Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Anticancer Res ; 43(6): 2551-2559, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37247915

RESUMEN

BACKGROUND/AIM: Arsenite is a radiosensitizer of glioma cells both in vitro and in vivo; however, the underlying mechanism of action is unclear. Radiosensitizers specific for p53-deficient tumors are a promising adjunct to radiotherapy because, unlike normal cells, many tumor cells lack p53. Previously, we demonstrated that arsenite sensitizes the p53-deficient glioma cell line U87MG-E6 to X-rays. MATERIALS AND METHODS: Using flowcytometry, we expand these findings to p53-proficient U87MG cells exposed to heavy ion beams, including carbon and iron ions. RESULTS: Arsenite sensitized U87MG-E6, but not U87MG, cells to heavy ion beams and X-rays. Cell cycle analysis indicated that sensitization of U87MG-E6 was related to an increase in the percentage of cells in the late S/G2/M phases after combined treatment with arsenite, especially when carbon ion beams were used. Induction of γH2AX was significant in U87MG-E6, but not in U87MG, cells after irradiation with carbon ion beams plus arsenite. CONCLUSION: Arsenite sensitizes cells by increasing the percentage of cells in the late S/G2/M phases after irradiation, possibly via inhibition of DNA repair in the context of p53 deficiency. The findings provide information that may be useful for the development of advanced radiotherapy protocols.


Asunto(s)
Arsenitos , Glioma , Humanos , Arsenitos/farmacología , Carbono , Carmustina , Línea Celular Tumoral , Supervivencia Celular , Glioma/patología , Proteína p53 Supresora de Tumor/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-36155141

RESUMEN

High atomic number and energy (HZE) particles such as iron-56 (Fe) ions are a major contributor to health risks in long-term manned space exploration. The aim of this study is to understand radiation-induced differential genotoxic effects between HZE particles and low linear energy transfer (LET) photons. C57BL/6J Jms female mice of 8 weeks old were exposed to total body irradiation of accelerated Fe-particles with a dose ranging from 0.1 to 3.0 Gy or of X-rays with a dose ranging from 0.1 to 5.0 Gy. Chromosomal aberrations (CAs) in splenocytes were examined by fluorescence in situ hybridization at 1- and 2-months after exposure. Clonal expansions of cells with CAs were found to be induced only by X-rays but not by Fe-particles. Dose-dependent increase in the frequencies of stable-type CAs was observed at 1- as well as 2-months after exposure to both radiation types. The frequencies of stable-type CAs in average were much higher in mice exposed to X-rays than those to Fe-particles and did not change significantly between 1- and 2-months after exposure to both radiation types. On the other hand, the frequencies of unstable-type CAs induced by X-rays and Fe-particles were not much different, and they appeared to decrease with time from 1- to 2-months after exposure. These results suggested that larger fraction of stable-type CAs induced by Fe-particles might be non-transmissible than those by X-rays because of some associating lethal alterations on themselves or on other chromosomes in the same cells and that these cells might be removed by 1-month after Fe-TBI. We also demonstrated that exposure to Fe-particles induced insertions at relatively higher frequency to other stable-type CAs than X-rays. Our findings suggest that insertions can be used as indicators of past exposure to high-LET particle radiation.


Asunto(s)
Hierro , Bazo , Animales , Aberraciones Cromosómicas , Relación Dosis-Respuesta en la Radiación , Femenino , Hibridación Fluorescente in Situ , Iones , Hierro/toxicidad , Ratones , Ratones Endogámicos C57BL , Rayos X
3.
J Radiat Res ; 63(4): 602-608, 2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35726341

RESUMEN

Psychological stress affects health. Radiation workers in the medical field or astronauts living in space have possible risks of exposure to radiation, and psychological stress is considered to be easily induced in them due to activities performed in small areas or stress conditions. The impact of psychological stress on the effects of radiation was evaluated in senescence-accelerated mouse prone 10 (SAMP10) mice and ddY mice using a confrontational housing model, which makes dominant and subordinate mice in a cage live together without severe quarrel. Mice of ddY and SAMP10 have been previously demonstrated to be influenced in terms of acute and late effects, respectively, under psychological stress by this model. In SAMP10 mice, irradiation with 4 Gy induced the death of irradiated mice under psychological stress. In ddY mice, irradiation with 5 Gy X-rays alone had almost no effect on the mouse survival, but irradiation in conditions of psychological stress promoted acute death of irradiated mice. In addition, hypocellular bone marrow was also observed histopathologically in irradiated ddY mice under stress. Psychological stress may promote damage caused by radiation through modulation of radio-sensitivity in bone marrow in mice. This model would be useful for evaluation of modulation of radiation-induced various effects by psychological stress.


Asunto(s)
Médula Ósea , Traumatismos por Radiación , Animales , Médula Ósea/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Ratones , Ratones Endogámicos C57BL , Modelos Teóricos , Estrés Psicológico/complicaciones , Rayos X
4.
Life (Basel) ; 12(4)2022 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-35455056

RESUMEN

Humans are exposed to both psychological stress (PS) and radiation in some scenarios such as manned deep-space missions. It is of great concern to verify possible enhanced deleterious effects from such concurrent exposure. Pioneer studies showed that chronic restraint-induced PS (CRIPS) could attenuate Trp53 functions and increase gamma-ray-induced carcinogenesis in Trp53-heterozygous mice while CRIPS did not significantly modify the effects on X-ray-induced hematopoietic toxicity in Trp53 wild-type mice. As high-linear energy transfer (LET) radiation is the most important component of space radiation in causing biological effects, we further investigated the effects of CRIPS on high-LET iron-particle radiation (Fe)-induced hematopoietic toxicity in Trp53-heterozygous mice. The results showed that CRIPS alone could hardly induce significant alteration in hematological parameters (peripheral hemogram and micronucleated erythrocytes in bone marrow) while concurrent exposure caused elevated genotoxicity measured as micronucleus incidence in erythrocytes. Particularly, exposure to either CRISP or Fe-particle radiation at a low dose (0.1 Gy) did not induce a marked increase in the micronucleus incidence; however, concurrent exposure caused a significantly higher increase in the micronucleus incidence. These findings indicated that CRIPS could enhance the deleterious effects of high-LET radiation, particularly at a low dose, on the hematopoietic toxicity in Trp53-heterozygous mice.

5.
Cancers (Basel) ; 13(15)2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34359734

RESUMEN

Hypoxia-Inducible Factor 1α (HIF-1α), which promotes cancer cell survival, is the main regulator of oxygen homeostasis. Hypoxia combined with photon and carbon ion irradiation (C-ions) stabilizes HIF-1α. Silencing HIF-1α under hypoxia leads to substantial radiosensitization of Head-and-Neck Squamous Cell Carcinoma (HNSCC) cells after both photons and C-ions. Thus, this study aimed to clarify a potential involvement of HIF-1α in the detection, signaling, and repair of DNA Double-Strand-Breaks (DSBs) in response to both irradiations, in two HNSCC cell lines and their subpopulations of Cancer-Stem Cells (CSCs). After confirming the nucleoshuttling of HIF-1α in response to both exposure under hypoxia, we showed that silencing HIF-1α in non-CSCs and CSCs decreased the initiation of the DSB detection (P-ATM), and increased the residual phosphorylated H2AX (γH2AX) foci. While HIF-1α silencing did not modulate 53BP1 expression, P-DNA-PKcs (NHEJ-c) and RAD51 (HR) signals decreased. Altogether, our experiments demonstrate the involvement of HIF-1α in the detection and signaling of DSBs, but also in the main repair pathways (NHEJ-c and HR), without favoring one of them. Combining HIF-1α silencing with both types of radiation could therefore present a potential therapeutic benefit of targeting CSCs mostly present in tumor hypoxic niches.

6.
Radiat Res ; 196(1): 100-112, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33901294

RESUMEN

Astronauts can develop psychological stress (PS) during space flights due to the enclosed environment, microgravity, altered light-dark cycles, and risks of equipment failure or fatal mishaps. At the same time, they are exposed to cosmic rays including high atomic number and energy (HZE) particles such as iron-56 (Fe) ions. Psychological stress or radiation exposure can cause detrimental effects in humans. An earlier published pioneering study showed that chronic restraint-induced psychological stress (CRIPS) could attenuate Trp53 functions and increase carcinogenesis induced by low-linear energy transfer (LET) γ rays in Trp53-heterozygous (Trp53+/-) mice. To elucidate possible modification effects from CRIPS on high-LET HZE particle-induced health consequences, Trp53+/- mice were received both CRIPS and accelerated Fe ion irradiation. Six-week-old Trp53+/- C57BL/6N male mice were restrained 6 h per day for 28 consecutive days. On day 8, they received total-body Fe-particle irradiation (Fe-TBI, 0.1 or 2 Gy). Metaphase chromosome spreads prepared from splenocytes at the end of the 28-day restraint regimen were painted with the fluorescence in situ hybridization (FISH) probes for chromosomes 1 (green), 2 (red) and 3 (yellow). Induction of psychological stress in our experimental model was confirmed by increase in urinary corticosterone level on day 7 of restraint regimen. Regardless of Fe-TBI, CRIPS reduced splenocyte number per spleen at the end of the 28-day restraint regimen. At 2 Gy, Fe-TBI alone induced many aberrant chromosomes and no modifying effect was detected from CRIPS on induction of aberrant chromosomes. Notably, neither Fe-TBI at 0.1 Gy nor CRIPS alone induced any increase in the frequency of aberrant chromosomes, while simultaneous exposure resulted in a significant increase in the frequency of chromosomal exchanges. These findings clearly showed that CRIPS could enhance the frequency of chromosomal exchanges induced by Fe-TBI at a low dose of 0.1 Gy.


Asunto(s)
Aberraciones Cromosómicas , Heterocigoto , Hierro/administración & dosificación , Restricción Física , Estrés Fisiológico , Proteína p53 Supresora de Tumor/genética , Animales , Relación Dosis-Respuesta en la Radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/metabolismo , Bazo/patología , Bazo/efectos de la radiación
7.
Dose Response ; 19(1): 1559325820982166, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33628149

RESUMEN

Radioadaptive response (RAR) describes a phenomenon in a variety of in vitro and in vivo systems that a low-dose of priming ionizing radiation (IR) reduces detrimental effects of a subsequent challenge IR at higher doses. Among in vivo investigations, studies using the mouse RAR model (Yonezawa Effect) showed that RAR could significantly extenuate high-dose IR-induced detrimental effects such as decrease of hematopoietic stem cells and progenitor cells, acute radiation hematopoietic syndrome, genotoxicity and genomic instability. Meanwhile, it has been demonstrated that diet intervention has a great impact on health, and dietary restriction shows beneficial effects on numerous diseases in animal models. In this work, by using the mouse RAR model and mild dietary restriction (MDR), we confirmed that combination of RAR and MDR could more efficiently reduce radiogenotoxic damage without significant change of the RAR phenotype. These findings suggested that MDR may share some common pathways with RAR to activate mechanisms consequently resulting in suppression of genotoxicity. As MDR could also increase resistance to chemotherapy and radiotherapy in normal cells, we propose that combination of MDR, RAR, and other cancer treatments (i.e., chemotherapy and radiotherapy) represent a potential strategy to increase the treatment efficacy and prevent IR risk in humans.

8.
Sci Rep ; 10(1): 21357, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33288855

RESUMEN

DNA double-strand breaks (DSBs) induced by photon irradiation are the most deleterious damage for cancer cells and their efficient repair may contribute to radioresistance, particularly in hypoxic conditions. Carbon ions (C-ions) act independently of the oxygen concentration and trigger complex- and clustered-DSBs difficult to repair. Understanding the interrelation between hypoxia, radiation-type, and DNA-repair is therefore essential for overcoming radioresistance. The DSBs signaling and the contribution of the canonical non-homologous end-joining (NHEJ-c) and homologous-recombination (HR) repair pathways were assessed by immunostaining in two cancer-stem-cell (CSCs) and non-CSCs HNSCC cell lines. Detection and signaling of DSBs were lower in response to C-ions than photons. Hypoxia increased the decay-rate of the detected DSBs (γH2AX) in CSCs after photons and the initiation of DSB repair signaling (P-ATM) in CSCs and non-CSCs after both radiations, but not the choice of DSB repair pathway (53BP1). Additionally, hypoxia increased the NHEJ-c (DNA-PK) and the HR pathway (RAD51) activation only after photons. Furthermore, the involvement of the HR seemed to be higher in CSCs after photons and in non-CSCs after C-ions. Taken together, our results show that C-ions may overcome the radioresistance of HNSCC associated with DNA repair, particularly in CSCs, and independently of a hypoxic microenvironment.


Asunto(s)
Hipoxia de la Célula/fisiología , Roturas del ADN de Doble Cadena/efectos de la radiación , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Ciclo Celular/genética , Ciclo Celular/efectos de la radiación , Hipoxia de la Célula/genética , Línea Celular Tumoral , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Técnica del Anticuerpo Fluorescente , Genotipo , Radioterapia de Iones Pesados , Humanos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/efectos de la radiación , Carcinoma de Células Escamosas de Cabeza y Cuello/radioterapia , Microambiente Tumoral/genética , Microambiente Tumoral/efectos de la radiación , Proteína p53 Supresora de Tumor/genética , Rayos X
9.
Radiother Oncol ; 150: 253-261, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32717360

RESUMEN

BACKGROUND AND PURPOSE: High-grade chondrosarcomas are chemo- and radio-resistant cartilage-forming tumors of bone that often relapse and metastase. Thus, new therapeutic strategies are urgently needed. MATERIAL AND METHODS: Chondrosarcoma cells (CH-2879) were exposed to carbon-ion irradiation, combined with miR-34 mimic and/or rapamycin administration. The effects of treatment on cancer stem cells, stemness-associated phenotype, radioresistance and tumor-initiating properties were evaluated. RESULTS: We show that high-grade chondrosarcoma cells contain a population of radioresistant cancer stem cells that can be targeted by a combination of carbon-ion therapy, miR-34 mimic administration and/or rapamycin treatment that triggers FOXO3 and miR-34 over-expression. mTOR inhibition by rapamycin triggered FOXO3 and miR-34, leading to KLF4 repression. CONCLUSION: Our results show that particle therapy combined with molecular treatments effectively controls cancer stem cells and may overcome treatment resistance of high-grade chondrosarcoma.


Asunto(s)
Neoplasias Óseas , Condrosarcoma , MicroARNs , Neoplasias Óseas/radioterapia , Carbono , Línea Celular Tumoral , Condrosarcoma/genética , Condrosarcoma/terapia , Terapia Combinada , Humanos , Iones , Factor 4 Similar a Kruppel , MicroARNs/genética , Recurrencia Local de Neoplasia , Células Madre Neoplásicas , Serina-Treonina Quinasas TOR
10.
Med Sci Monit ; 25: 8920-8927, 2019 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-31760404

RESUMEN

BACKGROUND Several factors found in foods are beneficial to human health and they may contribute to radiation protection. Taking food factors could be an easy way to reduce the effects of radiation after nuclear accidents, as well as secondary radiation risks after cancer radiotherapy or space missions. Here, diallyl disulfide (DADS), a component of garlic oil, was studied for its ability to mitigate radiation damage. MATERIAL AND METHODS We investigated the effects of DADS on micronucleus (MN) formation and apoptosis in HepG2 cells by use of 4-Gy X-ray irradiation. We also assessed the effects of DADS on radiation damage in vivo by evaluating MN formation in bone marrow cells in mice (BALB/c, 8-week-old females) after oral intake of DADS prior to irradiation with 4 Gy. Several tissue effects were also investigated. RESULTS The presence of DADS inhibited MN formation, whereas DADS had no influence on the radiation-induced inhibition of cell cycle progression in HepG2 cells. An increase in apoptosis in HepG2 cells was induced after irradiation, and this effect was stronger in the presence of DADS than in its absence. In mice, when DADS was administered daily for 3 days prior to irradiation, MN formation in irradiated mice was decreased. The decrease in MN formation in mice was greater with 0.5% DADS compared to 1% DADS. Moreover, an increase in spleen weight observed 3 weeks after irradiation was suppressed in mice administered DADS. CONCLUSIONS DADS is a potential radiation-protective agent that effectively mitigates DNA damage, and its effects in the spleen observed after irradiation may be related to inflammation and carcinogenesis.


Asunto(s)
Compuestos Alílicos/farmacología , Disulfuros/farmacología , Traumatismos por Radiación/prevención & control , Compuestos Alílicos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Daño del ADN/efectos de la radiación , Disulfuros/metabolismo , Femenino , Células Hep G2/efectos de la radiación , Humanos , Ratones , Ratones Endogámicos BALB C , Protectores contra Radiación/farmacología , Bazo/efectos de la radiación
11.
Technol Cancer Res Treat ; 18: 1533033819871309, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31495269

RESUMEN

Chondrosarcomas are malignant tumors of the cartilage that are chemoresistant and radioresistant to X-rays. This restricts the treatment options essential to surgery. In this study, we investigated the sensitivity of chondrosarcoma to X-rays and C-ions in vitro. The sensitivity of 4 chondrosarcoma cell lines (SW1353, CH2879, OUMS27, and L835) was determined by clonogenic survival assays and cell cycle progression. In addition, biomarkers of DNA damage responses were analyzed in the SW1353 cell line. Chondrosarcoma cells showed a heterogeneous sensitivity toward irradiation. Chondrosarcoma cell lines were more sensitive to C-ions exposure compared to X-rays. Using D10 values, the relative biological effectiveness of C-ions was higher (relative biological effectiveness = 5.5) with cells resistant to X-rays (CH2879) and lower (relative biological effectiveness = 3.7) with sensitive cells (L835). C-ions induced more G2 phase blockage and micronuclei in SW1353 cells as compared to X-rays with the same doses. Persistent unrepaired DNA damage was also higher following C-ions irradiation. These results indicate that chondrosarcoma cell lines displayed a heterogeneous response to conventional radiation treatment; however, treatment with C-ions irradiation was more efficient in killing chondrosarcoma cells, compared to X-rays.


Asunto(s)
Condrosarcoma/radioterapia , Transferencia Lineal de Energía , Radiografía , Rayos X/efectos adversos , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Condrosarcoma/patología , Daño del ADN/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Radiación Ionizante , Efectividad Biológica Relativa
12.
Cancers (Basel) ; 11(4)2019 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-30987217

RESUMEN

Although conventional radiotherapy promotes the migration/invasion of cancer stem cells (CSCs) under normoxia, carbon ion (C-ion) irradiation actually decreases these processes. Unraveling the mechanisms of this discrepancy, particularly under the hypoxic conditions that pertain in niches where CSCs are preferentially localized, would provide a better understanding of the origins of metastases. Invasion/migration, proteins involved in epithelial-to-mesenchymal transition (EMT), and expression of MMP-2 and HIF-1α were quantified in the CSC subpopulations of two head-and-neck squamous cell carcinoma (HNSCC) cell lines irradiated with X-rays or C-ions. X-rays triggered HNSCC-CSC migration/invasion under normoxia, however this effect was significantly attenuated under hypoxia. C-ions induced fewer of these processes in both oxygenation conditions. The differential response to C-ions was associated with a lack of HIF-1α stabilization, MMP-2 expression, or activation of kinases of the main EMT signaling pathways. Furthermore,we demonstrated a major role of reactive oxygen species (ROS) in the triggering of invasion/migration in response to X-rays. Monte-Carlo simulations demonstrated that HO● radicals are quantitatively higher after C-ions than after X-rays, however they are very differently distributed within cells. We postulate that the uniform distribution of ROS after X-rays induces the mechanisms leading to invasion/migration, which ROS concentrated in C-ion tracks are unable to trigger.

13.
J Cell Commun Signal ; 13(3): 343-356, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30903603

RESUMEN

While the dose-response relationship of radiation-induced bystander effect (RIBE) is controversial at low and high linear energy transfer (LET), mechanisms and effectors of cell-to-cell communication stay unclear and highly dependent of cell type. In the present study, we investigated the capacity of chondrocytes in responding to bystander factors released by chondrosarcoma cells irradiated at different doses (0.05 to 8 Gy) with X-rays and C-ions. Following a medium transfer protocol, cell survival, proliferation and DNA damages were quantified in bystander chondrocytes. The bystander factors secreted by chondrosarcoma cells were characterized. A significant and major RIBE response was observed in chondrocyte cells (T/C-28a2) receiving conditioned medium from chondrosarcoma cells (SW1353) irradiated with 0.1 Gy of X-rays and 0.05 Gy of C-ions, resulting in cell survivals of 36% and 62%, respectively. Micronuclei induction in bystander cells was observed from the same low doses. The cell survival results obtained by clonogenic assays were confirmed using impedancemetry. The bystander activity was vanished after a heat treatment or a dilution of the conditioned media. The cytokines which are well known as bystander factors, TNF-α and IL-6, were increased as a function of doses and LET according to an ELISA multiplex analysis. Together, the results demonstrate that irradiated chondrosarcoma cells can communicate stress factors to non-irradiated chondrocytes, inducing a wide and specific bystander response related to both doses and LET.

14.
Int J Mol Sci ; 19(12)2018 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-30513990

RESUMEN

Radiation has a wide variety of effects on the liver. Fibrosis is a concern in medical fields as one of the acute effects of high-dose irradiation, such as with cancer radiotherapies. Cancer is also an important concern following exposure to radiation. The liver has an active metabolism and reacts to radiations. In addition, effects are modulated by many environmental factors, such as high-calorie foods or alcohol beverages. Adaptations to other environmental conditions could also influence the effects of radiation. Reactions to radiation may not be optimally regulated under conditions modulated by the environment, possibly leading to dysregulation, disease or cancer. Here, we introduce some reactions to ionizing radiation in the liver, as demonstrated primarily in animal experiments. In addition, modulation of radiation-induced effects in the liver due to factors such as obesity, alcohol drinking, or supplements derived from foods are reviewed. Perspectives on medical applications by modulations of radiation effects are also discussed.


Asunto(s)
Estilo de Vida , Hígado/efectos de la radiación , Radiación Ionizante , Animales , Ambiente , Alimentos , Humanos , Tolerancia a Radiación
15.
Anticancer Res ; 37(8): 4111-4117, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28739695

RESUMEN

BACKGROUND/AIM: Glioblastoma is a frequent type of brain tumor and is radioresistant. Arsenite, which crosses the blood-brain barrier, shows synergistic effects with radiation in vitro and in vivo. The mechanism remains unclear. MATERIALS AND METHODS: As synergistic radiosensitization has been reported in p53-deficient cancer cells, radiosensitization was evaluated using the glioblastoma cell line, U87MG-E6, which harbors inactivated p53, in comparison with the cell line, HCT116 p53 (-/-). Radiosensitivity was evaluated using clonogenic assays and detection of abnormal amplification of centrosomes (AAC). RESULTS: Synergistic effects of arsenite on radiosensitivity were observed in both cell lines. The radiosensitization induced by arsenite was abolished by N-acetyl-l-cysteine, a reactive oxygen species (ROS) scavenger. Increased radiosensitivity by arsenite was also abolished following knock-down of BRCA2. In addition, the increased radiosensitization by arsenite was correlated with AAC, which was abolished by BRCA2 knock-down. CONCLUSION: We conclude that radiosensitization by arsenite is related to ROS and BRCA2 function.


Asunto(s)
Proteína BRCA2/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/radioterapia , Tolerancia a Radiación/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Apoptosis/efectos de los fármacos , Arsenitos/administración & dosificación , Barrera Hematoencefálica/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glioblastoma/patología , Células HCT116 , Humanos , Fármacos Sensibilizantes a Radiaciones , Especies Reactivas de Oxígeno/metabolismo
16.
J Radiat Res ; 58(4): 421-429, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28201773

RESUMEN

Molecular mechanisms of radiation dose-rate effects are not well understood. Among many possibilities, long-lasting sustained alterations in protein levels would provide critical information. To evaluate sustained effects after acute and chronic radiation exposure, we analyzed alterations in protein expression in the livers of mice. Acute exposure consisted of a lethal dose of 8 Gy and a sublethal dose of 4 Gy, with analysis conducted 6 days and 3 months after irradiation, respectively. Chronic irradiation consisted of a total dose of 8 Gy delivered over 400 days (20 mGy/day). Analyses following chronic irradiation were done immediately and at 3 months after the end of the exposure. Based on antibody arrays of protein expression following both acute lethal and sublethal dose exposures, common alterations in the expression of two proteins were detected. In the sublethal dose exposure, the expression of additional proteins was altered 3 months after irradiation. Immunohistochemical analysis showed that the increase in one of the two commonly altered proteins, MyD88, was observed around blood vessels in the liver. The alterations in protein expression after chronic radiation exposure were different from those caused by acute radiation exposures. Alterations in the expression of proteins related to inflammation and apoptosis, such as caspase 12, were observed even at 3 months after the end of the chronic radiation exposure. The alterations in protein expression depended on the dose, the dose rate, and the passage of time after irradiation. These changes could be involved in long-term effects of radiation in the liver.


Asunto(s)
Hígado/metabolismo , Hígado/efectos de la radiación , Proteínas/metabolismo , Animales , Caspasa 12/metabolismo , Relación Dosis-Respuesta en la Radiación , Inmunohistoquímica , Laminina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo
17.
Artículo en Inglés | MEDLINE | ID: mdl-28010925

RESUMEN

Both ionizing radiation (IR) and psychological stress (PS) cause detrimental effects on humans. A recent study showed that chronic restraint-induced PS (CRIPS) diminished the functions of Trp53 and enhanced radiocarcinogenesis in Trp53-heterozygous (Trp53+/-) mice. These findings had a marked impact on the academic field as well as the general public, particularly among residents living in areas radioactively contaminated by nuclear accidents. In an attempt to elucidate the modifying effects of CRIPS on radiation-induced health consequences in Trp53 wild-type (Trp53+/+) animals, investigations involving multidisciplinary analyses were performed. We herein demonstrated that CRIPS induced changes in the frequency of IR-induced chromosomal aberrations (CAs) in splenocytes. Five-week-old male Trp53+/+ C57BL/6J mice were restrained for 6h per day for 28 consecutive days, and total body irradiation (TBI) at a dose of 4Gy was performed on the 8th day. Metaphase chromosome spreads prepared from splenocytes at the end of the 28-day restraint regimen were painted with fluorescence in situ hybridization (FISH) probes for chromosomes 1, 2, and 3. The results obtained showed that CRIPS alone did not induce CAs, while TBI caused significant increases in CAs, mostly translocations. Translocations appeared at a lower frequency in mice exposed to TBI plus CRIPS than in those exposed to TBI alone. No significant differences were observed in the frequencies of the other types of CAs (insertions, dicentrics, and fragments) visualized with FISH between these experimental groups (TBI+CRIPS vs. TBI). These results suggest that CRIPS does not appear to synergize with the clastogenicity of IR.


Asunto(s)
Aberraciones Cromosómicas , Inmovilización , Bazo/efectos de la radiación , Estrés Fisiológico , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína p53 Supresora de Tumor/genética , Irradiación Corporal Total
18.
PLoS One ; 11(1): e0146730, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26752639

RESUMEN

Sake is a traditional Japanese alcoholic beverage that is gaining popularity worldwide. Although sake is reported to have beneficial health effects, it is not known whether chronic sake consumption modulates health risks due to radiation exposure or other factors. Here, the effects of chronic administration of sake on radiation-induced metabolic alterations in the livers of mice were evaluated. Sake (junmai-shu) was administered daily to female mice (C3H/He) for one month, and the mice were exposed to fractionated doses of X-rays (0.75 Gy/day) for the last four days of the sake administration period. For comparative analysis, a group of mice were administered 15% (v/v) ethanol in water instead of sake. Metabolites in the liver were analyzed by capillary electrophoresis-time-of-flight mass spectrometry one day following the last exposure to radiation. The metabolite profiles of mice chronically administered sake in combination with radiation showed marked changes in purine, pyrimidine, and glutathione (GSH) metabolism, which were only partially altered by radiation or sake administration alone. Notably, the changes in GSH metabolism were not observed in mice treated with radiation following chronic administration of 15% ethanol in water. Changes in several metabolites, including methionine and valine, were induced by radiation alone, but were not detected in the livers of mice who received chronic administration of sake. In addition, the chronic administration of sake increased the level of serum triglycerides, although radiation exposure suppressed this increase. Taken together, the present findings suggest that chronic sake consumption promotes GSH metabolism and anti-oxidative activities in the liver, and thereby may contribute to minimizing the adverse effects associated with radiation.


Asunto(s)
Consumo de Bebidas Alcohólicas , Bebidas Alcohólicas/efectos adversos , Hígado/efectos de los fármacos , Hígado/efectos de la radiación , Animales , Antioxidantes/metabolismo , Fraccionamiento de la Dosis de Radiación , Electroforesis Capilar , Etanol/efectos adversos , Femenino , Glutatión/metabolismo , Inflamación , Japón , Espectrometría de Masas , Metabolómica , Metionina/metabolismo , Ratones , Ratones Endogámicos C3H , Análisis de Componente Principal , Purinas/metabolismo , Pirimidinas/metabolismo , Triglicéridos/sangre , Valina/metabolismo
19.
J Radiat Res ; 56(5): 760-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26045492

RESUMEN

Both radiation and stresses cause detrimental effects on humans. Besides possible health effects resulting directly from radiation exposure, the nuclear plant accident is a cause of social psychological stresses. A recent study showed that chronic restraint-induced stresses (CRIS) attenuated Trp53 functions and increased carcinogenesis susceptibility of Trp53-heterozygous mice to total-body X-irradiation (TBXI), having a big impact on the academic world and a sensational effect on the public, especially the residents living in radioactively contaminated areas. It is important to investigate the possible modification effects from CRIS on radiation-induced health consequences in Trp53 wild-type (Trp53wt) animals. Prior to a carcinogenesis study, effects of TBXI on the hematopoietic system under CRIS were investigated in terms of hematological abnormality in the peripheral blood and residual damage in the bone marrow erythrocytes using a mouse restraint model. Five-week-old male Trp53wt C57BL/6J mice were restrained 6 h per day for 28 consecutive days, and TBXI (4 Gy) was given on the 8th day. Results showed that CRIS alone induced a marked decrease in the red blood cell (RBC) and the white blood cell (WBC) count, while TBXI caused significantly lower counts of RBCs, WBCs and blood platelets, and a lower concentration of hemoglobin regardless of CRIS. CRIS alone did not show any significant effect on erythrocyte proliferation and on induction of micronucleated erythrocytes, whereas TBXI markedly inhibited erythrocyte proliferation and induced a significant increase in the incidences of micronucleated erythrocytes, regardless of CRIS. These findings suggest that CRIS does not have a significant impact on radiation-induced detrimental effects on the hematopoietic system in Trp53wt mice.


Asunto(s)
Sistema Hematopoyético/efectos de la radiación , Inmovilización/psicología , Traumatismos por Radiación , Estrés Psicológico , Animales , Peso Corporal , Médula Ósea/patología , Médula Ósea/efectos de la radiación , Eritrocitos/citología , Eritrocitos/efectos de la radiación , Genes p53/genética , Leucocitos/citología , Leucocitos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Micronúcleos , Factores de Riesgo , Aumento de Peso , Rayos X
20.
Med Sci Monit ; 21: 1721-5, 2015 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-26071878

RESUMEN

Organisms must confront various environmental stresses. The liver is central to protecting against such stresses in mammals, and it has many detoxification and anti-oxidative stress functions. Radiation is a source of oxidative stress and is known to affect the liver and induce anti-oxidative responses. The detoxification enzyme rhodanese, which is also called thiosulfate sulfurtransferase (TST), has been demonstrated to be induced in the liver in response to radiation. Cyanide detoxification is a function of the liver, and rhodanese is a key enzyme involved in sulfur metabolism in that detoxification. Though the anti-oxidative stress system in which sulfur molecules such as thiol compounds are involved has attracted attention as a defense against radiation, detoxification enzymes may have other roles in this defense. Understanding how these functions are affected by alterations of sulfur metabolism (including thiol compounds) after irradiation would help uncover their roles in defense against cancer and other deleterious health effects, as well as environmental stress responses. This article reviews the roles of sulfur-related metabolism in oxidative stress regulation and detoxification for recovery from liver damage after radiation exposure, with particular attention to recent findings of sulfur-related enzymes such as rhodanese, which is unique in sulfur metabolism.


Asunto(s)
Hígado/metabolismo , Hígado/efectos de la radiación , Estrés Fisiológico/efectos de la radiación , Azufre/metabolismo , Tiosulfato Azufretransferasa/metabolismo , Animales , Cianuros/farmacocinética , Fase I de la Desintoxicación Metabólica/efectos de la radiación , Estrés Oxidativo/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...