first hour of the clamping period and by an additional factor of 7 over the second hour of the clamping period. If the tumors were clamped for 2 hr after the peak MISO concentration was reached and then BCNU administered 0-60 min after the clamp was released, this chemosensitization remained at a constant factor of approximately 20 for the first 10 min, and then decreased rapidly to a factor of approximately 3 by 20 min after the clamp was released. These data indicate that in sc 9L tumors, (1) at least two biochemical mechanisms are involved in this MISO-BCNU interaction, one of which depends on the duration and extent of the metabolism of MISO by hypoxic cells, and (2) reoxygenation does not immediately eliminate the potentiation of BCNU by MISO. These data also suggest that MISO should be given 2-4 hr before BCNU to achieve the maximum chemosensitization in clinical trials.


Subject(s)
Carmustine/administration & dosage , Misonidazole/administration & dosage , Neoplasms, Experimental/drug therapy , Animals , Carmustine/blood , Drug Synergism , Male , Misonidazole/blood , Neoplasm Transplantation , Neoplasms, Experimental/mortality , Rats , Rats, Inbred F344 , Survival Rate
13.
Radiat Environ Biophys ; 31(2): 101-15, 1992.
Article in English | MEDLINE | ID: mdl-1609056

ABSTRACT

Induction of unrepairable DNA damage, accumulation of misrepaired DNA damage, and generation of imbalances in competing biochemical and/or metabolic processes have been proposed to explain the relationship between radiation-induced DNA damage and cell lethality. Theoretically, the temperature dependence of the critical DNA repair process(es) should be 1) either independent of or identical to the temperature dependence of cell killing if the first two hypotheses are correct, and 2) different if the third hypothesis is correct. To test this, exponentially growing rat 9L brain tumor cells were left at 37 degrees C or equilibrated for 3-14 h at 20 degrees C before irradiation. Cells were irradiated and allowed to repair at either 20 degrees C or 37 degrees C. Alternatively, the cells were irradiated at one of these temperatures and immediately shifted to the other temperature for repair. DNA damage was assessed by the alkaline elution technique; cell kill was assessed by a clonogenic assay. 9L cells maintained at 20 degrees C or 37 degrees C sustained the same amount of DNA damage as measured by alkaline elution. DNA repair instantaneously assumed the rate characteristic of the postirradiation temperature. For 9L cells equilibrated, irradiated, and repaired at 20 degrees C, the half-time of the fast phase of the DNA repair decreased by a factor of approximately 2 and the half-time of the slow phase decreased by a factor of approximately 5 over that measured in cells incubated, irradiated and repaired at 37 degrees C. Although the rate of DNA repair decreased substantially at 20 degrees C, the survival of 9L cells that were equilibrated and irradiated at 20 degrees C was greater (p less than 10(-4)) than those incubated and irradiated at 37 degrees C, when assayed by an immediate plating protocol. In addition, the survival of 9L cells equilibrated and irradiated at 20 degrees C and then shifted to 37 degrees C immediately after irradiation was greater (p less than 10(-2)) than that obtained with any other delayed plating protocol. Thus, the temperature dependence of the DNA repair processes measured by alkaline elution was different from the temperature dependence of cell killing measured either by an immediate or delayed plating protocol. These data support the hypothesis that many irradiated 9L tumor cells die because of imbalances in sets of competing biochemical and/or metabolic processes.


Subject(s)
DNA Damage/physiology , DNA Repair/physiology , Animals , Cell Death/genetics , Cell Death/radiation effects , Cell Survival/radiation effects , Colony-Forming Units Assay , Gamma Rays , Temperature , Tumor Cells, Cultured
14.
Anal Biochem ; 198(1): 68-74, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1789433

ABSTRACT

The structure and composition of the nuclear matrices prepared from a mouse mammary adenocarcinoma cell (line 66) by digestion with DNase I and several proteases (PRT-matrices) were characterized by protein and DNA gel electrophoresis, flow cytometry, and scanning electron microscopy. The characteristics of these PRT-matrices were compared with the characteristics of conventionally prepared nuclear matrices that employ a high salt extraction step (HS-matrices) in order to select a preparation that can be used in biochemical and/or biophysical studies where salt extraction compromises either the analysis or the interpretation of the data. Of the characterized PRT-matrices, only those prepared with Type XIV protease (pronase) had most of the characteristics of HS-matrices. They, (i) maintained their structural integrity, (ii) had less than or equal to 5% of their nuclear DNA associated with the matrix, (iii) had no evidence of higher-order chromatin structure, and (iv) had a DNA size distribution in the range of 400-1100 bp. The major difference between the PRT-matrices and the HS-matrices was a decrease in the protein content of the PRT-matrices. Although the PRT-matrices may not be appropriate for studying the unique nuclear matrix associated proteins that are involved in functions such as replication, transcription, and differentiation, they are clearly suitable for studying the properties of the nuclear matrix associated DNA.


Subject(s)
Nuclear Matrix/chemistry , Adenocarcinoma , Animals , Cell Line, Transformed , DNA/analysis , Deoxyribonuclease I/metabolism , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Mammary Glands, Animal , Mice , Microscopy, Electron, Scanning , Nuclear Proteins/analysis , Peptide Hydrolases/metabolism , Sodium Chloride
15.
J Neurooncol ; 11(1): 17-25, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1833513

ABSTRACT

To determine if the 2-nitroimidazole (2-NI) and the nitrosourea (NU) in a brain tumor chemopotentiation trial should be selected on the basis of known structure-activity relationships (electron affinity, lipophilicity, alkylating activity, carbamoylating activity), s.c. implants of rat 9L brain tumor cells were treated with combinations of misonidazole (MISO) or etanidazole (SR-2508) administered under oxic and hypoxic conditions, and BCNU, CCNU or chlorozotocin (CLZ) administered under oxic conditions. Cell kill was assessed by an in vivo to in vitro colony formation assay. To mimic the 'preincubation effect', the 2-NI was injected i.p., and 30 min later the tumor was clamped. After 2 hr, the clamp was released, and the NU administered immediately. MISO (2.5 mmole/kg) and SR-2508 (3.75 mmole/kg) reached the same peak tumor concentration in 30 min. Both 2-NIs were metabolized at the same rate in the clamped tumors; however, metabolism of the 2-NIs by hypoxic cells over the 2 hr clamping period did not produce any measurable s.c. 9L cell kill. The relative effectiveness of the NUs for killing oxic s.c. 9L tumor cells was: BCNU greater than CCNU greater than CLZ. Clamping the tumor prior to NU administration did not change the NU cytotoxicity. No potentiation of the NU cytotoxicity by the 2-NIs was observed in oxic tumors. Although metabolism of MISO by hypoxic cells did not result in potentiation of CLZ cytotoxicity at any dose, it resulted in potentiation of BCNU cytotoxicity at all doses and CCNU cytotoxicity at high doses.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Carmustine/therapeutic use , Lomustine/therapeutic use , Misonidazole/therapeutic use , Nitroimidazoles/therapeutic use , Streptozocin/analogs & derivatives , Animals , Carmustine/administration & dosage , Carmustine/pharmacology , Cell Line , Cell Survival/drug effects , Drug Synergism , Etanidazole , Lomustine/administration & dosage , Lomustine/pharmacology , Male , Misonidazole/administration & dosage , Misonidazole/pharmacology , Nitroimidazoles/administration & dosage , Nitroimidazoles/pharmacology , Rats , Rats, Inbred F344 , Streptozocin/pharmacology , Streptozocin/therapeutic use , Tumor Stem Cell Assay
16.
Br J Cancer ; 63(4): 484-8, 1991 Apr.
Article in English | MEDLINE | ID: mdl-2021530

ABSTRACT

The acute toxicity, pharmacokinetics and hypoxic cytotoxicity of RSU-1069 were investigated using the subcutaneous (sc) rat 9L tumour model. The pharmacokinetics were studied after i.p. injection of RSU-1069 (20 mg kg-1 or 100 mg kg-1). For both doses, the elimination of RSU-1069 followed first-order kinetics in both plasma and unclamped tumours. After 100 mg kg-1, the peak plasma concentration of RSU-1069 was 40 micrograms ml-1; the elimination t1/2 was 39.3 +/- 11.1 min. After 20 mg kg-1, the peak plasma concentration was 3 micrograms ml-1; the elimination t1/2 was 47.8 +/- 6.3 min. In unclamped tumours, the peak concentration was 50 micrograms g-1 with an elimination t1/2 of 36.1 +/- 9.6 min for the 100 mg kg-1 dose, and 4 micrograms g-1 with an elimination t1/2 of 41.9 +/- 6.1 min for the 20 mg kg-1 dose. The tumour and plasma elimination half-times were not significantly different (P greater than 0.2) for the two doses. Clamping the tumour 30 min after administration of 100 mg kg-1 of RSU-1069 decreased the tumour elimination t1/2 to 10.9 +/- 1.4 min. After releasing the clamp, RSU-1069 returned rapidly to the unclamped tumour concentration. The unclamped tumour/plasma ratio reached a maximum of 4-6, then decreased to a constant value of about 2 for both doses, indicating that RSU-1069 accumulates in these 9L tumours. RSU-1069 kills hypoxic sc 9L cells more efficiently than oxic sc 9L cells; at a surviving fraction of 0.5, the SER was 4.8. For in vitro 9L cells, the SER was approximately 50 when the comparison was between those treated in 2.1% 0(2) and those treated in less than 7.5 x 10(-3)% 0(2); it was approximately 100 when the comparison was between those treated in 21% 0(2) and those treated in less than 7.5 x 10(-3)% 0(3). Tumours treated with RSU-1069 and clamped for various times exhibited biphasic cell-kill kinetics; at 50 mg kg-1, little additional cell kill was achieved after 40 min of clamping. Our data also indicate that RSU-1069 is 300-1000 fold more efficient than misonidazole or SR2508 for killing hypoxic sc 9L tumour cells in situ.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cell Hypoxia/physiology , Misonidazole/analogs & derivatives , Neoplasms, Experimental/metabolism , Oxygen/physiology , Animals , Antineoplastic Agents/therapeutic use , Injections, Subcutaneous , Male , Misonidazole/pharmacokinetics , Misonidazole/therapeutic use , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Rats , Rats, Inbred F344 , Tumor Stem Cell Assay
17.
J Med Chem ; 34(4): 1408-14, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1901911

ABSTRACT

Combinations of an amidoalkylphosphocholine, 8, and AZT have been found to cause an apparent synergistic action in suppressing infectious HIV-1 replication. In addition, amidoalkyl, oxyalkyl, and thioalkyl ether lipids have been chemically linked to anti-HIV-1 nucleosides (AZT and DDI) through phosphate and phosphonate linkages. These conjugates have shown promising in vitro anti-HIV-1 activity. Also, the conjugates have a 5-10-fold reduction in cell cytotoxicity compared to AZT alone. The most active compound, an amidoalkyl ether lipid-AZT conjugates, 4A, was found to have a differential selectivity of 1793 in a syncytial plaque assay. In comparison, AZT alone has a value of 1281.


Subject(s)
Antiviral Agents/chemical synthesis , Didanosine/analogs & derivatives , Didanosine/chemical synthesis , HIV-1/drug effects , Phospholipid Ethers/chemical synthesis , Zidovudine/analogs & derivatives , Zidovudine/chemical synthesis , Cell Line , Didanosine/chemistry , Didanosine/pharmacology , Dideoxynucleotides , Ethers , HIV-1/physiology , Humans , Indicators and Reagents , Molecular Structure , Phospholipid Ethers/chemistry , Phospholipid Ethers/pharmacology , Structure-Activity Relationship , Virus Replication/drug effects , Zidovudine/chemistry , Zidovudine/pharmacology
18.
J Urol ; 144(1): 159-63, 1990 Jul.
Article in English | MEDLINE | ID: mdl-2359170

ABSTRACT

The effects of focused high energy shock waves (SW) generated by the Dornier experimental XL-1 lithotripter alone or in combination with cisplatin (CDDP) on the AT-2 prostate tumor were examined. They were assessed by measuring 1) clonogenic cell survival 24 hours after treatment, 2) tumor growth delay, and 3) the number of lung metastases. The survival of clonogenic cells was reduced 38% by exposure to 2000 SW and tumor growth was delayed by 1.5 days. The limb bearing the tumor was excised in all animals when the tumor reached seven cm.3 The subsequent occurrence of lung metastases in three out of 13 unshocked animals and two out of 14 shocked animals indicated that SW did not promote the dissemination of tumor cells. At doses of one to four mg. CDDP/kg. body weight, SW exposure increased the effectiveness of the chemotherapy measured by a clonogenic assay. The fractional survival was 0.62 after 2000 SW alone, 0.23 with four mg. cisplatin/kg. alone and 0.10 after the combination treatment. At higher doses of CDDP, there was no added effect of SW over that of CDDP therapy alone. Tumor growth to one cm.3 was delayed by seven days after treatment with four mg. CDDP/kg., in comparison to untreated controls. SW exposure combined with CDDP treatment delayed the time taken for the tumor to reach one cm.3 by 13 days in comparison with untreated controls. However, the combination treatment increased animal mortality from 9% with CDDP alone to 29%. These results indicate that SW could be used focally to enhance the cell killing effects of CDDP.


Subject(s)
Cisplatin/therapeutic use , Lithotripsy , Prostatic Neoplasms/therapy , Animals , Combined Modality Therapy , Male , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Rats , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology
19.
Int J Radiat Oncol Biol Phys ; 18(5): 1043-50, 1990 May.
Article in English | MEDLINE | ID: mdl-2140824

ABSTRACT

Alterations of the pharmacokinetics and cytotoxic effects of the nitrosoureas, 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) and 1-(2-chloroethyl)-3-(cyclohexyl)-1-nitrosourea (CCNU) by the 2-nitroimidazoles, misonidazole (MISO) and SR-2508 were investigated using the subcutaneous (sc) 9L tumor model in male Fisher 344 rats. When 50 mg/kg of CCNU was given i.p., the peak plasma concentration of CCNU was about 3 micrograms/ml. CCNU was eliminated with biphasic kinetics that had a terminal half-time (T1/2) of approximately 47 min. When 2.5 mmole/kg of MISO was given i.p. 150 min before CCNU, the peak plasma concentration of CCNU was increased by approximately 63% with no change in the elimination kinetics. Clamping did not change the pharmacokinetics of CCNU in either plasma or tumors. MISO pretreatment increased the peak CCNU concentration in unclamped tumors by 3-fold, but had no effect on the CCNU pharmacokinetics in clamped tumors. With the exception of a decrease in the peak BCNU concentration in tumors similar to that observed with MISO, SR-2508 (3.75 mmole/kg, i.p.) did not change the pharmacokinetics of BCNU or CCNU in plasma and tumors. CCNU had no effect on the MISO concentration in plasma and unclamped tumors. However, in the clamped tumors, CCNU delayed the return of the MISO concentration to the unclamped tumor level by about an additional 60 min after the clamp was released. SR-2508 was eliminated from the plasma with biphasic kinetics having an initial and terminal T1/2 of approximately 11 and approximately 76 min, respectively. SR-2508 reached a peak tumor concentration of about 500 micrograms/ml in 30 min. The elimination T1/2 for SR-2508 in unclamped and clamped tumors was approximately 81 and approximately 42 min, respectively. When the clamp was released, the SR-2508 concentration returned to the level found in the unclamped tumors approximately 90 min after it reached its nadir; BCNU and CCNU had no effect on the kinetics of this process. MISO significantly potentiated the cytotoxicity of BCNU in clamped tumors at surviving fractions less than or equal to 0.5. MISO did not potentiate the cytotoxicity of CCNU until the surviving fraction reached 0.05. SR-2508 did not potentiate the cytotoxicity of either BCNU or CCNU.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Neoplasms, Experimental/pathology , Nitroimidazoles/pharmacology , Nitrosourea Compounds/pharmacokinetics , Animals , Carmustine/pharmacokinetics , Carmustine/pharmacology , Drug Synergism , Etanidazole , Lomustine/pharmacokinetics , Lomustine/pharmacology , Male , Misonidazole/pharmacology , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Nitroimidazoles/pharmacokinetics , Nitrosourea Compounds/pharmacology , Rats , Rats, Inbred F344 , Structure-Activity Relationship
20.
Radiat Environ Biophys ; 29(2): 93-102, 1990.
Article in English | MEDLINE | ID: mdl-2339199

ABSTRACT

The radiosensitivities and the kinetics for removal of radiation-induced DNA damage were compared for proliferative (P) and quiescent (Q) cells of the lines 66 and 67 derived from a mouse mammary adenocarcinoma. As determined from cell survival assays, the 66 and 67 Q cells were more radiosensitive than their 66 and 67 P counterparts. The rank order of their radiosensitivity was: 67 Q greater than 66 Q greater than or equal to 67 P greater than 66 P. Induction of radiation damage in the DNA of these cells, as measured by the alkaline elution technique, was identical for 66 and 67 P and Q cells. The repair of this DNA damage was biphasic for 66 and 67 P and Q cells. The half-times for the fast and slow repair phases in 66 Q cells were identical to those previously measured in 67 Q cells. The half-times of the fast and slow repair phases in 66 P cells were also identical to those previously measured in 67 P cells. However, the half-times for the fast and slow repair phases in 66 and 67 Q cells were longer than those measured in their 66 and 67 P counterparts. The 66 cell data are consistent with our previously published hypothesis that Q cells are more radiosensitive than their corresponding P cells because they repair their radiation-induced DNA damage slower. However, our results are not consistent with hypotheses that attempt to explain the radiosensitivity differences between lines 66 and 67 solely on the basis of measurable induction and repair of DNA damage.


Subject(s)
Cell Survival/radiation effects , DNA Damage , DNA Repair , Animals , Mammary Neoplasms, Experimental/pathology , Mice , Radiation Tolerance , Tumor Cells, Cultured/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...