首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Within 3-6 h of small doses of radiation (gamma-rays) the number of dead cells (apoptotic cells) in the crypts of the small intestine reaches peak values. These return to normal levels only after times later than 1 day. After higher doses elevated levels of cell death persist for longer times. The dead cells first occur most frequently at the lower positions of the crypt (median value for the distribution of apoptotic fragments is about cell position 6). At later times more dead cells are observed at higher positions. Two doses of radiation separated by various time intervals have been used to investigate when after irradiation the cell population susceptible to acute cell death is re-established. Dead cells were scored 3 or 6 h after the second dose. The yield of dead cells after two doses represents the sum of the dead cells produced by, and persisting from, the first dose and new apoptotic cells induced by the second dose. Since the temporal and dose-dependence aspects of the dead-cell yield after the first dose alone is known, the additional dead cells attributable to the second dose alone can be determined by subtraction. Within 1-2 days of small doses (0.5 Gy) the sensitive cells, recognized histologically as apoptotic cells, are re-established at the base of the crypt (around cell position 6). After higher doses (9.0 Gy) they are not re-established until about the fourth day after irradiation. Even in the enlarged regenerating crypts the sensitive cells are found at the same position at the crypt base. It has been estimated that the crypt contains five or six cells that are susceptible to low doses (0.5 Gy) (hypersensitive cells) and up to a total of only seven or eight susceptible cells that can be induced by any dose to enter the sequence of changes implicit in apoptosis. Between 4 and 10 days after an initial irradiation of 9.0 Gy the total number of susceptible cells increased from seven to eight to about 10 to 13 per crypt.  相似文献   

2.
The aim of the present study was to analyse the dose rate effect of gamma radiation at the level of mutations, chromosomal aberrations, and cell growth in TK6 cells with normal as well as reduced levels of hMTH1 protein. TK6 cells were exposed to gamma radiation at dose rates ranging from 1.4 to 30.0 mGy/h (chronic exposure) as well as 24 Gy/h (acute exposure). Cell growth, frequency of thymidine kinase mutants, and of chromosomal aberrations in painted chromosomes 2, 8, and 14 were analysed. A decline in cell growth and an increase in unstable-type chromosomal aberrations with increasing dose rate were observed in both cell lines. A dose rate effect was not seen on mutations or stable-type chromosomal aberrations in any of the two cell lines. Reduction in the hMTH1 protein does not influence the sensitivity of TK6 cells to gamma radiation. This result fits well with data of others generated with the same cell line.  相似文献   

3.
Irradiation time and dose rate are important factors in the evaluation of radiation risk for human health. We previously proposed a novel dose-rate effect model, the modified exponential (MOE) model, which predicts that radiation risks decline exponentially as the dose rate decreases. Here we show that, during the early phase of exposure, up to 1000 h, the proliferation of cells continuously exposed to γ rays at a constant dose rate is gradually suppressed, even as the total dose increases. This trend holds for a number of cell lines including tumor cells, nontransformed fibroblasts and leukocytes. The accumulation of total dose by longer exposure times does not increase this suppressive effect even in cells with a defective DNA repair system, suggesting that risk is determined solely by dose rate in the later phase. The dose-rate effect in the early phase follows the MOE model in DNA repair-proficient cell lines, while cells with impaired DNA-PK or ATM show no dose-rate effect. In the later phase, however, a certain dose-rate effect is observed even in mutant cell lines, and suppression of cell proliferation no longer follows the MOE model. Our results suggest that a distinct mechanism that can operate in the absence of intact DNA-PK or ATM influences the dose-rate effect in the later phase of continuous radiation exposure.  相似文献   

4.
Cultures of radioresistant (LY-R) and radiosensitive (LY-S) strains of L5178Y mouse lymphoma cells were exposed continuously to X-rays delivered at dose rates ranging from 0.003 to 0.025 Gy/h for up to 35 days. Populations of both strains proliferated actively during the exposure, but the growth rates were reduced in a dose rate-dependent manner. The reduction of growth rate occurred for strain LY-S earlier during the exposure and at lower dose rates than for strain LY-R. The survival (as measured by colony forming ability) of strain LY-R was affected only slightly at all dose rates applied. For strain LY-S, a decrease in the surviving fraction was observed in the initial part of the exposure. This decrease was followed by a plateau and eventually by an increase, in some cases to values close to the control level. The increase in the surviving fraction indicated that the radioresistance of the exposed LY-S cells had increased. This pattern was particularly clear for dose rates greater than 0.014 Gy/h. The pre-irradiated cells exhibited radioresistance when exposed to acute X-radiation after termination of the chronic exposure. The increase in radiation resistance was stable for at least 70 days after termination of the protracted exposure. These results show that mutagenic and/or selective phenomena leading to an increase in radiation resistance of mammalian cells can be caused by protracted exposures to X-rays at dose rates permitting active proliferation.  相似文献   

5.
Different types of ionizing radiation produce different dependences of cancer risk on radiation dose/dose rate. Sparsely ionizing radiation (e.g. γ-rays) generally produces linear or upwardly curving dose responses at low doses, and the risk decreases when the dose rate is reduced (direct dose rate effect). Densely ionizing radiation (e.g. neutrons) often produces downwardly curving dose responses, where the risk initially grows with dose, but eventually stabilizes or decreases. When the dose rate is reduced, the risk increases (inverse dose rate effect). These qualitative differences suggest qualitative differences in carcinogenesis mechanisms. We hypothesize that the dominant mechanism for induction of many solid cancers by sparsely ionizing radiation is initiation of stem cells to a pre-malignant state, but for densely ionizing radiation the dominant mechanism is radiation-bystander-effect mediated promotion of already pre-malignant cell clone growth. Here we present a mathematical model based on these assumptions and test it using data on the incidence of dysplastic growths and tumors in the mammary glands of mice exposed to high or low dose rates of γ-rays and neutrons, either with or without pre-treatment with the chemical carcinogen 7,12-dimethylbenz-alpha-anthracene (DMBA). The model provides a mechanistic and quantitative explanation which is consistent with the data and may provide useful insight into human carcinogenesis.  相似文献   

6.
Friend erythroleukemia cells require high doses (15 Gy) of ionizing radiation to display a reduced rate of proliferation and an increased number of dead cells. Since ionizing radiation can activate several signaling pathways at the plasma membrane which can lead to the nuclear translocation of a number of proteins, we looked at the intranuclear signaling system activated by Protein Kinases C, being this family of enzymes involved in the regulation of cell growth and death. Our results show an early and dose-dependent increased activity of zeta and epsilon isoforms, although PKC zeta is the only isoform significantly active and translocated into the nuclear compartment upon low (1.5 Gy) and high (15 Gy) radiation doses. These observations are concomitant and consistent with an increase in the anti-apoptotic protein Bcl-2 level upon both radiation doses. Our results point at the involvement of the PKC pathway in the survival response to ionizing radiation of this peculiar cell line, offering PKC zeta for consideration as a possible target of pharmacological treatments aimed at amplifying the effect of such a genotoxic agent.  相似文献   

7.
The lethal and mutagenic effects of ionizing radiation delivered at high (53 Gy/h) and low (0.02 Gy/h) dose rates were measured in two closely related strains of mouse lymphoma L5178Y cells differing in radiation sensitivity (LY-R and LY-S). Strain LY-R was more resistant to the lethal effects of radiation than strain LY-S when exposed at either the high or low dose rate. The survival of strain LY-R was markedly enhanced by the reduction in dose rate. The dose-rate dependence of the survival of strain LY-S was less clear, because of the biphasic nature of its survival curve following low dose-rate radiation. However, if the initial slope of the low dose-rate survival curve is compared to the slope of the high dose-rate survival curve for strain LY-S, only a slight increase in survival at the low dose rate is apparent. Although more sensitive to the lethal effects of radiation, strain LY-S was less mutable at the hypoxanthine/guanine phosphoribosyl transferase locus by both low dose-rate and high dose-rate radiation than strain LY-R. Little dose-rate dependence was exhibited by either strain with regard to the mutagenic effects of radiation. Thus, for strain LY-R, which showed marked dose-rate dependence for survival but not for mutation, the ratio of mutational to lethal lesions was much greater following exposure to low dose-rate than to high dose-rate radiation.  相似文献   

8.
Apoptosis in HeLa Hep2 cells is induced by low-dose,low-dose-rate radiation   总被引:5,自引:0,他引:5  
Radioimmunotherapy with radiolabeled antibodies may cause inhibition of the growth of epithelial tumors, despite low total radiation doses and comparatively low radiosensitivity of epithelial tumor cells. The induction of apoptosis by low-dose radiation, such as delivered in radioimmunotherapy, was investigated in vitro in human HeLa Hep2 carcinoma cells. The cultured cells were exposed to defined radiation doses from a (60)Co radiation therapy source. The radiation source delivered 0.80 +/- 0.032 (mean +/- SD) Gy/min and the cells were given total doses of 1, 2, 5, 10 and 15 Gy. Using fluorescein-labeled Annexin V, followed by flow cytometry and DNA ladder analysis, apoptotic cells were detected and quantified. Radiation doses below 2 Gy did not cause any significant increase in apoptosis. Compared to control cells, apoptosis was pronounced after 5-10 Gy irradiation and was correlated to the radiation dose, with up to 42 +/- 3.5% of the cells examined displaying apoptosis. Clonogenic assays confirmed significantly decreased viability of the cells in the interval 2 to 10 Gy with low-dose-rate radiation, 60 +/- 2% compared to 2 +/- 2%. Lethal effects on the tumor cells were also evaluated by an assay of the cytotoxic effects of the release of (51)Cr. Significant cytotoxicity, with up to 64 +/- 6% dead cells, was observed at 5 Gy. Similar results were obtained when the dose rate was reduced to 0.072 +/- 0.003 Gy/min (mean +/- SD). In the case of the (137)Cs source, the dose rate could be reduced to 0.045 Gy/h, a level comparable to radioimmunotherapy, which induced significant apoptosis, and was most pronounced at 72-168 h postirradiation. It can be concluded that in vitro low-dose and low-dose-rate radiation induces apoptosis in epithelial HeLa Hep2 cells and thus may explain a mechanism by which pronounced inhibition of growth of HeLa Hep2 tumors at doses used in radioimmunotherapy has been obtained previously.  相似文献   

9.
Whether natural radiation can be a conditioning factor for the growth and survival of a living organism was investigated using diploid yeast S. cerevisiae D7. Yeast cells were conditioned by growing them continuously for at least 100 generation in 3 different radiation background such as i) ambient radiation (1.1 mSv/y), ii) sub-ambient radiation (0.44 mSv/y, within a shielded chamber) and iii) an elevated background radiation (88 and 880 mSv/y in a gamma-field). At the end, the cells were challenged with 60Co gamma, 100 Gy and the viable fractions were determined. Conditioning the cells in 880 mSv/y and in ambient radiation, enabled the cells to reduce the deleterious effect of the challenging dose significantly (P < 0.05) compared to that of sub-ambient radiation. The cellular viability of yeast cultures seems to be influenced by the prevailing radiation background, apart from starvation. Comparatively, a rapid decline in viability was noticed when the cultures were incubated for 60 days in the shielded chamber. The results indicate that some amount of radiation equivalent to background level or little above is needed to confer fitness in biological systems against stress factors, including radiation. The adaptive dose for the diploid yeast was also determined by single exposure. The priming dose ranged from 0.01 to 1.2 Gy. An adaptive dose of 0.25 or 0.4 Gy, almost nullified the deleterious effect of the challenging dose. The adaptive response may have a greater role in the field of cancer therapy and in radiation risk assessment. Understanding the response of an organism at different radiation-background will be helpful for successful space management.  相似文献   

10.
The dose response for adaption to radiation at low doses was compared in normal human fibroblasts (AG1522) exposed to either (60)Co gamma rays or (3)H beta particles. Cells were grown in culture to confluence and exposed at either 37 degrees C or 0 degrees C to (3)H beta-particle or (60)Co gamma-ray adapting doses ranging from 0.1 mGy to 500 mGy. These cells, and unexposed control cells, were allowed to adapt during a fixed 3-h, 37 degrees C incubation prior to a 4-Gy challenge dose of (60)Co gamma rays. Adaption was assessed by measuring micronucleus frequency in cytokinesis-blocked, binucleate cells. No adaption was detected in cells exposed to (60)Co gamma radiation at 37 degrees C after a dose of 0.1 mGy given at a low dose rate or to 500 mGy given at a high dose rate. However, low-dose-rate exposure (1-3 mGy/min) to any dose between 1 and 500 mGy from either radiation, delivered at either temperature, caused cells to adapt and reduced the micronucleus frequency that resulted from the subsequent 4-Gy exposure. Within this dose range, the magnitude of the reduction was the same, regardless of the dose or radiation type. These results demonstrate that doses as low as (on average) about one track per cell (1 mGy) produce the same maximum adaptive response as do doses that deposit many tracks per cell, and that the two radiations were not different in this regard. Exposure at a temperature where metabolic processes, including DNA repair, were inactive (0 degrees C) did not alter the result, indicating that the adaptive response is not sensitive to changes in the accumulation of DNA damage within this range. The results also show that the RBE for low doses of tritium beta-particle radiation is 1, using adaption as the end point.  相似文献   

11.
12.
There are various types of radiation in space including high energy particles. It is, therefore, becoming to be important to study the low dose and low dose-rate effects in space radiation biology. Radiation adaptive response (RAR) for cell growth and its mechanism were examined using cultured glial cells. The cells from hippocampus of Wistar rats were irradiated with a low dose (0.1 Gy) of X-rays and 3 h after with a high dose (2 Gy). Decrease in the rate of cell growth with 2 Gy was suppressed by the 0.1 Gy preirradiation, when cells were counted 2 days after irradiation. The inhibitors of protein kinase C (PKC) and DNA-dependent protein kinase (DNAPK) or phosphatidylinositol 3-kinase (PI3K) suppressed RAR. The treatment with the activators of PKC instead of 0.1 Gy-preirradiation also caused adaptive response to 2 Gy-irradiation. Moreover, glial cells cultured from severe combined immunodeficiency (scid) mice, which have lost DNAPK activity, and AT-2KY cells, fibroblasts of an ataxia-telangiectasia (AT) patient, showed no RAR. These results indicated that PKC, ATtM, DNAPK and/or PI3K were involved in RAR for growth of cultured glial cells. Proteomics [correction of preteomics] analysis of these cells exposed to low dose irradiation in now underway.  相似文献   

13.
This study concerns with the multiple treatment of the target site to potent carcinogen and the super imposition of low level radiofrequency and microwave radiation. Swiss albino mice (male) were used for this investigation. The study has been divided in two parts, part A: a single dose of 7,12-dimethylbenz(a)anthracene (DMBA) 100 μg/animal was applied topically on the skin of mice and were exposed to 112 MHz amplitude modulated (AM) at 16 Hz (power density 1.0 mW/cm(2), specific absorption rate (SAR) 0.75 W/kg). Similarly after a single dose of DMBA, mice were exposed to 2.45 GHz radiation (power density of 0.34 mW/cm(2), SAR, 0.1 W/kg), 2 h/day, 3 days a week for a period of 16 weeks. The two sets of experiments were carried out separately. Part B: mice were transplanted intraperitoneally (ip) with ascites 8 × 10(8) (Ehrlich-Lettre ascites, strain E) carcinoma cells per mouse. These mice were exposed to 112 MHz amplitude modulated at 16 Hz and 2.45 GHz radiation separately for a period of 14 days. There was no tumor development in mice exposed to RF and MW. Similarly a topical application of single dose of DMBA followed by RF/MW exposure also did not produce any visible extra tumor on the skin of mice. On the other hand mice were transplanted intraperitoneally with ascites (8 × 10(8) cell/ml) and subsequently exposed to above mentioned fields for 14 days showed a slight increase in the cell numbers as compared to the control group. However, the increase is insignificant. There were insignificant differences either in the mortality or cell proliferation among the control and exposed group. This results show that low level RF or MW do not alter tumor growth and development as evidenced by no observable change in tumor size.  相似文献   

14.
To investigate the effects of extremely low frequency magnetic fields on ultraviolet radiation (UV) exposed budding yeast, haploid yeast (Saccharomyces cerevisiae) cells of the strain SEy2101a were exposed to 50 Hz sine wave magnetic field (MF) of 120 microT with simultaneous exposure to UV radiation. Most of the UV energy was in the UVB range (280-320 nm). The biologically weighted (CIE action spectrum) dose level for the UV radiation was 175 J/m2. We examined whether 50 Hz MF affected the ability of UV irradiated yeast cells to form colonies (Colony Forming Units, CFUs). In addition, the effect of coexposure on cell cycle kinetics was investigated. Although the significant effect of MF on the cell cycle phases of UV exposed yeast cells was seen only at one time point, the overall results showed that MF exposure may influence the cell cycle kinetics at the first cycle after UV irradiation. The effect of our particular MF exposure on the colony forming ability of the UV irradiated yeast cells was statistically significant 420 min after UV irradiation. Moreover, at 240, 360, and 420 min after UV irradiation, there were fewer CFUs in every experiment in (UV+MF) exposed populations than in only UV exposed yeast populations. These results could indicate that MF exposure in conjunction with UV may have some effects on yeast cell survival or growth.  相似文献   

15.
Liu G  Gong P  Zhao H  Wang Z  Gong S  Cai L 《Radiation research》2006,165(4):379-389
Hormetic and adaptive responses induced by low-level radiation in hematopoietic and immune systems have been observed, as shown by stimulatory effects on cell growth and resistance to subsequent radiation-induced cytogenetic damage. However, in terms of cell death by apoptosis, the effects of low-level radiation are controversial: Some studies showed decreased apoptosis in response to low-level radiation while others showed increased apoptosis. This controversy may be related to the radiation doses or dose rates and also, more importantly, to the cell types. Testes are one of the most radiosensitive organs. The loss of male germ cells after exposure to ionizing radiation has been attributed to apoptosis. In the present study, the effects of low-level radiation at doses up to 200 mGy on mouse male germ cells in terms of apoptosis and the expression of apoptosis-related proteins were examined at different times after whole-body exposure of mice to low-level radiation. In addition, the effect of pre-exposure to low-level radiation on subsequent cell death induced by high doses of radiation was examined to explore the possibility of low-level radiation-induced adaptive response. The results showed that low-level radiation in the dose range of 25-200 mGy induced significant increases in apoptosis in both spermatogonia and spermatocytes, with the maximal effect at 75 mGy. The increased apoptosis is most likely associated with Trp53 protein expression. Furthermore, 75 mGy low-level radiation given pre-irradiation led to an adaptive response of seminiferous germ cells to subsequent high-level radiation-induced apoptosis. These results suggest that low-level radiation induces increased apoptosis in male germ cells but also induces a significant adaptive response that decreases cell death after a subsequent high-dose irradiation.  相似文献   

16.
Low‐dose (≤0.1 Gy) radiation‐induced adaptive responses could protect cells from high‐challenge dose radiation‐induced killing. The protective role is believed to promote the repair of DNA double‐strand breaks (DSBs) that are a severe threat to cell survival. However, it remains unclear which repair pathway, homologous recombination repair (HRR) or non‐homologous end‐joining (NHEJ), is promoted by low‐dose radiation. To address this question, we examined the effects of low‐dose (0.1 Gy) on high‐challenge dose (2–4 Gy) induced killing in NHEJ‐ or HRR‐deficient cell lines. We showed that 0.1 Gy reduced the high‐dose radiation‐induced killing for wild‐type or HRR‐deficient cells, but enhanced the killing for NHEJ‐deficient cells. Interestingly, low‐dose radiation also enhanced the killing for wild‐type cells exposed to high‐challenge dose radiation with high‐linear energy transfer (LET). Because it is known that high‐LET radiation induces an inefficient NHEJ, these results support that the low‐dose radiation‐stimulated protective role in reducing high‐challenge dose (low‐LET)‐induced cell killing might depend on NHEJ. In addition, we showed that low‐dose radiation activated the DNA‐PK catalytic subunit (DNA‐PKcs) and the inhibitor of DNA‐PKcs destroyed the low‐dose radiation‐induced protective role. These results suggest that low‐dose radiation might promote NHEJ through the stimulation of DNA‐PKcs activity and; therefore, increase the resistance of cells to high‐challenge dose radiation‐induced killing. J. Cell. Physiol. 226: 369–374, 2011. © 2010 Wiley‐Liss, Inc.  相似文献   

17.
Treatment of mammalian cells with lactate or inhibitors of glycolysis alters their radiation response, particularly in the low dose region of the dose response curve. The occurrence of both high lactate levels and high glycolytic metabolism in tumours is well known and therefore the effect of lactate on a cell line sensitive to radiation induced transformation was examined using a single exposure to Cobalt 60 gamma rays as the carcinogen challenge. The results indicate that cells treated with 5mM lactate before irradiation exhibit changes in morphology and growth rate and that the transformation frequency is increased by three to ten fold following 24 hours lactate treatment just prior to irradiation. Examination of radiation survival curves showed a positive correlation between transformation frequency and size of the shoulder, but increasing transformation frequency was associated with a decrease in Do. A mechanism involving altered Redox potential in lactate treated cells is suggested. The results are discussed in terms of their possible significance for radiotherapy.  相似文献   

18.
Heptaplatin, cis-malonato [(4R,5R)-4,5-bis (amino-methyl)-2-isopropyl-1,3-dioxolane] platinum(II) (SKI-2053R, Sunpla) is a new platinum derivative with anti-tumor activity comparable to cisplatin on various cancer cell lines. Preclinical studies suggest that it is less nephrotoxic than cisplatin. This study was undertaken to examine the combined effect of heptaplatin and ionizing radiation on two established human squamous carcinoma cell lines (NCI-H520, SQ20B). The cytotoxic activity of heptaplatin was concentration-dependent in both cell lines. When low dose heptaplatin was combined with high dose ionizing radiation, there was an additive cytotoxic effect on NCI-H520 cells (P < 0.05), while a moderate dose of heptaplatin and a low dose of ionizing radiation had an additive cytotoxic effect on the growth of SQ20B cells (P < 0.05). FACS analysis and DAPI staining showed that their additive cytotoxic effects were correlated with the induction of apoptosis. Further studies are warranted using heptaplatin and ionizing radiation in squamous cell carcinoma as a substitute for cisplatin.  相似文献   

19.
Improved risk characterization for stochastic biological effects of low doses of low-LET radiation is important for protecting nuclear workers and the public from harm from radiation exposure. Here we present a Bayesian approach to characterize risks of stochastic effects from low doses of low-LET radiation. The stochastic effect considered is neoplastic transformation of cells because it relates closely to cancer induction. We have used a published model of neoplastic transformation called NEOTRANS1. It is based on two different classes of cellular sensitivity for asynchronous, exponentially growing populations (in vitro). One sensitivity class is the hypersensitive cell; the other is the resistant cell. NEOTRANS1 includes the effects of genomic damage accumulation, DNA repair during cell cycle arrest, and DNA misrepair (non-lethal repair errors). The model-associated differential equations are solved for conditions of in vitro irradiation at a fixed rate. Previously published solutions apply only to high dose rates and were incorrectly assumed to apply to only high-LET radiation. Solutions provided here apply to any fixed dose rate and to both high- and low-LET radiations. Markov chain Monte Carlo methods are used to carry out the Bayesian inference of the low-dose risk for neoplastic transformation of aneuploid C3H 10T1/2 cells for X-ray doses from 0 to 1000 mGy. We have assumed that for this low-dose range only the hypersensitive fraction of the cells are affected. Our results indicate that the initial slope of the risk vs dose relationship for neoplastic transformation is as follows: (1) directly proportional to the fraction, f1, of hypersensitive cells; (2) directly proportional to the radiosensitivity of the genomic target; and (3) inversely proportional to the rate at which hypersensitive cells with radiation-induced damage are committed to undergo correct repair of genomic damage. Further, our results indicate that very fast molecular events are associated with the commitment of cells to the correct repair pathway. Results also indicate a relatively large probability for misrepair that leads to genomic instability. Our results are consistent with the view that for very low doses, dose rate is not an important variable for characterizing low-LET radiation risks so long as age-related changes in sensitivity do not occur during irradiation.  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号