Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Life Sci Space Res (Amst) ; 41: 202-209, 2024 May.
Article in English | MEDLINE | ID: mdl-38670648

ABSTRACT

Explorations of the Moon and Mars are planned as future manned space missions, during which humans will be exposed to both radiation and microgravity. We do not, however, know the health effects for such combined exposures. In a ground-based experiment, we evaluated the combined effects of radiation and simulated microgravity on tumorigenesis by performing X-irradiation and tail suspension in C3B6F1 ApcMin/+ mice, a well-established model for intestinal tumorigenesis. Mice were irradiated at 2 weeks of age and underwent tail suspension for 3 or 11 weeks using a special device that avoids damage to the tail. The tail suspension treatment significantly reduced the thymus weight after 3 weeks but not 11 weeks, suggesting a transient stress response. The combination of irradiation and tail suspension significantly increased the number of small intestinal tumors less than 2 mm in diameter as compared with either treatment alone. The combined treatment also increased the fraction of malignant tumors among all small intestinal tumors as compared with the radiation-only treatment. Thus, the C3B6F1 ApcMin/+ mouse is a useful model for assessing cancer risk in a simulated space environment, in which simulated microgravity accelerates tumor progression when combined with radiation exposure.


Subject(s)
Intestinal Neoplasms , Weightlessness Simulation , Animals , Mice , Intestinal Neoplasms/pathology , Intestinal Neoplasms/etiology , Carcinogenesis/radiation effects , Mice, Inbred C57BL , Hindlimb Suspension , Male , X-Rays , Disease Models, Animal , Female , Intestine, Small/radiation effects , Intestine, Small/pathology , Thymus Gland/radiation effects , Thymus Gland/pathology , Neoplasms, Radiation-Induced/pathology , Neoplasms, Radiation-Induced/etiology
2.
J Radiat Res ; 64(3): 622-631, 2023 May 25.
Article in English | MEDLINE | ID: mdl-37117033

ABSTRACT

Recent studies have identified interstitial deletions in the cancer genome as a radiation-related mutational signature, although most of them do not fall on cancer driver genes. Pioneering studies in the field have indicated the presence of loss of heterozygosity (LOH) spanning Apc in a subset of sporadic and radiation-induced intestinal tumors of ApcMin/+ mice, albeit with a substantial subset in which LOH was not detected; whether copy number losses accompany such LOH has also been unclear. Herein, we analyzed intestinal tumors of C3B6F1 ApcMin/+ mice that were either left untreated or irradiated with 2 Gy of γ-rays. We observed intratumor mosaicism with respect to the nuclear/cytoplasmic accumulation of immunohistochemically detectable ß-catenin, which is a hallmark of Apc+ allele loss. An immunoguided laser microdissection approach enabled the detection of LOH involving the Apc+ allele in ß-catenin-overexpressing cells; in contrast, the LOH was not observed in the non-overexpressing cells. With this improvement, LOH involving Apc+ was detected in all 22 tumors analyzed, in contrast to what has been reported previously. The use of a formalin-free fixative facilitated the LOH and microarray-based DNA copy number analyses, enabling the classification of the aberrations as nondisjunction/mitotic recombination type or interstitial deletion type. Of note, the latter was observed only in radiation-induced tumors (nonirradiated, 0 of 8; irradiated, 11 of 14). Thus, an analysis considering intratumor heterogeneity identifies interstitial deletion involving the Apc+ allele as a causative radiation-related event in intestinal tumors of ApcMin/+ mice, providing an accurate approach for attributing individual tumors to radiation exposure.


Subject(s)
Intestinal Neoplasms , Neoplasms, Radiation-Induced , Mice , Animals , beta Catenin/genetics , Neoplasms, Radiation-Induced/genetics , Mutation , Loss of Heterozygosity/genetics , Intestinal Neoplasms/genetics
3.
In Vivo ; 36(2): 618-627, 2022.
Article in English | MEDLINE | ID: mdl-35241514

ABSTRACT

BACKGROUND/AIM: An enriched environment (EE) modifies apoptotic cell death and promotes cell proliferation in the central nervous system (CNS) in mice. However, few studies have examined the effects of an EE on apoptosis in non-CNS organs in model orgamisms. In addition, the intestinal tract is one of organs at high-risk of carcinogenesis after radiation exposure. Herein we evaluated the effects of an EE on spontaneous and radiation-induced apoptosis in intestinal crypt cells of mice. MATERIALS AND METHODS: Juvenile (3-week-old) and adult (11-week-old) male B6C3F1 mice were housed in a standard environment or EE for 8 weeks and then were whole-body irradiated with 2 Gy X-rays. Apoptosis in the small intestine and colon was analyzed with antibody against cleaved caspase 3. RESULTS: The EE significantly reduced body weight; adipose tissue weight; and serum levels of total cholesterol, triglyceride, leptin, and insulin. Although EE did not change the spontaneous apoptotic index without irradiation, it significantly increased the index after irradiation in the colonic crypt. The apoptotic index in the small intestinal crypt showed similar patterns. CONCLUSION: An EE enhances radiation-induced apoptosis of stem/progenitor cells in the small intestine and colon without affecting spontaneous apoptosis. An EE may thus reduce the risk of cancer in the intestinal tract after radiation exposure such as radiotherapy.


Subject(s)
Apoptosis , Intestinal Mucosa , Animals , Cell Proliferation , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Male , Mice , Mice, Inbred Strains
4.
Biology (Basel) ; 11(3)2022 Mar 16.
Article in English | MEDLINE | ID: mdl-35336821

ABSTRACT

The risk of radiation-induced carcinogenesis depends on age at exposure. We previously reported principal causative genes in lymphomas arising after infant or adult exposure to 4-fractionated irradiation as Pten or Ikzf1, respectively, suggesting that cells with mutation in these genes might be the origin of lymphomas arising after irradiation depending on age at exposure. Here, we clarified the age-dependent differences in thymus-cell dynamics in mice during the initial post-irradiation period. The thymocyte number initially decreased, followed by two regeneration phases. During the first regeneration, the proportion of phosphorylated-AKT-positive (p-AKT+) cells in cell-cycle phases S+G2/M of immature CD4-CD8- and CD4+CD8+ thymocytes and in phases G0/G1 of mature CD4+CD8- and CD4-CD8+ thymocytes was significantly greater in irradiated infants than in irradiated adults. During the second regeneration, the proportion of p-AKT+ thymocytes in phases G0/G1 increased in each of the three populations other than CD4-CD8- thymocytes more so than during the first regeneration. Finally, PI3K-AKT-mTOR signaling in infants contributed, at least in part, to biphasic thymic regeneration through the modification of cell proliferation and survival after irradiation, which may be associated with the risk of Pten mutation-associated thymic lymphoma.

5.
Biology (Basel) ; 10(12)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34943185

ABSTRACT

The intergenerational effects from chronic low-dose exposure are matters of concern. It is thus important to elucidate the radiation-induced effects of germ cell maturation, fertilization and embryonic development. It is well known that DNA methylation levels in CpG sites in gametes are reprogrammed in stages during their maturity. Furthermore, the binding of Izumo on the surface of sperm and Juno on the surface of oocytes is essential for fertilization. Thus, there is a possibility that these genes are useful indicators to evaluate fertility in mice after irradiation exposure. Therefore, in this study, we analyzed global DNA methylation patterns in the testes and gene expression of Izumo1 and Izumo1r (Juno) in the gonads of mice after neonatal acute high-dose ionizing radiation (HDR) and chronic low-dose ionizing radiation (LDR). One-week-old male and female mice were irradiated with a total dose of 4 Gy, with acute HDR at 7 days at a dose rate of 30 Gy/h and LDR continuously at a dose rate of 6 mGy/h from 7 to 35 days. Their gonads were subsequently analyzed. The results of global DNA methylation patterns in the testes showed that methylation level increased with age in the control group, the LDR group maintained its DNA methylation level, and the HDR group showed decreased DNA methylation levels with age. In the control group, the gene expression level of Izumo1 in the testis did not show age-related changes, although there was high expression at 100 days of age. However, in the LDR group, the expression level recovered after the end of irradiation, while it remained low regardless of age in the HDR group. Conversely, gene expression of Izumo1r (Izumo1 receptor) in the ovary decreased with age in the control group. Although the gene expression of Izumo1r decreased with age in the LDR group, it remained low in the HDR group. Our results indicate that LDR can induce different DNA methylation patterns, and both high- and low-dose radiation before sexual maturity might affect gametogenesis and fertility.

6.
Radiat Res ; 196(2): 225-234, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34046685

ABSTRACT

Neutron radiation, a high-linear energy transfer radiation, has a high relative biological effectiveness (RBE) for various end points. The age at exposure is an important modifier of the effects of radiation, including carcinogenesis, with infants being generally more radiosensitive. Ptch1+/- mice offer a unique experimental system for assessing radiation carcinogenesis. Spontaneous development of medulloblastoma tumors occurs in nonirradiated animals that lose their Ptch1+ allele, most frequently by a loss of heterozygosity (LOH) of chromosome 13 via recombination or non-disjunction (referred to as S-type tumors). In contrast, tumors occur in irradiated Ptch1+/- mice as a result of chromosome 13 LOH with an interstitial deletion (R-type), making spontaneous and radiation-induced tumors discernible. To elucidate the influence of age on the effect of fast neutrons, we irradiated Ptch1+/- mice with neutrons (mean energy, ∼2 MeV) or γ rays on embryonic day (E)14 and E17 and on postnatal day (P)1, 4 or 10 and classified the resulting medulloblastomas based on chromosome 13 aberrations. Instead of LOH, some tumors harbored mutations in their Ptch1+ gene via a nonirradiation-associated mechanism such as duplication, insertion, base substitution or deletion with microhomology-mediated end joining; thus, these tumors were classified as S-type. The RBE regarding the induction of R-type tumors was 12.9 (8.6, 17.2), 9.6 (6.9, 12.3), 21.5 (17.2, 25.8), and 7.1 (4.7, 9.5) (mean and 95% confidence interval) for mice irradiated on E14, E17, P1 and P4, respectively, with the highest value seen during the most active development of the tissue and P10 being completely resistant. These results indicate that the developmental stage at exposure of the tissue influences the RBE of neutrons.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 13/radiation effects , Medulloblastoma/genetics , Neoplasms, Radiation-Induced/genetics , Patched-1 Receptor/genetics , Animals , Chromosomes, Human, Pair 13/genetics , Dose-Response Relationship, Radiation , Fast Neutrons/adverse effects , Humans , Loss of Heterozygosity/genetics , Loss of Heterozygosity/radiation effects , Medulloblastoma/etiology , Medulloblastoma/pathology , Mice , Neoplasms, Radiation-Induced/pathology , Radiation Tolerance/genetics , Radiation Tolerance/radiation effects , Relative Biological Effectiveness
7.
Int J Radiat Oncol Biol Phys ; 109(3): 813-818, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33190969

ABSTRACT

PURPOSE: Radiation therapy (RT) modulates the immune characteristics of the tumor microenvironment (TME). It is not known whether these effects are dependent on the type of RT used. METHODS AND MATERIALS: We evaluated the immunomodulatory effects of carbon-ion therapy (CiRT) compared with biologically equivalent doses of photon therapy (PhRT) on solid tumors. Orthotopic 4T1 mammary tumors in immunocompetent hosts were treated with CiRT or biologically equivalent doses of PhRT. Seventy-two hours after RT, tumors were harvested and the immune characteristics of the TME were quantified by flow cytometry and multiplex cytokine analyses. RESULTS: PhRT decreased the abundance of CD4+ and CD8+ T cells in the TME at all doses tested, with compensatory increases in proliferation. By contrast, CiRT did not significantly alter CD8+ T-cell infiltration. High-dose CiRT increased secretion of proinflammatory cytokines by tumor-infiltrating CD8+ T cells, including granzyme B, IL-2, and TNF-α, with no change in IFN-γ. Conversely, high-dose PhRT increased CD8+ T-cell secretion of IFN-γ only. At most of the doses studied, PhRT increased proliferation of immunosuppressive regulatory T cells; this was only seen with high-dose CiRT. Cytokine analyses of bulk dissociated tumors showed that CiRT significantly increased levels of IFN-γ, IL-2, and IL-1ß, whereas PhRT increased IL-6 levels alone. CONCLUSIONS: At low doses, lymphocytes differ in their sensitivity to CiRT compared with PhRT. Unlike PhRT, low-dose CiRT is generally lymphocyte-sparing. At higher doses, CiRT is a more potent inducer of proinflammatory cytokines and merits further study as a modulator of the immunologic characteristics of the TME.


Subject(s)
CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/radiation effects , Heavy Ion Radiotherapy , Mammary Neoplasms, Animal/radiotherapy , Photons/therapeutic use , Tumor Microenvironment/radiation effects , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Female , Flow Cytometry , Granzymes/metabolism , Granzymes/radiation effects , Immunocompetence , Interferon-gamma/metabolism , Interferon-gamma/radiation effects , Interleukin-1beta/metabolism , Interleukin-1beta/radiation effects , Interleukin-2/metabolism , Interleukin-2/radiation effects , Interleukin-6/metabolism , Interleukin-6/radiation effects , Mammary Neoplasms, Animal/immunology , Mice , Relative Biological Effectiveness , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/radiation effects , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/radiation effects
8.
Biomed Res Int ; 2020: 4703286, 2020.
Article in English | MEDLINE | ID: mdl-32337251

ABSTRACT

Space travel has advanced significantly over the last six decades with astronauts spending up to 6 months at the International Space Station. Nonetheless, the living environment while in outer space is extremely challenging to astronauts. In particular, exposure to space radiation represents a serious potential long-term threat to the health of astronauts because the amount of radiation exposure accumulates during their time in space. Therefore, health risks associated with exposure to space radiation are an important topic in space travel, and characterizing space radiation in detail is essential for improving the safety of space missions. In the first part of this review, we provide an overview of the space radiation environment and briefly present current and future endeavors that monitor different space radiation environments. We then present research evaluating adverse biological effects caused by exposure to various space radiation environments and how these can be reduced. We especially consider the deleterious effects on cellular DNA and how cells activate DNA repair mechanisms. The latest technologies being developed, e.g., a fluorescent ubiquitination-based cell cycle indicator, to measure real-time cell cycle progression and DNA damage caused by exposure to ultraviolet radiation are presented. Progress in examining the combined effects of microgravity and radiation to animals and plants are summarized, and our current understanding of the relationship between psychological stress and radiation is presented. Finally, we provide details about protective agents and the study of organisms that are highly resistant to radiation and how their biological mechanisms may aid developing novel technologies that alleviate biological damage caused by radiation. Future research that furthers our understanding of the effects of space radiation on human health will facilitate risk-mitigating strategies to enable long-term space and planetary exploration.


Subject(s)
Cosmic Radiation/adverse effects , Space Flight , Ultraviolet Rays , Animals , Astronauts , Carcinogenesis/radiation effects , Central Nervous System/radiation effects , Chromosome Aberrations/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , Extraterrestrial Environment , Genomic Instability/radiation effects , Humans , Micronuclei, Chromosome-Defective/radiation effects , Protective Agents/pharmacology , Radiation Dosage , Radiation Exposure/adverse effects , Radiation Exposure/prevention & control , Stress, Psychological , Weightlessness
9.
J Radiat Res ; 61(2): 187-196, 2020 Mar 23.
Article in English | MEDLINE | ID: mdl-31909805

ABSTRACT

The risk of cancer due to exposure to ionizing radiation is higher in infants than in adults. In a previous study, the effect of adult-onset calorie restriction (CR) on carcinogenesis in mice after early-life exposure to X-rays was examined (Shang, Y, Kakinuma, S, Yamauchi, K, et al. Cancer prevention by adult-onset calorie restriction after infant exposure to ionizing radiation in B6C3F1 male mice. Int J Cancer. 2014; 135: 1038-47). The results showed that the tumor frequency was reduced in the CR group. However, the mechanism of tumor suppression by CR is not yet clear. In this study, we examined the effects of CR on radiation-induced mutations using gpt delta mice, which are useful to analyze mutations in various tissues throughout the whole body. Infant male mice (1-week old) were exposed to 3.8 Gy X-rays and fed a control (95 kcal/week/mouse) or CR (65 kcal/week/mouse) diet from adult stage (7-weeks old). Mice were sacrificed at the age of 7 weeks, 8 weeks and 100 days, and organs (spleen, liver, lung, thymus) were harvested. Mutations at the gpt gene in the DNA from the spleen were analyzed by using a gpt assay protocol that detects primarily point mutations in the gpt gene. The results showed that mutation frequencies were decreased in CR groups compared with non-CR groups. Sequence analysis of the gpt gene in mutants revealed a reduction in the G:C to T:A transversion in CR groups. Since it is known that 8-oxoguanine could result in this base substitution and that CR has an effect of reducing oxidative stress, these results indicate that the suppression of oxidative stress by CR is the cause of the reduction of this transversion.


Subject(s)
Caloric Restriction , Mutation/genetics , Radiation, Ionizing , Spleen/pathology , Spleen/radiation effects , Animals , Animals, Newborn , Body Weight/radiation effects , Female , Male , Mice, Inbred C57BL , Mutation Rate , X-Rays
10.
Cancer Sci ; 111(3): 840-848, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31925975

ABSTRACT

Ionizing radiation can damage DNA and, therefore, is a risk factor for cancer. Eker rats, which carry a heterozygous germline mutation in the tumor-suppressor gene tuberous sclerosis complex 2 (Tsc2), are susceptible to radiation-induced renal carcinogenesis. However, the molecular mechanisms involved in Tsc2 inactivation are unclear. We subjected Fischer 344 × Eker (Long Evans Tsc2+/- ) F1 hybrid rats to gamma-irradiation (2 Gy) at gestational day 19 (GD19) or postnatal day 5 (PND5) and investigated the patterns of genomic alterations in the Tsc2 allele of renal tumors that developed at 1 year after irradiation (N = 24 tumors for GD19, N = 10 for PND5), in comparison with spontaneously developed tumors (N = 8 tumors). Gamma-irradiation significantly increased the multiplicity of renal tumors. The frequency of LOH at the chromosome 10q12 region, including the Tsc2 locus, was 38%, 29% and 60% in renal carcinomas developed from the nonirradiated, GD19 and PND5 groups, respectively. Array comparative genomic hybridization analysis revealed that the LOH patterns on chromosome 10 in renal carcinomas were classified into chromosomal missegregation, mitotic recombination and chromosomal deletion types. LOH of the interstitial chromosomal deletion type was observed only in radiation-associated carcinomas. Sequence analysis for the wild-type Tsc2 allele in the LOH-negative carcinomas identified deletions (nonirradiated: 26%; GD19: 21%) and base-substitution mutations (GD19: 4%). Reduced expression of Tsc2 was also observed in the majority of the LOH-negative carcinomas. Our results suggest that interstitial chromosomal deletion is a characteristic mutagenic event caused by ionizing radiation, and it may contribute to the assessment of radiation-induced cancer risk.


Subject(s)
Kidney Neoplasms/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis/genetics , Alleles , Animals , Chromosome Deletion , Chromosomes, Human, Pair 10/genetics , Comparative Genomic Hybridization/methods , Gamma Rays/adverse effects , Heterozygote , Humans , Male , Mutation/genetics , Rats , Rats, Inbred F344 , Rats, Long-Evans , Risk , Tumor Suppressor Proteins/genetics
11.
Cytokine ; 114: 92-97, 2019 02.
Article in English | MEDLINE | ID: mdl-30467095

ABSTRACT

Whole body irradiation causes significant apoptosis in various tissues such as the thymus. If apoptotic cells outnumber the phagocytic capacity of macrophages, apoptosis becomes secondary necrosis, inducing inflammatory cytokine expression in macrophages. Radiation also induces thymic lymphomas in C57BL/6 mice after four consecutive irradiations with 1.6 Gy X-rays with nearly 100% incidence. Since cancer development is modulated by a microenvironment involving macrophages, we examined the kinetics of thymocyte number and plastic adherent cell number in the thymus as well as cytokine mRNA expression by plastic adherent cells in the thymus after split-dose irradiation. Upon split-dose irradiation, thymocyte number changed dramatically, whereas plastic adherent cell number did not. Among cytokine mRNAs tested, IL-1ß, IL-11 and IL-12p40 mRNAs were up regulated 2 days after the 1st and 2nd, 3rd and 4th, and 2nd and 3rd irradiations, respectively. On the other hand, TNF-α mRNA was up regulated 2 days after the 3rd irradiation and 2 weeks after the 4th irradiation. The level of IL-11 protein was also increased 2 days after 3rd and 4th irradiations. These results suggest that, upon split-dose irradiation, macrophages in the thymus produce various cytokines in a time-dependent manner, thereby contributing to induction of thymic lymphomas.


Subject(s)
Cytokines/genetics , Plastics/pharmacology , Radiation Dosage , Thymus Gland/cytology , Thymus Gland/radiation effects , Animals , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Count , Cytokines/blood , Cytokines/metabolism , Female , Kinetics , Mice, Inbred C57BL , Phagocytosis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thymocytes/cytology , Thymocytes/metabolism , Thymocytes/radiation effects
12.
Radiat Res ; 188(2): 235-241, 2017 08.
Article in English | MEDLINE | ID: mdl-28581892

ABSTRACT

There is a natural tendency to expect that irradiation of an infant organ prior to development-related expansion will result in a higher risk of developing cancer than that of fully-developed adult tissue, and this has generally been observed. However, if tissues also vary in their initial responses to radiation depending on age, the interplay between tissue- and age-dependent risk would potentially be quite complex. We have previously shown opposing age-dependent induction of apoptosis for the intestinal epithelium and hematopoietic cells in mice, but such data are not yet available for the liver. Here, we have examined markers of DNA damage, initiation of DNA damage responses, cell cycle arrest, apoptosis and proliferation, as well as gene expression, in the B6C3F1 mouse liver over the hours and days after irradiation of mice at 1 or 7 weeks of age. We found that induction and resolution of radiation-induced DNA damage is not accompanied by significant changes in these cellular end points in the adult liver, while in infant hepatocytes modest induction of p53 accumulation and p21-mediated cell cycle arrest in a small fraction of damaged cells was overshadowed by a further stimulation of proliferation over the relatively high levels already found in the neonatal liver. We observed distinct expression of genes that regulate cell division between the ages, which may contribute to the differential responses. These data suggest that the growth factor signaling environment of the infant liver may mediate radiation-induced proliferation and increased liver cancer risk after irradiation during early life.


Subject(s)
Aging/radiation effects , Hepatocytes/cytology , Hepatocytes/radiation effects , Animals , Animals, Newborn , Cell Proliferation/radiation effects , DNA Damage , Female , Gene Expression Regulation/radiation effects , Hepatocytes/metabolism , Male , Mice , Mice, Inbred C57BL
13.
Radiat Res ; 186(4): 407-414, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27690174

ABSTRACT

Recently reported studies have led to a heightened awareness of the risks of cancer induced by diagnostic radiological imaging, and in particular, the risk of brain cancer after childhood CT scans. One feature of Ptch1+/- mice is their sensitivity to radiation-induced medulloblastomas (an embryonic cerebellar tumor) during a narrow window of time centered on the days around birth. Little is known about the dynamics of how dose protraction interacts with such narrow windows of sensitivity in individual tissues. Using medulloblastomas from irradiated Ptch1+/- mice with a hybrid C3H × C57BL/6 F1 genetic background, we previously showed that the alleles retained on chromosome 13 (which harbors the Ptch1 gene) reveal two major mechanisms of loss of the wild-type allele. The loss of parental alleles from the telomere extending up to or past the Ptch1 locus by recombination (spontaneous type) accounts for almost all medulloblastomas in nonirradiated mice, while tumors in irradiated mice often exhibited interstitial deletions, which start downstream of the wild-type Ptch1 and extend up varying lengths towards the centromere (radiation type). In this study, Ptch1+/- mice were exposed to an acute dose of either 100 or 500 mGy gamma rays in utero or postnatally, or the same radiation doses protracted over a four-day period, and were monitored for medulloblastoma development. The results showed dose- and age-dependent radiation-induced type tumors. Furthermore, the size of the radiation-induced deletion differed with the dose rate. The results of this work suggest that tumor latency may be related to the size of the deletion. In this study, 500 mGy exposure produced radiation-induced type tumors at all ages and dose rates, while 100 mGy exposure did not significantly produce radiation-induced type tumors. The radiation signature allows for unique mechanistic insight into the action of radiation to induce DNA lesions with known causal relationship to a specific tumor type, particularly for doses and dose rates that are relevant to both diagnostic and accidental radiological exposures.


Subject(s)
Cerebellar Neoplasms/genetics , Gamma Rays/adverse effects , Heterozygote , Medulloblastoma/genetics , Neoplasms, Radiation-Induced/genetics , Patched-1 Receptor/genetics , Animals , Cerebellar Neoplasms/etiology , Cerebellar Neoplasms/pathology , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/radiation effects , Dose-Response Relationship, Radiation , Kaplan-Meier Estimate , Loss of Heterozygosity/radiation effects , Medulloblastoma/etiology , Medulloblastoma/pathology , Mice , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/pathology , Time Factors
14.
Nihon Rinsho ; 74 Suppl 7: 77-82, 2016 09.
Article in Japanese | MEDLINE | ID: mdl-30634733
15.
PLoS One ; 10(6): e0130666, 2015.
Article in English | MEDLINE | ID: mdl-26125582

ABSTRACT

Monitoring mice exposed to carbon ion radiotherapy provides an indirect method to evaluate the potential for second cancer induction in normal tissues outside the radiotherapy target volume, since such estimates are not yet possible from historical patient data. Here, male and female B6C3F1 mice were given single or fractionated whole-body exposure(s) to a monoenergetic carbon ion radiotherapy beam at the Heavy Ion Medical Accelerator in Chiba, Japan, matching the radiation quality delivered to the normal tissue ahead of the tumour volume (average linear energy transfer = 13 keV x µm(-1)) during patient radiotherapy protocols. The mice were monitored for the remainder of their lifespan, and a large number of T cell lymphomas that arose in these mice were analysed alongside those arising following an equivalent dose of 137Cs gamma ray-irradiation. Using genome-wide DNA copy number analysis to identify genomic loci involved in radiation-induced lymphomagenesis and subsequent detailed analysis of Notch1, Ikzf1, Pten, Trp53 and Bcl11b genes, we compared the genetic profile of the carbon ion- and gamma ray-induced tumours. The canonical set of genes previously associated with radiation-induced T cell lymphoma was identified in both radiation groups. While the pattern of disruption of the various pathways was somewhat different between the radiation types, most notably Pten mutation frequency and loss of heterozygosity flanking Bcl11b, the most striking finding was the observation of large interstitial deletions at various sites across the genome in carbon ion-induced tumours, which were only seen infrequently in the gamma ray-induced tumours analysed. If such large interstitial chromosomal deletions are a characteristic lesion of carbon ion irradiation, even when using the low linear energy transfer radiation to which normal tissues are exposed in radiotherapy patients, understanding the dose-response and tissue specificity of such DNA damage could prove key to assessing second cancer risk in carbon ion radiotherapy patients.


Subject(s)
Heavy Ion Radiotherapy/adverse effects , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/radiotherapy , Neoplasms, Radiation-Induced/genetics , Neoplasms, Second Primary/genetics , Animals , Chromosome Deletion , DNA Damage/genetics , DNA Damage/radiation effects , Dose-Response Relationship, Radiation , Female , Gamma Rays/adverse effects , Genetic Testing/methods , Heavy Ions/adverse effects , Japan , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
16.
J Radiat Res ; 55(4): 812-5, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24699001

ABSTRACT

We investigated the relative biological effectiveness (RBE) of therapeutic proton beams at six proton facilities in Japan with respect to cell lethality of HSG cells. The RBE of treatments could be determined from experimental data. For this purpose, we used a cell survival assay to compare the cell-killing efficiency of proton beams. Among the five linear accelerator (LINAC) X-ray machines at 4 or 6 MeV that were used as reference beams, there was only a small variation (coefficient of variation CV = 3.1% at D10) in biological effectiveness. The averaged value of D10 for the proton beams at the middle position of the spread-out Bragg peak (SOBP) was 4.98. These values showed good agreement, with a CV of 4.3% among the facilities. Thus, the average RBE10 (RBE at the D10 level) at the middle position of the SOBP beam for six facilities in Japan was 1.05 with a CV of 2.8%.


Subject(s)
Proton Therapy , Salivary Gland Neoplasms/radiotherapy , Cell Line, Tumor , Cell Survival/radiation effects , Humans , Japan , Relative Biological Effectiveness , Salivary Gland Neoplasms/pathology , Tumor Stem Cell Assay
17.
Radiat Res ; 177(5): 614-27, 2012 May.
Article in English | MEDLINE | ID: mdl-22206232

ABSTRACT

We measured the yield and spectrum of strand breaks and nucleobase lesions produced in fully hydrated plasmid DNA films to determine the linear energy transfer (LET) dependence of DNA damage induced by ion-beam irradiation in relation to the change in the atomic number of ions. The yield of isolated damage was revealed as a decrease in prompt SSBs with increasing LET of He(2+), C(5+,6+) and Ne(8+,10+) ions. On the other hand, the yields of prompt DSBs increased with increasing ion LET. SSBs were additionally induced in ion-irradiated DNA film by treatment with two kinds of base excision repair proteins (glycosylases), Nth and Fpg, indicating that base lesions are produced in the hydrated DNA film. This result shows that nucleobase lesions are produced via both chemical reactions with diffusible water radicals, such as OH radicals, and direct energy deposition onto DNA and the hydrated water layer. Nth-sensitive sites deduced to be pyrimidine lesions, such as 5,6-dihydrothymine (DHT), showed a relatively larger yield than Fpg-sensitive sites deduced to be purine lesions, such as 7,8-dihydro-8-oxo-2'deoxyguanine (8-oxoGua), for all ion exposures tested. The yield of SSBs or DSBs observed by enzyme treatment decreased noticeably with increasing LET for all tested ions. These results indicated that higher-LET ions preferentially produce a complex type of damage that might compromise the activities of the glycosylases used in this study. These findings are biologically important since, under cell mimicking conditions, persistent DNA damage occurs in part due to direct energy deposition on the DNA or hydrated water shell that is specifically induced by dense ionization in the track.


Subject(s)
Alpha Particles/adverse effects , DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA Damage , DNA, Bacterial/radiation effects , Heavy Ions/adverse effects , Plasmids/genetics , Carbon , DNA-Formamidopyrimidine Glycosylase , Deoxyribonuclease (Pyrimidine Dimer) , Free Radicals , Linear Energy Transfer , Neon , Purines/radiation effects , Pyrimidines/radiation effects , Water
18.
Mutat Res ; 701(1): 47-51, 2010 Aug 14.
Article in English | MEDLINE | ID: mdl-20420934

ABSTRACT

In previous studies we have shown that the linear energy transfer (LET)-relative biological effectiveness (RBE) curves were affected by LET and ion species [1,2]. In this paper we have examined the difference in the repair kinetics of G1-prematurely condensed chromosome breaks in normal human fibroblasts following irradiation with different heavy-ion beams of similar LET values. Normal human fibroblasts were irradiated with about 110 keV/microm of carbon (135 MeV/n), neon (400 MeV/n) and silicon ions (490 MeV/n), and the doses of carbon (3.25 Gy), neon (2.94+/-0.01 Gy) and silicon (2.31 Gy) were chosen to produce approximately the same number of initially measured G1-premature chromosome condensation (PCC) breaks (about 37 excess fragments per cell). The number of G1-PCC breaks was counted as excess fragments of prematurely condensed chromosomes using the PCC technique in the G1/G0 phase. The fractions of residual G1-PCC breaks after 24 h post-irradiation and half time, which is the time point where 50% of initially measured G1-PCC breaks are rejoined (t1/2), of the slow components of rejoining in carbon- and neon-ion irradiated cells were different from those of silicon-ion irradiated cells. However, no difference was observed in the half time of the fast components of rejoining in each ion beam. The results suggest that the difference in the fractions of residual G1-PCC breaks after 24 h post-irradiation reflect the result of the slow repair process for induced G1-PCC breaks, and that the repair process is dependent on the ion species, not the LET values.


Subject(s)
Chromosome Aberrations , Heavy Ions/adverse effects , Linear Energy Transfer , DNA Repair , Fibroblasts/radiation effects , G1 Phase , Humans
19.
J Radiat Res ; 50(5): 395-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19680011

ABSTRACT

We studied cellular responses in normal human fibroblasts induced with low-dose (rate) or low-fluence irradiations of different radiation types, such as gamma rays, neutrons and high linear energy transfer (LET) heavy ions. The cells were pretreated with low-dose (rate) or low-fluence irradiations (approximately 1 mGy/7-8 h) of 137Cs gamma rays, 241Am-Be neutrons, helium, carbon and iron ions before irradiations with an X-ray challenging dose (1.5 Gy). Helium (LET = 2.3 keV/microm), carbon (LET = 13.3 keV/microm) and iron (LET = 200 keV/microm) ions were produced by the Heavy Ion Medical Accelerator in Chiba (HIMAC), Japan. No difference in cell-killing effect, measured by a colony forming assay, was observed among the pretreatment with different radiation types. In mutation induction, which was detected in the hypoxanthine-guanine phosphoribosyltransferase (hprt) locus to measure 6-thioguanine resistant clones, there was no difference in mutation frequency induced by the X-ray challenging dose between unpretreated and gamma-ray pretreated cells. In the case of the pretreatment of heavy ions, X-ray-induced mutation was around 1.8 times higher in helium-ion pretreated and 4.0 times higher in carbon-ion pretreated cells than in unpretreated cells (X-ray challenging dose alone). However, the mutation frequency in cells pretreated with iron ions was the same level as either unpretreated or gamma-ray pretreated cells. In contrast, it was reduced at 0.15 times in cells pretreated with neutrons when compared to unpretreated cells. The results show that cellular responses caused by the influence of hprt mutation induced in cells pretreated with low-dose-rate or low-fluence irradiations of different radiation types were radiation-quality dependent manner.


Subject(s)
Cell Survival/radiation effects , Fibroblasts/physiology , Fibroblasts/radiation effects , Mutation/physiology , Mutation/radiation effects , Radiation Tolerance/physiology , Cells , Dose-Response Relationship, Radiation , Humans , Radiation Dosage , Radiation Tolerance/radiation effects
20.
Radiat Res ; 170(2): 163-71, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18666815

ABSTRACT

We studied the LET and ion species dependence of the induction of chromatin breaks measured immediately after irradiation as initially measured breaks and after 24 h postirradiation incubation (37 degrees C) as non-rejoined breaks in normal human fibroblasts with different heavy ions, such as carbon, neon, silicon and iron, generated by the Heavy Ion Medical Accelerator in Chiba (HIMAC) at the National Institute of Radiological Science (NIRS). Chromatin breaks were measured as an excess number of fragments of prematurely condensed chromosomes using premature chromosome condensation (PCC). The results showed that the number of excess fragments per cell per Gy for initially measured chromatin breaks was dependent on LET in the range from 13.3 to 113.1 keV/mum but was not dependent on ion species. On the other hand, the number of non-rejoined chromatin breaks detected after 24 h postirradiation incubation was clearly dependent on both LET and ion species. No significant difference was observed in the cross section for initially measured breaks, but a statistically significant difference was observed in the cross section for non-rejoined breaks among carbon, neon, silicon and iron ions. This suggests that the LET-dependent structure in the biological effects is reflected in biological consequences of repair processes.


Subject(s)
Chromatin/genetics , Chromatin/radiation effects , DNA Breaks , DNA Repair/genetics , DNA Repair/radiation effects , Fibroblasts/radiation effects , Linear Energy Transfer/physiology , Cell Line , Dose-Response Relationship, Radiation , Fibroblasts/physiology , Heavy Ions , Humans , Linear Energy Transfer/radiation effects , Radiation Dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...