64. 2011年4月06日 02:25:11: cqRnZH2CUM
あ、一応、あったか 0.35 or 1.2 mGy/h程度を、5週間照射すると免疫系が活性化して寿命が延びるということか確かに、これを人にあてはめられるなら、浪江町程度(〜100μSv)なら、寿命が延びるってことになる ただ人だとどうなるか、もっと強い線量だとどうなるかをチェックした方がいいから やはり浪江町の残留男の出番だなw Radiat Res. 2005 Apr;163(4):418-23. Further study of prolongation of life span associated with immunological modification by chronic low-dose-rate irradiation in MRL-lpr/lpr mice: effects of whole-life irradiation. Ina Y, Sakai K. Low Dose Radiation Research Center, Central Research Institute of Electric Power Industry, 2-11-1 Iwado-kita, Komae, Tokyo, 201-8511, Japan. Abstract MRL-lpr/lpr mice carry a deletion in the apoptosis-regulating Fas gene that markedly shortens life due to multiple severe diseases. In our previous study (Radiat. Res. 161, 168- 173, 2004), chronic low-dose-rate gamma irradiation of mice at 0.35 or 1.2 mGy/h for 5 weeks markedly prolonged the life span, accompanied by immunological activation. This report shows that extension of the irradiation period to the entire life of the mice at the same dose rates improved survival further. The 50% survival time for untreated mice, 134 days, was prolonged to 502 days by 1.2 mGy/h life-long irradiation. Also obtained were a time course and a radiation dose-rate response for the activation of the immune system as indicated by a significant increase in CD4+ CD8+ T cells in the thymus and CD8+ T cells in the spleen and also by a significant decrease in CD3+ CD45R/B220+ cells and CD45R/B220+ CD40+ cells in the spleen. Drastic ameliorations of multiple severe diseases, i.e. total-body lymphadenopathy, splenomegaly and serious autoimmune diseases including proteinuria, and kidney and brain-central nervous system syndromes, were found in parallel with these immunological activations, with lifelong low-dose-rate irradiation being more effective than 5-week irradiation at low dose rates. PMID: 15799698 [PubMed - indexed for MEDLINE] http://www.ncbi.nlm.nih.gov/pubmed/14731073 Prolongation of life span associated with immunological modification by chronic low-dose-rate irradiation in MRL-lpr/lpr mice.
Ina Y, Sakai K. Low Dose Radiation Research Center, Central Research Institute of Electric Power Industry, 2-11-1 Iwado-kita, Komae, Tokyo 201-8511, Japan. y-i@criepi.denken.or.jp Abstract Chronic low-dose-rate gamma irradiation at 0.35 or 1.2 mGy/h prolonged the life span of MRL-lpr/lpr mice carrying a deletion in the apoptosis-regulating Fas gene that markedly shortens life due to severe autoimmune disease. Immunological modifications as indicated by a significant increase of CD8(+) T cells and a significant decrease of CD3(+) CD45R/B220(+) as well as CD45R/B220(+) CD40(+) cells were found in parallel with amelioration of total-body lymphadenopathy, splenomegaly, proteinuria, and kidney and brain syndromes. http://informahealthcare.com/doi/abs/10.1080/09553000500519808 Activation of immunological network by chronic low-dose-rate irradiation in wild-type mouse strains: Analysis of immune cell populations and surface molecules
2005, Vol. 81, No. 10 , Pages 721-729 (doi:10.1080/09553000500519808) HTML PDF (382 KB) PDF Plus (390 KB) Reprints Permissions Yasuhiro Ina, DMSc1† and Kazuo Sakai1 1LDRC (Low Dose Radiation Research Center), CRIEPI (Central Research Institute of Electric Power Industry), Komae, Tokyo, Japan †Correspondence: Yasuhiro Ina, LDRC (Low Dose Radiation Research Center), CRIEPI (Central Research Institute of Electric Power Industry), 2-11-1 Iwado-kita, Komae, Tokyo, 201-8511, Japan, +81 33480 2111, +81 33480 3113 hormesis@yasuhiro-ina-dmsc.jp Purpose: To analyse the effects of chronic whole body low-dose-rate irradiation on the immune system in various wild-type mouse strains in comparison with the effects from acute high-dose-rate irradiation. Materials and methods: Wild-type mouse strains (C57BL/6, BALB/c, C3H/He, DBA/1, DBA/2 and CBA) were observed after chronic low-dose-rate γ irradiation at 1.2 mGy hour−1 by intensive analysis of immune cell populations and their various surface molecules, together with antibody-producing activity both with and without immunization by sheep red blood cells (SRBC). The cell surface functional molecules [cluster of differentiation (CD) 3, CD4, CD8, CD19, CD45R/B220, intercellular adhesion molecule (ICAM)-1, Fas, natural killer (NK)-1.1, chemokine {C-X-C motif } receptor 4 (CXCR4) and chemokine {C-C motif } receptor 5 (CCR5)] and activation molecules [thymocyte-activating molecule (THAM), CD28, CD40, CD44H, CD70, B7-1, B7-2, OX-40 antigen, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), CD30 ligand and CD40 ligand] were studied in the bone marrow, thymus, spleen, lymph nodes and peripheral blood by flow cytometry. Results: By chronic low-dose-rate irradiation alone, CD4+ T cells and CD8 molecule expression increased significantly by a maximum of 30%, while CD40+ B cells decreased significantly. Increases of CD4+ T cells, CD40+ B cells and anti-SRBC antibody-producing cells by immunization were significantly enhanced by continuous low-dose-rate irradiation at 1.2 mGy hour−1. CD3− CD4+ T cells, representative of abnormal immune cells, were absent in the chronically low-dose-rate-irradiated mice, while a dose-dependent increase of these cells was found in acutely high-dose-rate-irradiated mice given the same total doses. Conclusion: Chronic low-dose-rate radiation activated the immune system of the whole body. Keywords Chronic low-dose-rate irradiation, immunological network activation, immune cell populations, cell surface molecules, abnormal immune cells, wild-type mouse strains Read More: http://informahealthcare.com/doi/abs/10.1080/09553000500519808 http://yasuhiro-ina-dmsc.jp/profile.html 世界中で公刊発表された 稲 博士の世界初の 低線量率放射線療法 関連の代表的な英文学術論文 全論文とも、 稲 博士が 筆頭著者 兼 代表著者、かつ、 Corresponding Author になっておられ、 論文に関する連絡先も、 全論文とも 稲 博士 宛になっております。 1. Yasuhiro Ina et al., Prolongation of Life Span Associated with Immunological Modification by Chronic Low-Dose-Rate Irradiation in MRL-lpr/lpr Mice. Radiation Research, 161, 168━173 (2004). 2. Yasuhiro Ina et al., Suppression of Thymic Lymphoma Induction by Life-Long Low-Dose-Rate Irradiation Accompanied by Immune Activation in C57BL/6 Mice. Radiation Research, 163, 153━158 (2005). 3. Yasuhiro Ina et al., Further Study of Prolongation of Life Span Associated with Immunological Modification by Chronic Low-Dose-Rate Irradiation in MRL-lpr/lpr Mice : Effects of Whole-Life Irradiation. Radiation Research, 163, 418━423 (2005). 4. Yasuhiro Ina et al., Activation of Immunological Network by Chronic Low-Dose-Rate Irradiation in Wild-Type Mouse Strains : Analysis of Immune Cell Populations and Surface Molecules. International Journal of Radiation Biology, 81, 721━729 (2005). |
|
65. 2011年4月06日 02:37:37: cqRnZH2CUM
http://www.groenerekenkamer.nl/grkfiles/images/Dr_INA_Rad_Res_lpr2.pdf RADIATION RESEARCH 163, 418–423 (2005) q 2005 by Radiation Research Society. All rights of reproduction in any form reserved.Further Study of Prolongation of Life Span Associated with Immunological Modification by Chronic Low-Dose-Rate Irradiation in MRL-lpr/lpr Mice: Effects of Whole-Life Irradiation Yasuhiro Ina1 and Kazuo Sakai Low Dose Radiation Research Center, Central Research Institute of Electric Power Industry, 2-11-1 Iwado-kita, Komae, Tokyo, 201-8511, Japan Ina, Y. and Sakai, K. Further Study of Prolongation of Life Span Associated with Immunological Modification by Chronic Low-Dose-Rate Irradiation in MRL-lpr/lpr Mice: Effects of Whole-Life Irradiation. Radiat. Res. 163, 418–423 (2005). MRL-lpr/lpr mice carry a deletion in the apoptosis-regulating Fas gene that markedly shortens life due to multiple severe diseases. In our previous study (Radiat. Res. 161, 168– 173, 2004), chronic low-dose-rate g irradiation of mice at 0.35 or 1.2 mGy/h for 5 weeks markedly prolonged the life span, accompanied by immunological activation. This report shows that extension of the irradiation period to the entire life of the mice at the same dose rates improved survival further. The 50% survival time for untreated mice, 134 days, was prolonged to 502 days by 1.2 mGy/h life-long irradiation. Also obtained were a time course and a radiation dose-rate response for the activation of the immune system as indicated by a significant increase in CD41 CD81 T cells in the thymus and CD81 T cells in the spleen and also by a significant decrease in CD31 CD45R/B2201 cells and CD45R/B2201 CD401 cells in the spleen. Drastic ameliorations of multiple severe diseases, i.e. total-body lymphadenopathy, splenomegaly and serious autoimmune diseases including proteinuria, and kidney and brain-central nervous system syndromes, were found in parallel with these immunological activations, with lifelong low-dose-rate irradiation being more effective than 5-week irradiation at low dose rates. q 2005 by Radiation Research Society INTRODUCTION MRL-lpr/lpr mice, which carry a mutation of the apoptosis- regulating Fas gene, develop multiple severe diseases and have a very short life span, thus providing interesting animal models for experimental therapy (1–7). In our previous study, we demonstrated a marked prolongation of the life span of MRL-lpr/lpr mice by 5-week g irradiation at a low dose rate of 0.35 mGy/h and more effectively at 1.2 mGy/h (8). These life-span prolongations were associated with immunological activation and amelioration of au- 1 Address for correspondence: Low Dose Radiation Research Center, Central Research Institute of Electric Power Industry, 2-11-1 Iwado-kita, Komae, Tokyo, 201-8511, Japan. toimmune diseases. The present study shows a greater effect of chronic low-dose-rate irradiation continued for almost the entire life span of the mice, together with the results of observations completed for the entire life span of the mice after a 5-week irradiation at low dose rates. Low-dose radiation induces various effects on living organisms, including augmentation of immunological functions (9–17), prevention and cure of diseases (18–22), and prolongation of life span (23–25). Low-dose radiation activates immune activity in wild-type and lpr mice as measured by proliferation of spleen cells to mitogens (11, 13) and modifies the latency for radiation-induced myeloid leukemia in CBA/H mice (26). Furthermore, low-dose radiation increases the latency of spontaneous lymphomas and spinal osteosarcomas in cancer-prone, radiation-sensitive Trp53 heterozygous mice (27). Recently, Ootsuyama et al. (28) reported that MRL-gld/gld mice, which carry a mutation within the Fas ligand (FasL) and autoimmune diseases similar to lpr mice (29–35), showed remission of the diseases after low-dose irradiation, in parallel with a decrease in the population of splenic CD42 CD82 T cells. They attributed this effect to augmentation of apoptosis. We show in this report that both the amelioration of multiple severe diseases throughout the body and the prolongation of life span by chronic low-dose-rate irradiation involve the activation of the immune system. MATERIALS AND METHODS Materials and methods used were essentially as described previously (8). Animals Female MRL/MpJUmmCrj-lpr/lpr mice (5 weeks old) were purchased from Charles River Japan, Inc., Yokohama, and were kept under specificpathogen- free conditions. All the animals were maintained on a light schedule from 7:00 to 19:00 and were fed a standard mouse diet CE-2 (Clea Japan, Inc., Tokyo) with water allowed ad libitum. The study was reviewed by the Institutional Animal Care and Use Committee, and the mice were treated in accordance with governmental guidelines and the guidelines of the Central Research Institute of Electric Power Industry (CRIEPI). The urine was tested weekly using commercial test strips (Wako Pure PROLONGATION OF LIFE SPAN BY WHOLE-LIFE IRRADIATION 419 FIG. 1. Prolongation of life span in MRL-lpr/lpr mice with chronic low-dose-rate g radiation. a: Nonirradiated controls. The survival results obtained in our previous observation (8) were combined. b: Mice irradiated with 0.35 mGy/h for 5 weeks. The observations in our previous report (8) were extended to the entire life span. c: Mice irradiated with 0.35 mGy/h for the whole life span. d: Mice irradiated with 1.2 mGy/h for 5 weeks. Observation until day 203 (8) was extended to the entire life span. e: Mice irradiated with 1.2 mGy/h for 521 days. *P , 0.0001 compared to controls. P , 0.0001 for b 2 c, d 2 e, b 2 d, c 2 e and b 2 e. Chemical Industries, Ltd., Osaka). Specimens of organs and tissues were fixed in 10% formalin/PBS solution. After embedding in paraffin, 3-mmthick sections were prepared, stained with hematoxylin and eosin (H&E), and examined histologically under a microscope. Irradiation Mice were irradiated in a clean irradiation room equipped with a 370 GBq 137Cs g-ray source (Chiyoda Technol Co., Tokyo). The dose rates were 1.2 and 0.35 mGy/h, respectively, at a distance of 5 and 10 m from the source as measured by an ionization chamber and glass dosimeter in 2000. The tissue dose rates measured with a glass dosimeter embedded in a mouse’s abdomen were 0.95 and 0.30 mGy/h at each point as measured in a separate experiment (36). The mice were irradiated continuously except for 1 h in the morning on weekdays. Group a mice (n 5 20) were unirradiated and were treated in the same way as the unirradiated controls (n 5 12) in the previous experiment (8). Group b mice (n 5 12) were irradiated with 0.35 mGy/h for 5 weeks, which is the same treatment used in the previous study (8), but were observed for the entire life span. Group c mice (n 5 20) were irradiated at 0.35 mGy/h for the whole life span. Group d mice (n 5 12) were irradiated with 1.2 mGy/h for 5 weeks, which is the same treatment used in the previous study (8), but were observed for the entire life span. Group e mice (n 5 20) were irradiated at 1.2 Gy for 521 days. The ages of the mice at the start of continuous irradiation were 5 weeks for Groups c and e, and 7 weeks for Groups b and d. Analysis of Immune Cell Status Single cell suspensions were prepared from the thymus and spleen in RPMI 1640 medium supplemented with 10% FCS. Immunological changes were examined in terms of cell surface molecules after analysis of cell populations. The cell surface functional molecules CD3, CD4, CD8, CD45R/B220 (B220) and Fas and activation-marker molecule CD40 were analyzed by flow cytometry. For immunofluorescence studies, the following monoclonal antibodies (mAbs) from BD PharMingen (San Diego, CA) were used: purified 2.4G2 mAb (rat IgG2b, k), which recognizes CD16/32 (Fcg III/II receptor) for FcBlock; fluorescein isothiocyanate (FITC)-conjugated 17A2 mAb (rat IgG2b, k), which recognizes CD3 molecular complex; FITC-conjugated GK1.5 mAb (rat IgG2b, k), CD4; R-phycoerythrin (R-PE)-conjugated 53- 6.7 mAb (rat IgG2a, k), CD8a (Ly-2); FITC-conjugated and R-PE-conjugated RA3-6B2 mAbs (rat IgG2a, k), CD45R/B220 (B220); R-PE-conjugated Jo2 mAb (hamster IgG, group 2, l), Fas (CD95); R-PE-conjugated 3/23 mAb (rat IgG2a, k), CD40. The single cell suspensions of thymus and spleen were preincubated with unlabeled anti-Fcg receptor mAbs for 10 min at 48C to avoid nonspecific Fc-mediated binding of the labeled antibodies. The cells were stained with FITC-conjugated mAbs and R-PE-conjugated mAbs simultaneously for 20 min at 48C. After they were washed, the stained samples were analyzed using an EPICS XL flow cytometry system (Beckman Coulter, Inc., Fullerton, CA). Statistical Analysis The Kaplan-Meier method was used to estimate the survival curves and the significance of differences between the percentages of survival was evaluated by the logrank test. The significance of differences in the percentages of mice with lymphadenopathy or proteinuria was evaluated by the x2 test. The weights of spleen and inguinal lymph nodes and the percentages of cell populations are presented as means 6 standard errors of the mean. The statistical significance of the differences was evaluated by the Student’s t test. RESULTS Chronic Low-Dose-Rate g Irradiation Prolonged Life Span MRL-lpr/lpr mice were continuously g-irradiated at a low dose rate of 0.35 or 1.2 mGy/h. In our previous study (8), the mice were irradiated for 5 weeks, beginning at 7 weeks of age, and observed for 203 days. In the present study, this observation period was extended to the entire life span, and continuous irradiation was extended throughout the whole life span for the 0.35-mGy/h group and for the 1.2-mGy/h group. Figure 1 shows survival of these groups plotted as a function of time and clearly shows that the life span of the lpr mice was significantly prolonged by continuous low-dose-rate irradiation in a dose-rate-dependent manner (P , 0.0001 for b 2 c, d 2 e, b 2 d, c 2 e and b 2 e). Table 1 shows the 50% survival time for the lpr mice irradiated continuously at two different low dose rates for two different periods together with changes in immunological parameters at the two low dose rates. The prolongations of survival are in parallel with immunological activities, as discussed later. Chronic Low-Dose-Rate g Irradiation Suppressed Lymphadenopathy, Splenomegaly, and Kidney and Brain- Central Nervous System Syndromes In parallel with the survival study, we examined the effects of continuous low-dose-rate g irradiation on specific diseases in the lpr mice, i.e. lymphadenopathy (Figs. 2A and 3B), proteinuria (Fig. 2B) and splenomegaly (Fig. 3A). In the nonirradiated controls, these symptoms developed over time. However, the symptoms were suppressed re420 INA AND SAKAI TABLE 1 50% Survival Time and Immunological Parameters for MRL-lpr/lpr Mice g-Irradiated at Low Dose Rates Group Dose rate (mGy/h) Irradiation period (week) 50% survival time (days) Relative immunological parametersa Thymus CD41 CD81 T cells Spleen CD81 T cells CD31 B2201 cells B2201 CD401 cells a 0 0 134b 1.0 1.0 1.0 1.0 b 0.35 5 159c ― ― ― ― c 0.35 35 228 1.1 1.9 0.62 0.81 d 1.2 5 301d ― ― ― ― e 1.2 74 502 1.2 2.2 0.45 0.37 a Measured in 12 mice in each group irradiated for 7 weeks beginning at 5 weeks of age and killed at 12 weeks of age. b Identical results from previous (8) and present studies. c Previous observation (8). d Previous observation (8) was extended to whole life. FIG. 2. Development of lymphadenopathy and proteinuria in MRLlpr/ lpr mice over time and its suppression by chronic g irradiation at low dose rates beginning at 5 weeks of age. Upper curves: nonirradiated mice. Middle curves: mice irradiated with 0.35 mGy/h. Lower curves: mice irradiated with 1.2 mGy/h. n 5 10 per point. Panel A: Mice with palpable inguinal lymph nodes. *P , 0.01, **P , 0.001, ***P , 0.0001 compared to controls. Panel B: Mice with proteinuria over 30 mg/dl. *P , 0.05, **P , 0.01, ***P , 0.001, ****P , 0.0001 compared to controls. markably by continuous low-dose-rate irradiation, and this suppression was dependent on the dose rate. Furthermore, the severity of proteinuria in each mouse was reduced by the low-dose-rate irradiation (data not shown). We previously demonstrated a remarkable amelioration of glomerulonephritis in the kidney and hemorrhage and inflammation in the brain in low-dose-rate-irradiated lpr mice using histological sections (8). In this study, we again observed histological amelioration of disease-specific damage to the kidney and the brain and also so observed that the whole brain, central nervous system, liver, intestine, lung, joints, blood vessels and skin in the low-dose-rateirradiated mice appeared much less affected compared to the nonirradiated group (data not shown). Chronic Low-Dose-Rate g Irradiation Increased Immune Activities CD41 CD81 T cells in the thymus are very important for maturation of the immune system in the entire body. The population of these cells is small in lpr mice. However, chronic low-dose-rate irradiation significantly increased these cell populations (Fig. 4 and Table 1). CD81 T cells are important for the biological defense system. The cell population in the spleen was low in the nonirradiated mice. However, chronic low-dose-rate irradiation increased the cell population significantly (Fig. 5A and Table 1). CD31 CD45R/B2201 (CD31 B2201) cells are cells that attack the mouse’s own organs and tissues (37). CD45R/B2201 CD401 (B2201 CD401) cells are characteristic in autoimmune diseases (38). The percentages of these cells in spleens are very high in the lpr mice. However, these high levels of abnormal cells were significantly decreased in the chronic low-dose-rate-irradiated lpr mice. Furthermore, suppressions of the abnormal cell populations were dependent on the dose rate (Fig. 5B, C and Table 1). Also noted was a tendency for an increase in the percentage of CD41 T cells in the spleen after chronic irradiation at a low dose rate, indicating augmentation of the immune system in the whole body. Furthermore, no changes in the total cell numbers in the bone marrow, no injury in the intestine, and no radiation-induced lymphomas were observed (data not shown). DISCUSSION The present report summarizes results obtained from lifelong observation of MRL-lpr/lpr mice that were continuPROLONGATION OF LIFE SPAN BY WHOLE-LIFE IRRADIATION 421 FIG. 3. Suppression of splenomegaly and lymphadenopathy in MRLlpr/ lpr mice by chronic g irradiation at low dose rates from 5 weeks of age. Upper curves: nonirradiated mice. Middle curves: mice irradiated with 0.35 mGy/h. Lower curves: mice irradiated with 1.2 mGy/h. n 5 12 per point. Panel A: The weights of the spleen. *P , 0.01, **P , 0.001, ***P , 0.0001 compared to controls. Panel B: The weights of the inguinal lymph nodes. *P , 0.0001 compared to controls. FIG. 5. Changes in immune cell populations in the spleens of MRLlpr/ lpr mice by chronic g irradiation at low dose rates from 5 weeks of age. Solid curves: mice irradiated with 1.2 mGy/h. Dashed curves: mice irradiated with 0.35 mGy/h. Dotted curves: nonirradiated mice. n 5 12 per point. Panel A: CD81 T cells. *P , 0.001, **P , 0.0001 compared to controls. Panel B: CD31 B2201 cells. *P , 0.01, **P , 0.0001 compared to controls. Panel C: B2201 CD401 cells. *P , 0.05, **P , 0.01, ***P , 0.001, ****P , 0.0001 compared to controls. FIG. 4. Increases in normal immune cell populations in the thymuses of MRL-lpr/lpr mice by chronic g irradiation at low dose rates from 5 weeks of age. Upper curve: mice irradiated with 1.2 mGy/h. Middle curve: mice irradiated with 0.35 mGy/h. Lower curve: nonirradiated mice. n 5 12 per point. *P , 0.01, **P , 0.001, ***P , 0.0001 compared to controls. ously g-irradiated at low dose rates. Previous observations (8) already indicated a prolongation of the life span of lpr mice accompanied by amelioration of disease and immune activation after 5 weeks of irradiation at low dose rates. The present study includes whole-life observation of these mice and provides information on the more profound effects of life-long irradiation that accompany immune activation over time. In this study, whole-life g irradiation at low dose rates significantly prolonged the life span of lpr mice. The radiation suppressed total-body lymphadenopathy, splenomegaly and proteinuria together with kidney and brain-central nervous system syndromes, which are possible causes of death in these animals. It significantly increased CD41 CD81 T cells in the thymus and CD81 T cells in the spleen, 422 INA AND SAKAI which indicates activation of the immune system, and decreased the representative abnormal cell populations, i.e. CD31 B2201 cells and B2201 CD401 cells. The CD31 B2201 cells are autoreactive to the mouse’s own organs and tissues throughout the body. The B2201 CD401 cells are thought to be the major cell population in the progression of autoimmune diseases. We believe that the immunological activations in the chronically low-dose-rate-irradiated mice are highly important in disease suppression in lpr mice. We believe that the increase in CD81 T cells may bring about an effective attack against abnormal cells and suppress the disease-specific abnormal cell populations, i.e. CD31 B2201 cells and B2201 CD401 cells. The B2201 CD401 cells in the lowdose- rate-irradiated lpr mice decreased to the levels of those cells in normal mice without the lpr mutation, although the CD31 B2201 cells still existed. We therefore consider that these decreases explain the amelioration of severe autoimmune diseases throughout the body. It appeared that irradiation with 1.2 mGy/h was more effective than irradiation with 0.35 mGy/h in the prolongation of life and the enhancement of the immune system. We think it is necessary to irradiate mice at a higher dose rate for the full development of immunity and for suppression of these diseases. An optimum dose rate may exist beyond the dose-rate range employed in this study. Furthermore, lpr mice spontaneously develop inflammation in the cerebral vessels, meninges and choroid plexus (5, 6). Our previous observations (8) showed that chronic low-dose-rate irradiation significantly suppressed cerebral and central nervous inflammation and hemorrhage in lpr mice, an observation that was confirmed in the present study (data not shown). Pathways to apoptosis through non-Fas-mediated mechanisms may exist in chronically low-dose-rate-irradiated lpr mice. When Ootsuyama et al. (28) observed the effects of low-dose radiation in the amelioration of disease in MRLgld/ gld mice, which have a mutation in the Fas ligand, they explained the effects of such radiation by recovery of the apoptotic activity as indicated in spleen cells. However, in this study, we found no significant difference between the nonirradiated control mice and the low-dose-rate-irradiated mice in terms of the percentage of thymocytes stained by propidium iodide, trypan blue and erythrosin B, respectively, and the expression of phosphatidylserine on the thymocytes. Furthermore, there was no expression of Fas, the apoptosis-regulating molecule, on the thymocytes, splenocytes and lymph node cells (data not shown). The difference between lpr and gld mutations or between the dose rates of radiation in the experiments of Ootsuyama et al. (28) and our experiments may explain the difference in apoptosis. Therefore, we consider the immunological modification in the low-dose-rate-irradiated mice to be the primary reason for increased survival through amelioration of diseases, although changes in non-Fas-mediated apoptotic activity might be indirectly involved. Finally, it is intriguing that immunological activation by low-dose-rate irradiation may be effective in suppressing tumor induction. Indeed, we have found suppression of radiation- induced thymic lymphomas in C57BL/6 mice to less than half (40%) by continuous low-dose-rate g irradiation under conditions identical to those in the present study (39). In summary, we found ameliorations of multiple severe diseases, including total-body lymphadenopathy, splenomegaly and serious autoimmune diseases throughout the body, including kidney and brain-central nervous system syndromes, in MRL-lpr/lpr mice chronically irradiated at low dose rates. The life span of the mice was prolonged remarkably in parallel with the activation of the immune system without severe tissue damage. Thus chronic lowdose- rate irradiation is expected to be useful in the treatment of certain lymphomas and autoimmune diseases. ACKNOWLEDGMENTS We thank Ms. Ikuno Suzuki and Mr. Takeshi Oda for their excellent technical assistance and animal care. We are also indebted to Dr. Hiroshi Tanooka for his critical reading of this manuscript and to Dr. Harumi Ohyama for her helpful discussions. Received: March 25, 2004; accepted: October 26, 2004 REFERENCES 1. E. D. Murphy and J. B. Roths, Autoimmunity and lymphoproliferation: Induction by mutant gene lpr, and acceleration by a male-associated factor in strain BXSB mice. In Genetic Control of Autoimmune Disease (N. R. Rose, P. E. Bigazzi and N. L. Warner, Eds.), pp. 207–221. Elsevier, Amsterdam, 1978. 2. B. S. Andrews, R. A. Eisenberg, A. N. Theofilopoulos, S. Izui, C. B. Wilson, P. J. McConahey, E. D. Murphy, J. B. Roths and F. J. Dixon, Spontaneous murine lupus-like syndromes: Clinical and immunopathological manifestations in several strains. J. Exp. Med. 148, 1198–1215 (1978). 3. E. D. Murphy and J. B. Roths, A Y chromosome associated factor in strain BXSB producing accelerated autoimmunity and lymphoproliferation. Arthritis Rheum. 22, 1188–1194 (1979). 4. J. L. Chu, J. Drappa, A. Parnassa and K. B. Elkon, The defect in Fas mRNA expression in MRL/lpr mice is associated with insertion of the retrotransposon, ETn. J. Exp. Med. 178, 723–730 (1993). 5. E. L. Alexander, E. D. Murphy, J. B. Roths and G. E. Alexander, Congenic autoimmune murine models of central nervous system disease in connective tissue disorders. Ann. Neurol. 14, 242–248 (1983). 6. E. L. Alexander, C. Moyer, G. S. Travlos, J. B. Roths and E. D. Murphy, Two histopathologic types of inflammatory vascular disease in MRL/Mp autoimmune mice. Model for human vasculitis in connective tissue disease. Arthritis Rheum. 28, 1146–1155 (1985). 7. T. Watanabe, Y. Sakai, S. Miyawaki, A. Shimizu, O. Koiwai and K. Ohno, A molecular genetic linkage map of mouse chromosome 19, including the lpr, Ly-44, and Tdt genes. Biochem. Genet. 29, 325– 335 (1991). 8. Y. Ina and K. Sakai, Prolongation of life span associated with immunological modification by chronic low-dose-rate irradiation in MRL-lpr/lpr mice. Radiat. Res. 161, 168–173 (2004). 9. R. E. Anderson and I. Lefkovits, In vitro evaluation of radiationinduced augmentation of the immune response. Am. J. Pathol. 97, 456–472 (1979). 10. S. Z. Liu, W. H. Liu and J. B. Sun, Radiation hormesis: Its expression in the immune system. Health Phys. 52, 579–583 (1987). PROLONGATION OF LIFE SPAN BY WHOLE-LIFE IRRADIATION 423 11. S. J. James and T. Makinodan, T cell potentiation in normal and autoimmune-prone mice after extended exposure to low doses of ionizing radiation and/or caloric restriction. Int. J. Radiat. Biol. 53, 137– 152 (1988). 12. S. Z. Liu, Radiation hormesis. A new concept in radiological science. Chin. Med. J. 102, 750–755 (1989). 13. S. J. James, S. M. Enger, W. J. Peterson and T. Makinodan, Immune potentiation after fractionated exposure to very low doses of ionizing radiation and/or caloric restriction in autoimmune-prone and normal C57Bl/6 mice. Clin. Immunol. Immunopathol. 55, 427–437 (1990). 14. J. Matsubara, V. Turcanu, P. Poindron and Y. Ina, Immune effects of low-dose radiation: Short-term induction of thymocyte apoptosis and long-term augmentation of T-cell-dependent immune responses. Radiat. Res. 153, 332–338 (2000). 15. S. Kojima, S. Matsumori, H. Ishida and K. Yamaoka, Possible role of elevation of glutathione in the acquisition of enhanced proliferation of mouse splenocytes exposed to small-dose g-rays. Int. J. Radiat. Biol. 76, 1641–1647 (2000). 16. S. Kojima, H. Ishida, M. Takahashi and K. Yamaoka, Elevation of glutathione induced by low-dose gamma rays and its involvement in increased natural killer activity. Radiat. Res. 157, 275–280 (2002). 17. Y. Ina and K. Sakai, Immunological network activation by low-dose rate irradiation: Analysis of cell populations and cell surface molecules in whole body irradiated mice. In Molecular Mechanisms for Radiation-induced Cellular Response and Cancer Development (K. Tanaka, T. Takabatake, K. Fujikawa, T. Matsumoto and F. Sato, Eds.), pp. 227–233. Art Press, Aomori, 2003. 18. H. J. Dobbs, A. Barrett, A. Y. Rostom and M. J. Peckham, Totalbody irradiation in advanced non-Hodgkin’s lymphoma. Br. J. Radiol. 54, 878–881 (1981). 19. P. Jacobs and H. S. King, A randomized prospective comparison of chemotherapy to total body irradiation as initial treatment for the indolent lymphoproliferative diseases. Blood 69, 1642–1646 (1987). 20. R. N. Shen, L. Lu, H. E. Kaiser and H. E. Broxmeyer, Murine AIDS cured by low dosage total body irradiation. Adv. Exp. Med. Biol. 407, 451–458 (1997). 21. S. Hashimoto, H. Shirato, M. Hosokawa, T. Nishioka, Y. Kuramitsu, K. Matsushita, M. Kobayashi and K. Miyasaka, The suppression of metastases and the change in host immune response after low-dose total-body irradiation in tumor-bearing rats. Radiat. Res. 151, 717– 724 (1999). 22. M. Takahashi, S. Kojima, K. Yamaoka and E. Niki, Prevention of type I diabetes by low-dose gamma irradiation in NOD mice. Radiat. Res. 154, 680–685 (2000). 23. E. Lorenz, J. W. Hollcroft, E. Miller, C. C. Congdon and R. Schweisthal, Long-term effects of acute and chronic irradiation in mice. I. Survival and tumor incidence following chronic irradiation of 0.11 r per day. J. Natl. Cancer Inst. 15, 1049–1058 (1955). 24. H. S. Ducoff, Form of the increased longevity of Tribolium after Xirradiation. Exp. Gerontol. 10, 189–193 (1975). 25. M. Mine, Y. Okumura, M. Ichimaru, T. Nakamura and S. Kondo, Apparently beneficial effect of low to intermediate doses of A-bomb radiation on human lifespan. Int. J. Radiat. Biol. 58, 1035–1043 (1990). 26. R. E. J. Mitchel, J. S. Jackson, R. A. McCann and D. R. Boreham, The adaptive response modifies latency for radiation-induced myeloid leukemia in CBA/H mice. Radiat. Res. 152, 273–279 (1999). 27. R. E. J. Mitchel, J. S. Jackson, D. P. Morrison and S. M. Carlisle, Low doses of radiation increase the latency of spontaneous lymphomas and spinal osteosarcomas in cancer-prone, radiation-sensitive Trp53 heterozygous mice. Radiat. Res. 159, 320–327 (2003). 28. A. Ootsuyama, R. Okazaki and T. Norimura, Effect of extended exposure to low-dose radiation on autoimmune diseases of immunologically suppressed MRL/MpTn-gld/gld mice. J. Radiat. Res. 44, 243–247 (2003). 29. R. Watanabe-Fukunaga, C. I. Brannan, N. G. Copeland, N. A. Jenkins and S. Nagata, Lymphoproliferation disorder in mice explained by defects in Fas antigen that mediates apoptosis. Nature 356, 314–317 (1992). 30. J. B. Roths, E. D. Murphy and E. M. Eicher, A new mutation, gld, that produces lymphoproliferation and autoimmunity in C3H/HeJ mice. J. Exp. Med. 159, 1–20 (1984). 31. W. F. Davidson, F. J. Dumont, H. G. Bedigian, B. J. Fowlkes and H. C. Morse, III, Phenotypic, functional, and molecular genetic comparisons of the abnormal lymphoid cells of C3H-lpr/lpr and C3H-gld/ gld mice. J. Immunol. 136, 4075–4084 (1986). 32. M. Nose, M. Nishimura and M. Kyogoku, Analysis of granulomatous arteritis in MRL/Mp autoimmune disease mice bearing lymphoproliferative genes. The use of mouse genetics to dissociate the development of arteritis and glomerulonephritis. Am. J. Pathol. 135, 271– 280 (1989). 33. P. L. Cohen and R. A. Eisenberg, Lpr and gld: Single gene models of systemic autoimmunity and lymphoproliferative disease. Annu. Rev. Immunol. 9, 243–269 (1991). 34. M. L. Watson, P. D’Eustachio, B. A. Mock, A. D. Steinberg, H. C. Morse, III, R. J. Oakey, T. A. Howard, J. M. Rochelle and M. F. Seldin, A linkage map of mouse chromosome 1 using an interspecific cross segregating for the gld autoimmunity mutation. Mamm. Genome 2, 158–171 (1992). 35. T. Takahashi, M. Tanaka, C. I. Brannan, N. A. Jenkins, N. G. Copeland, T. Suda and S. Nagata, Generalized lymphoproliferative disease in mice, caused by a point mutation in the Fas ligand. Cell 76, 969–976 (1994). 36. Y. Hoshi, T. Nomura, T. Oda, T. Iwasaki, K. Fujita, T. Ishikawa, A. Kato, K. Sakai, H. Tanooka and T. Yamada, Application of a newly developed photoluminescence glass dosimeter for measuring the absorbed dose in individual mice exposed to low-dose rate 137Cs g-rays. J. Radiat. Res. 41, 129–137 (2000). 37. C. Diaz-Gallo and V. R. Kelley, Self-regulation of autoreactive kidney- infiltrating T cells in MRL-lpr nephritis. Kidney Int. 44, 692– 699 (1993). 38. C. Mohan, Y. Shi, J. D. Laman and S. K. Datta, Interaction between CD40 and its ligand gp39 in the development of murine lupus nephritis. J. Immunol. 154, 1470–1480 (1995). 39. Y. Ina, H. Tanooka, T. Yamada and K. Sakai, Suppression of thymic lymphoma induction by life-long low-dose-rate irradiation accompanied by immune activation in C57BL/6 mice. Radiat. Res. 163, 153–158 (2005). |
|