Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 415
Filtrar
1.
Food Funct ; 15(15): 8116-8127, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39011610

RESUMEN

Research on plant and animal peptides has garnered significant attention, but there is a lack of studies on the functional properties of Tenebrio molitor peptides, particularly in relation to their potential mitigating effect on radiation damage and the underlying mechanisms. This study aims to explore the protective effects of Tenebrio molitor peptides against radiation-induced damage. Mice were divided into five groups: normal, radiation model, and low-, medium-, and high-dose Tenebrio molitor peptide (TMP) groups (0.15 g per kg BW, 0.30 g per kg BW, and 0.60 g per kg BW). Various parameters such as blood cell counts, bone marrow DNA content, immune organ indices, serum levels of D-lactic acid, diamine oxidase (DAO), endotoxin (LPS), and inflammatory factors were assessed at 3 and 15 days post gamma irradiation. Additionally, the intestinal tissue morphology was examined through H&E staining, RT-qPCR experiments were conducted to analyze the expression of inflammatory factors in the intestine, and immunohistochemistry was utilized to evaluate the expression of tight junction proteins ZO-1 and Occludin in the intestine. The findings revealed that high-dose TMP significantly enhanced the hematopoietic system function in mice post radiation exposure, leading to increased spleen index, thymus index, blood cell counts, and bone marrow DNA production (p < 0.05). Moreover, TMP improved the intestinal barrier integrity and reduced the intestinal permeability. Mechanistic insights suggested that these peptides may safeguard intestinal barrier function by downregulating the gene expression of inflammatory factors TNF-α, IL-1ß, and IL-6, while upregulating the expression of tight junction proteins ZO-1 and Occludin (p < 0.05). Overall, supplementation with TMP mitigates radiation-induced intestinal damage by enhancing the hematopoietic system and the intestinal barrier, offering valuable insights for further investigations into the mechanisms underlying the protective effects of these peptides against ionizing radiation.


Asunto(s)
Mucosa Intestinal , Péptidos , Tenebrio , Animales , Ratones , Péptidos/farmacología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de la radiación , Mucosa Intestinal/efectos de los fármacos , Masculino , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/efectos de la radiación , Protectores contra Radiación/farmacología , Proteína de la Zonula Occludens-1/metabolismo , Proteína de la Zonula Occludens-1/genética , Rayos gamma/efectos adversos , Ocludina/metabolismo , Ocludina/genética , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación
2.
Immunology ; 172(4): 614-626, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38685744

RESUMEN

Ionising radiation exposure can lead to acute haematopoietic radiation syndrome. Despite significant advancements in the field of radioprotection, no drugs with high efficacy and low toxicity have yet been approved by the Food and Drug Administration. FG-4592, as a proline hydroxylase inhibitor, may play an important role in radioprotection of the haematopoietic system. Mice were peritoneal injected with FG-4592 or normal saline. After irradiation, the survival time, body weight, peripheral blood cell and bone marrow cell (BMC) count, cell apoptosis, pathology were analysed and RNA-sequence technique (RNA-Seq) was conducted to explore the mechanism of FG-4592 in the haematopoietic system. Our results indicated that FG-4592 improved the survival rate and weight of irradiated mice and protected the spleen, thymus and bone marrow from IR-induced injury. The number of BMCs was increased and protected against IR-induced apoptosis. FG-4592 also promoted the recovery of the blood system and erythroid differentiation. The results of RNA-Seq and Western blot showed that the NF-κB signalling pathway and hypoxia-inducible factor-1 (HIF-1) signalling pathway were upregulated by FG-4592. Meanwhile, RT-PCR results showed that FG-4592 could promote inflammatory response significantly. FG-4592 exhibited radioprotective effects in the haematopoietic system by promoting inflammatory response and targeting the NF-κB, HIF signalling pathway.


Asunto(s)
Apoptosis , Radiación Ionizante , Protectores contra Radiación , Animales , Ratones , Protectores contra Radiación/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Transducción de Señal/efectos de los fármacos , FN-kappa B/metabolismo , Masculino , Ratones Endogámicos C57BL , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/efectos de la radiación , Síndrome de Radiación Aguda/prevención & control , Síndrome de Radiación Aguda/tratamiento farmacológico , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Traumatismos Experimentales por Radiación/prevención & control , Traumatismos Experimentales por Radiación/metabolismo , Irradiación Corporal Total , Glicina/análogos & derivados , Isoquinolinas
3.
Curr Radiopharm ; 17(2): 200-208, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38231059

RESUMEN

BACKGROUND: The modern world faces a growing concern about the possibility of accidental radiation events. The Hematopoietic system is particularly vulnerable to radiationinduced apoptosis, which can lead to death. Metformin, a drug used to treat diabetes, has been shown to protect normal cells and tissues from the toxic effects of radiation. This study aimed to evaluate the effectiveness of metformin in mitigating radiation injury to the gastrointestinal and hematological systems of rats. MATERIALS AND METHODS: The study involved 73 male rats. After total body irradiation with 7.5 Gy of X-rays, rats were treated with metformin. Seven days later, the rats were sacrificed and blood samples were taken for evaluation. RESULTS: The study found that metformin was not effective in mitigating radiation injury. The histopathological assessment showed no significant changes in goblet cell injury, villi shortening, inflammation, or mucous layer thickness. In terms of biochemical evaluation, metformin did not significantly affect oxidative stress markers, but irradiation increased the mean MDA level in the radiation group. The complete blood count revealed a significant decrease in WBC and platelet, counts in the radiation group compared to the control group, but no significant difference was found between the radiation and radiation + metformin groups. CONCLUSION: In conclusion, metformin may not be a good option for reducing radiation toxicity after accidental exposure. Despite treatment, there was no improvement in platelet, white blood cell, and lymphocyte counts, nor was there any decrease in oxidative stress. Further research is needed to explore other potential treatments for radiation injury.


Asunto(s)
Metformina , Estrés Oxidativo , Traumatismos Experimentales por Radiación , Irradiación Corporal Total , Animales , Metformina/farmacología , Ratas , Masculino , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/prevención & control , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/efectos de la radiación , Tracto Gastrointestinal/efectos de la radiación , Tracto Gastrointestinal/efectos de los fármacos , Protectores contra Radiación/farmacología , Rayos X
4.
Sci Rep ; 12(1): 3485, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35241733

RESUMEN

The threat of a nuclear attack has increased in recent years highlighting the benefit of developing additional therapies for the treatment of victims suffering from Acute Radiation Syndrome (ARS). In this work, we evaluated the impact of a PEGylated thrombopoietin mimetic peptide, JNJ-26366821, on the mortality and hematopoietic effects associated with ARS in mice exposed to lethal doses of total body irradiation (TBI). JNJ-26366821 was efficacious as a mitigator of mortality and thrombocytopenia associated with ARS in both CD2F1 and C57BL/6 mice exposed to TBI from a cobalt-60 gamma-ray source. Single administration of doses ranging from 0.3 to 1 mg/kg, given 4, 8, 12 or 24 h post-TBI (LD70 dose) increased survival by 30-90% as compared to saline control treatment. At the conclusion of the 30-day study, significant increases in bone marrow colony forming units and megakaryocytes were observed in animals administered JNJ-26366821 compared to those administered saline. In addition, enhanced recovery of FLT3-L levels was observed in JNJ-26366821-treated animals. Probit analysis of survival in the JNJ-26366821- and saline-treated cohorts revealed a dose reduction factor of 1.113 and significant increases in survival for up to 6 months following irradiation. These results support the potential use of JNJ-26366821 as a medical countermeasure for treatment of acute TBI exposure in case of a radiological/nuclear event when administered from 4 to 24 h post-TBI.


Asunto(s)
Síndrome de Radiación Aguda , Materiales Biomiméticos , Sistema Hematopoyético , Trombopoyetina , Síndrome de Radiación Aguda/tratamiento farmacológico , Síndrome de Radiación Aguda/patología , Animales , Materiales Biomiméticos/farmacología , Sistema Hematopoyético/patología , Sistema Hematopoyético/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/patología , Trombopoyetina/farmacología , Irradiación Corporal Total
5.
Radiat Res ; 196(6): 686-689, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34644390

RESUMEN

Exposure to high dose radiation causes life-threatening acute and delayed effects. Defining the mechanisms of lethal radiation-induced acute toxicity of gastrointestinal and hematopoietic tissues are critical steps to identify drug targets to mitigate and protect against the acute radiation syndrome (ARS). For example, one rational approach would be to design pharmaceuticals that block cell death pathways to preserve tissue integrity in radiation-sensitive organ systems including the gastrointestinal tract and hematopoietic compartment. A previous study reported that the inflammasome pathway, which mediates inflammatory cell death through pyroptosis, promotes ARS. However, we show that mice lacking the inflammatory executioner caspases, caspase-1 and caspase-11, are not protected from ARS when compared directly to littermates expressing caspase-1 and caspase-11. These results suggest that alternative pathways will need to be targeted by drugs that successfully mitigate and protect against the ARS.


Asunto(s)
Síndrome de Radiación Aguda/enzimología , Caspasa 1/metabolismo , Caspasas Iniciadoras/metabolismo , Inflamasomas/metabolismo , Animales , Sistema Hematopoyético/efectos de la radiación , Inflamasomas/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Piroptosis/efectos de la radiación
6.
PLoS One ; 16(8): e0256208, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34449797

RESUMEN

Our laboratory has demonstrated that captopril, an angiotensin converting enzyme inhibitor, mitigates hematopoietic injury following total body irradiation in mice. Improved survival in mice is correlated with improved recovery of mature blood cells and bone marrow, reduction of radiation-induced inflammation, and suppression of radiation coagulopathy. Here we investigated the effects of captopril treatment against radiation injuries in the Göttingen mini pig model of Hematopoietic-Acute Radiation Syndrome (H-ARS). Minipigs were given captopril orally (0.96 mg/kg) twice daily for 12 days following total body irradiation (60Co 1.79 Gy, 0.42-0.48 Gy/min). Blood was drawn over a time course following irradiation, and tissue samples were collected at euthanasia (32-35 days post-irradiation). We observed improved survival with captopril treatment, with survival rates of 62.5% in vehicle treated and 87.5% in captopril treated group. Additionally, captopril significantly improved recovery of peripheral blood mononuclear cells, and a trend toward improvement in recovery of red blood cells and platelets. Captopril significantly reduced radiation-induced expression of cytokines erythropoietin and granulocyte-macrophage colony-stimulating factor and suppressed radiation-induced acute-phase inflammatory response cytokine serum amyloid protein A. Using quantitative-RT-PCR to monitor bone marrow recovery, we observed significant suppression of radiation-induced expression of redox stress genes and improved hematopoietic cytokine expression. Our findings suggest that captopril activities in the Göttingen minipig model of hematopoietic-acute radiation syndrome reflect findings in the murine model.


Asunto(s)
Síndrome de Radiación Aguda/tratamiento farmacológico , Captopril/farmacología , Sistema Hematopoyético/efectos de los fármacos , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Síndrome de Radiación Aguda/patología , Animales , Modelos Animales de Enfermedad , Eritropoyetina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Sistema Hematopoyético/lesiones , Sistema Hematopoyético/patología , Sistema Hematopoyético/efectos de la radiación , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/efectos de la radiación , Ratones , Oxidación-Reducción/efectos de los fármacos , Traumatismos Experimentales por Radiación/patología , Porcinos , Porcinos Enanos , Irradiación Corporal Total/efectos adversos
7.
Free Radic Res ; 55(3): 230-245, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34044723

RESUMEN

Radiation-induced hematopoietic dysfunction is one of the most common problems during unplanned radiation exposures and also in cancer patients receiving radiotherapy. Management of the hematopoietic system is necessary to promote survival against radiation. The present study was undertaken to demonstrate the protective potential of Quercetin 3 rutinoside (Q-3-R), against gamma radiation-induced hematopoietic injuries. C57BL/6 male mice exposed either to radiation or pretreated with Q-3-R (10 mg/kg body weight) were checked for hematopoietic protection using hematotoxicity indices, histopathological, and genotoxic evaluations. To elucidate the underlying mechanisms of Q-3-R mediated hematopoietic protection, oxidative/nitrosative stress, inflammatory and apoptotic markers as well as PCNA expression in spleen cross-sections were assessed. Studies revealed Q-3-R pretreatment inhibited radiation-induced ROS in spleen cells and better maintained the total antioxidant levels in serum that were otherwise altered post 7.5 Gy exposure. The NO levels and nitrotyrosine expression were also found inhibited by Q-3-R in the spleen. Differential regulations of Bcl2, Bax and NF-κB with reduced serum TNF-α level indicated anti-apoptotic and anti-inflammatory roles of Q-3-R. Q-3-R attenuated radiation mediated spleen damage by minimizing cell death and promoting proliferation. Restoration of abnormal histopathological changes in bone marrow following Q-3-R administration correlated to reduced apoptosis and altered cell cycle distributions. Chromosomal aberrations were also found reduced in Q-3-R pretreated bone marrow. Q-3-R restored the total leukocyte counts and serum IL-6 levels, further supporting its role in promoting hematopoiesis. These findings suggest that Q-3-R can potentially be used to minimize radiation inflicted hematopoietic toxicities during accidental as well as radiotherapy scenarios.


Asunto(s)
Apoptosis/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Rayos gamma/efectos adversos , Sistema Hematopoyético/efectos de la radiación , Inflamación/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Protectores contra Radiación/uso terapéutico , Rutina/uso terapéutico , Bazo/efectos de los fármacos , Animales , Humanos , Masculino , Ratones , Protectores contra Radiación/farmacología , Rutina/farmacología
8.
Radiat Res ; 196(1): 55-65, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33914879

RESUMEN

The growing risk of accidental radiation exposure due to increased usage of ionizing radiation, such as in nuclear power, industries and medicine, has increased the necessity for the development of radiation countermeasures. Previously, we demonstrated the therapeutic potential of the acetylated diacylglycerol, 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG), as a radiation countermeasure by mitigating radiation-associated mortality and hematopoietic acute radiation syndrome (H-ARS) in BALB/c mice after a lethal dose (LD70/30) of gamma-ray total-body irradiation (TBI). In this study, we show that PLAG mitigates symptoms of H-ARS, as characterized by mature blood cell recovery and restoration of bone marrow cellularity, by regulating systemic inflammation. Log-rank test demonstrated that high levels of WBCs, lymphocytes and neutrophils on day 10 post-TBI resulted in significantly improved survival rate. PLAG significantly enhanced the nadir values of all major blood cell types as well as bone marrow cellularity. A single TBI at LD70/30 induced an immediate increase in the blood levels of CXCL1 (12.5 fold), CXCL2 (1.5 fold), IL-6 (86.9 fold), C-reactive protein (CRP; 1.3 fold) and G-CSF (15.7 fold) at 6 h post-TBI, but the cytokine levels returned to baseline level afterward. When the irradiated mice started to die around 15 days post-TBI, they exhibited a second surge in blood levels of CXCL1 (49.3 fold), CXCL2 (87.1 fold), IL-6 (208 fold), CRP (3.6 fold) and G-CSF (265.7 fold). However, PLAG-treated groups showed a significant decrease in these same blood levels (P < 0.001). Considering the inverse correlation between inflammatory cytokine levels and hematological nadirs, PLAG exerts its therapeutic effects on H-ARS by regulating inflammatory cytokine production. These data suggest that PLAG has high potential as a radiation countermeasure to mitigate H-ARS after accidental exposure to radiation.


Asunto(s)
Síndrome de Radiación Aguda/tratamiento farmacológico , Diglicéridos/uso terapéutico , Sistema Hematopoyético/efectos de la radiación , Inflamación/tratamiento farmacológico , Síndrome de Radiación Aguda/complicaciones , Animales , Modelos Animales de Enfermedad , Femenino , Inflamación/etiología , Recuento de Leucocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Neutrófilos/citología , Análisis de Supervivencia , Irradiación Corporal Total
9.
Radiat Res ; 195(4): 307-323, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33577641

RESUMEN

Medical countermeasures (MCMs) for hematopoietic acute radiation syndrome (H-ARS) should be evaluated in well-characterized animal models, with consideration of at-risk populations such as pediatrics. We have developed pediatric mouse models of H-ARS and delayed effects of acute radiation exposure (DEARE) for efficacy testing of MCMs against radiation. Male and female C57BL/6J mice aged 3, 4, 5, 6, 7 and 8 weeks old (±1 day) were characterized for baseline hematopoietic and gastrointestinal parameters, radiation response, efficacy of a known MCM, and DEARE at six and 12 months after total-body irradiation (TBI). Weanlings (age 3 weeks) were the most radiosensitive age group with an estimated LD50/30 of 712 cGy, while mice aged 4 to 8 weeks were more radioresistant with an estimated LD50/30 of 767-787 cGy. Female weanlings were more radiosensitive than males at 3 and 4 weeks old but became significantly more radioresistant after the pubertal age of 5 weeks. The most dramatic increase in body weight, RBC counts and intestinal circumference length occurred from 3 to 5 weeks of age. The established radiomitigator Neulasta® (pegfilgrastim) significantly increased 30-day survival in all age groups, validating these models for MCM efficacy testing. Analyses of DEARE among pediatric survivors revealed depressed weight gain in males six months post-TBI, and increased blood urea nitrogen at 12 months post-TBI which was more severe in females. Hematopoietic DEARE at six months post-TBI appeared to be less severe in survivors from the 3- and 4-week-old groups but was equally severe in all age groups by 12 months of age. Similar to our other acute radiation mouse models, there was no appreciable effect of Neulasta used as an H-ARS MCM on the severity of DEARE. In summary, these data characterize a pediatric mouse model useful for assessing the efficacy of MCMs against ARS and DEARE in children.


Asunto(s)
Síndrome de Radiación Aguda/tratamiento farmacológico , Filgrastim/farmacología , Sistema Hematopoyético/efectos de los fármacos , Polietilenglicoles/farmacología , Tolerancia a Radiación/efectos de los fármacos , Síndrome de Radiación Aguda/etiología , Síndrome de Radiación Aguda/fisiopatología , Animales , Modelos Animales de Enfermedad , Sistema Hematopoyético/fisiopatología , Sistema Hematopoyético/efectos de la radiación , Humanos , Ratones , Pediatría , Tolerancia a Radiación/efectos de la radiación , Irradiación Corporal Total/efectos adversos
10.
J Cell Mol Med ; 25(8): 3785-3792, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33609010

RESUMEN

Severe ionizing radiation causes the acute lethal damage of haematopoietic system and gastrointestinal tract. Here, we found CL429, the novel chimeric TLR2/NOD2 agonist, exhibited significant radioprotective effects in mice. CL429 increased mice survival, protected mice against the lethal damage of haematopoietic system and gastrointestinal tract. CL429 was more effective than equivalent amounts of monospecific (TLR2 or NOD2) and combination (TLR2 + NOD2) of molecules in preventing radiation-induced death. The radioprotection of CL429 was mainly mediated by activating TLR2 and partially activating NOD2. CL429-induced radioprotection was largely dependent on the activation of TLR2-MyD88-NF-κB signalling pathway. In conclusion, the data suggested that the co-activation of TLR2 and NOD2 could induce significant synergistic radioprotective effects and CL429 might be a potential high-efficiency selective agent.


Asunto(s)
Acetilmuramil-Alanil-Isoglutamina/análogos & derivados , Síndrome de Radiación Aguda/prevención & control , Sistema Hematopoyético/efectos de los fármacos , Intestinos/efectos de los fármacos , Proteína Adaptadora de Señalización NOD2/agonistas , Protectores contra Radiación/farmacología , Receptor Toll-Like 2/agonistas , Irradiación Corporal Total/efectos adversos , Acetilmuramil-Alanil-Isoglutamina/farmacología , Síndrome de Radiación Aguda/etiología , Síndrome de Radiación Aguda/patología , Animales , Sistema Hematopoyético/efectos de la radiación , Intestinos/lesiones , Intestinos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL
11.
Health Phys ; 119(5): 647-658, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32947490

RESUMEN

Lymphoid lineage recovery and involution after exposure to potentially lethal doses of ionizing radiation have not been well defined, especially the long-term effects in aged survivors and with regard to male/female differences. To examine these questions, male and female C57BL/6 mice were exposed to lethal radiation at 12 wk of age in a model of the Hematopoietic-Acute Radiation Syndrome, and bone marrow, thymus, spleen, and peripheral blood examined up to 24 mo of age for the lymphopoietic delayed effects of acute radiation exposure. Aged mice showed myeloid skewing and incomplete lymphocyte recovery in all lymphoid tissues. Spleen and peripheral blood both exhibited a monophasic recovery pattern, while thymus demonstrated a biphasic pattern. Naïve T cells in blood and spleen and all subsets of thymocytes were decreased in aged irradiated mice compared to age-matched non-irradiated controls. Of interest, irradiated males experienced significantly improved reconstitution of thymocyte subsets and peripheral blood elements compared to females. Bone marrow from aged irradiated survivors was significantly deficient in the primitive lymphoid-primed multipotent progenitors and common lymphoid progenitors, which were only 8-10% of levels in aged-matched non-irradiated controls. Taken together, these analyses define significant age- and sex-related deficiencies at all levels of lymphopoiesis throughout the lifespan of survivors of the Hematopoietic-Acute Radiation Syndrome and may provide a murine model suitable for assessing the efficacy of potential medical countermeasures and therapeutic strategies to alleviate the severe immune suppression that occurs after radiation exposure.


Asunto(s)
Síndrome de Radiación Aguda/patología , Sistema Hematopoyético/inmunología , Reconstitución Inmune , Exposición a la Radiación/efectos adversos , Traumatismos Experimentales por Radiación/patología , Bazo/inmunología , Timo/inmunología , Síndrome de Radiación Aguda/etiología , Animales , Modelos Animales de Enfermedad , Femenino , Sistema Hematopoyético/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Dosis de Radiación , Traumatismos Experimentales por Radiación/etiología , Bazo/efectos de la radiación , Timo/efectos de la radiación
12.
Free Radic Res ; 54(7): 497-516, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32746646

RESUMEN

The present study was conceptualized to delineate radioprotective efficacy of a formulation G-003M (a combination of podophyllotoxin and rutin) against radiation-induced damage to the lymphohematopoietic system of mice. C57BL/6J mice, treated with G-003M 1 h prior to 9 Gy lethal dose, were assessed for reactive oxygen species (ROS)/nitric oxide (NO) generation, antioxidant alterations, Annexin V/PI and TUNEL staining for apoptosis, modulation of apoptotic proteins, cell proliferation, histological alterations in thymus and cell cycle arrest in bone marrow cells. Induction of granulocyte colony-stimulating factor (G-CSF), granulocytes macrophage colony-stimulating factor (GM-CSF), interleukin-IL-6, IL-10, IL-1α, and IL-1ß in response to G-003M was also evaluated in different groups of mice. Haematopoietic reconstitution with G-003M was explored by examining endogenous spleen colony-forming units (CFU-S) in irradiated animals. G-003M significantly inhibited ROS/NO, malondialdehyde (MDA) and restored cellular antioxidant glutathione in the thymus of irradiated animals. G-003M pre-treatment significantly (p < 0.001) restrained apoptosis in thymocytes via upregulation of Bcl2 and down-regulation of Bax, p53 and caspase-3. Stimulation of cell proliferation and inhibition of apoptosis by G-003M, restored architecture of thymus in irradiated animals within 30 days as evaluated by histological analysis. G-003M arrested cells at the G2/M phase by inducing reversible cell cycle arrest. Peak expression of G-CSF (45-fold) and IL-6 (60-fold) as well as moderate induction of GM-CSF, IL-10, IL-1α by G-003M helped in haematopoietic recovery of irradiated mice. A higher number of endogenous CFU-S in G-003M pre-treated irradiated mice suggested haematopoietic recovery. Data obtained from the current study affirms that G-003M can be proved as a potential radioprotective agent against radiation damage.


Asunto(s)
Citocinas/metabolismo , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/efectos de la radiación , Podofilotoxina/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Rutina/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Combinación de Medicamentos , Sistema Hematopoyético/metabolismo , Sistema Hematopoyético/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Distribución Aleatoria , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo
13.
Int J Radiat Oncol Biol Phys ; 108(5): 1357-1367, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32758640

RESUMEN

PURPOSE: Recombinant human thrombopoietin (rhTPO) has been evaluated as a therapeutic intervention for radiation-induced myelosuppression. However, the immunogenicity induced by a repeated-dosing strategy raises concerns about the therapeutic use of rhTPO. In this study, single-dose administration of rhTPO was evaluated for efficacy in the hematopoietic response and survival effect on mice and nonhuman primates exposed to total body irradiation (TBI). METHODS AND MATERIALS: Survival of lethally (9.0 Gy) irradiated C57BL/6J male mice was observed for 30 days after irradiation. Hematologic evaluations were performed on C57BL/6J male mice given a sublethal dose of radiation (6.5 Gy). Furthermore, in sublethally irradiated mice, we performed bone marrow (BM) histologic evaluation and evaluated BM-derived clonogenic activity. Next, the proportion and number of hematopoietic stem cells (HSCs) were analyzed. Competitive repopulation experiments were conducted to assess the multilineage engraftment of irradiated HSCs after BM transplantation. Flow cytometry was used to evaluate DNA damage, cell apoptosis, and cell cycle stage in HSCs after irradiation. Finally, we evaluated the efficacy of a single dose of rhTPO administered after 7 Gy TBI in male and female rhesus monkeys. RESULTS: A single administration of rhTPO 2 hours after irradiation significantly mitigated TBI-induced death in mice. rhTPO promoted multilineage hematopoietic recovery, increasing peripheral blood cell counts, BM cellularity, and BM colony-forming ability. rhTPO administration led to an accelerated recovery of BM HSC frequency and multilineage engraftment after transplantation. rhTPO treatment reduced radiation-induced DNA damage and apoptosis and promoted HSC proliferation after TBI. Notably, a single administration of rhTPO significantly promoted multilineage hematopoietic recovery and improved survival in nonhuman primates after TBI. CONCLUSIONS: These findings indicate that early intervention with a single administration of rhTPO may represent a promising and effective radiomitigative strategy for victims of radiation disasters.


Asunto(s)
Médula Ósea/efectos de la radiación , Traumatismos Experimentales por Radiación/prevención & control , Trombopoyetina/administración & dosificación , Irradiación Corporal Total/efectos adversos , Animales , Apoptosis , Recuento de Células Sanguíneas , Médula Ósea/efectos de los fármacos , Médula Ósea/lesiones , Médula Ósea/patología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/efectos de la radiación , Ciclo Celular , Daño del ADN/efectos de los fármacos , Femenino , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/lesiones , Sistema Hematopoyético/patología , Sistema Hematopoyético/efectos de la radiación , Humanos , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/administración & dosificación , Factores de Tiempo
14.
Int J Radiat Oncol Biol Phys ; 108(4): 1091-1102, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32629081

RESUMEN

PURPOSE: Humans are exposed to charged particles in different scenarios. The use of protons and high-linear energy transfer (LET) in cancer treatment is steadily growing. In outer space, astronauts will be exposed to a mixed radiation field composed of both protons and heavy ions, in particularly the long-term space missions outside of earth's magnetosphere. Thus, understanding the radiobiology and transforming potential of these types of ionizing radiation are of paramount importance. METHODS AND MATERIALS: We examined the effect of 10 or 100 cGy of whole-body doses of protons or 28Si ions on the hematopoietic system of a genetic model of aging based on recent studies that showed selective loss of the MLH1 protein in human hematopoietic stems with age. RESULTS: We found that Mlh1 deficient animals are highly prone to develop lymphomas when exposed to either low doses of protons or 28Si ions. The lymphomas that develop are genetically indistinguishable, in spite of different types of damage elicited by low- and high-LET radiation. RNA sequencing analyses reveal similar gene expression patterns, similar numbers of altered genes, similar numbers of single nucleotide variants and insertions and deletions, and similar activation of known leukemogenic loci. CONCLUSIONS: Although the incidence of malignancy is related to radiation quality, and increased due to loss of Mlh1, the phenotype of the tumors is independent of LET.


Asunto(s)
Sistema Hematopoyético/efectos de la radiación , Transferencia Lineal de Energía , Linfoma/genética , Homólogo 1 de la Proteína MutL/deficiencia , Neoplasias Inducidas por Radiación/genética , Protones/efectos adversos , Silicio/efectos adversos , Envejecimiento , Animales , Reparación de la Incompatibilidad de ADN , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Sistema Hematopoyético/fisiología , Humanos , Linfoma/patología , Masculino , Ratones , Homólogo 1 de la Proteína MutL/genética , Neoplasias Inducidas por Radiación/patología , Penetrancia , Exposición a la Radiación/efectos adversos , Análisis de Secuencia de ARN/métodos , Vuelo Espacial , Irradiación Corporal Total/efectos adversos , Irradiación Corporal Total/métodos
15.
Int J Mol Sci ; 21(14)2020 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-32708958

RESUMEN

Acute exposure to ionizing radiation leads to Hematopoietic Acute Radiation Syndrome (H-ARS). To understand the inter-strain cellular and molecular mechanisms of radiation sensitivity, adult males of two strains of minipig, one with higher radiosensitivity, the Gottingen minipig (GMP), and another strain with comparatively lower radiosensitivity, the Sinclair minipig (SMP), were exposed to total body irradiation (TBI). Since Insulin-like Growth Factor-1 (IGF-1) signaling is associated with radiation sensitivity and regulation of cardiovascular homeostasis, we investigated the link between dysregulation of cardiac IGF-1 signaling and radiosensitivity. The adult male GMP; n = 48, and SMP; n = 24, were irradiated using gamma photons at 1.7-2.3 Gy doses. The animals that survived to day 45 after irradiation were euthanized and termed the survivors. Those animals that were euthanized prior to day 45 post-irradiation due to severe illness or health deterioration were termed the decedents. Cardiac tissue analysis of unirradiated and irradiated animals showed that inter-strain radiosensitivity and survival outcomes in H-ARS are associated with activation status of the cardiac IGF-1 signaling and nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated induction of antioxidant gene expression. Our data link H-ARS with dysregulation of cardiac IGF-1 signaling, and highlight the role of oxidative stress and cardiac antioxidant response in radiation sensitivity.


Asunto(s)
Síndrome de Radiación Aguda/metabolismo , Corazón/efectos de la radiación , Sistema Hematopoyético/efectos de la radiación , Factor I del Crecimiento Similar a la Insulina/metabolismo , Transducción de Señal/efectos de la radiación , Síndrome de Radiación Aguda/etiología , Síndrome de Radiación Aguda/patología , Animales , Rayos gamma/efectos adversos , Sistema Hematopoyético/metabolismo , Sistema Hematopoyético/patología , Masculino , Miocardio/metabolismo , Miocardio/patología , Estrés Oxidativo/efectos de la radiación , Tolerancia a Radiación/efectos de la radiación , Porcinos , Porcinos Enanos
16.
Arch Pharm Res ; 42(11): 1021-1029, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31549342

RESUMEN

Some flavonoids have been shown to exhibit good antioxidant activity and protect mice from damage induced by radiation. Amentoflavone (AMF), a biflavonoid derived from the traditional herb-Selaginella tamariscina, has been reported to have antioxidant properties. The protective effects and mechanism of action of AMF against radiation injury remain unknown. In this study, male C57BL/6 mice were subjected to total-body 60Co γ-irradiation at 7.5 or 3.0 Gy. The survival rate and mean survival time were evaluated to determine the radioprotective effect of AMF. Number of peripheral blood cells, frequency of colony forming unit-granulocytes, monocytes and micronuclei were measured to assess the protective effects of AMF on the hematopoietic system. Levels of superoxide dismutase and glutathione, and pathological changes in the bone marrow were determined. Additionally, next-generation sequencing technology was used to explore potential targets of AMF. We observed that AMF markedly extends average survival time, reduces injury to the hematopoietic system and promotes its recovery. Furthermore, treatment with AMF significantly attenuated radiation-induced oxidative stress. In addition, AMF had a significant effect on gene tumor necrosis factor alpha-induced protein 2. Together, the results of this study suggest that AMF is a potential protective agent against radiation injury.


Asunto(s)
Antioxidantes/administración & dosificación , Biflavonoides/administración & dosificación , Sistema Hematopoyético/efectos de los fármacos , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/administración & dosificación , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Rayos gamma/efectos adversos , Sistema Hematopoyético/efectos de la radiación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Exposición a la Radiación/efectos adversos , Traumatismos Experimentales por Radiación/etiología , Factores de Necrosis Tumoral/metabolismo , Irradiación Corporal Total
17.
Int J Radiat Biol ; 95(8): 1094-1102, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30831047

RESUMEN

Purpose: The present study aimed to investigate the potential protective effects of icariin both in vivo and in vitro, an active flavonoid glucoside derived from medicinal herb Epimedium, and its possible mechanisms against radiation-induced injury. Methods: Male C57BL/6 mice were exposed to lethal dose (7 Gy) or sub-lethal dose (4 Gy) of whole body radiation by X-ray at a dose rate of ∼0.55 Gy/min, and icariin was given three times at 24 h and 30 min before and 24 h after the irradiation. After irradiation, hematological, biochemical, and histological evaluations were performed. We further determined the effect of icariin on radiation-induced cytotoxicity and changes in apoptosis-related protein expression. Results: Icariin enhanced the 30-day survival rates (20 and 40 mg/kg) in a dose-dependent manner, and protected the radiosensitive organs such as intestine and testis from the radiation damages. Moreover, hematopoietic damage by radiation was significantly decreased in icariin-treated mice as demonstrated by the increases in number of peripheral blood cells, bone marrow cells (1.7-fold), and spleen colony forming units (1.7-fold). In addition, icariin decreased the radiation-induced oxidative stress by modulating endogenous antioxidant levels. Subsequent in vitro studies showed that icariin effectively increased cell viability (1.4-fold) and suppressed the expression of apoptosis-related proteins after irradiation. Conclusion: These results suggest that icariin has significant protective effects against radiation-induced damages partly through its anti-oxidative and anti-apoptotic properties.


Asunto(s)
Flavonoides/farmacología , Protectores contra Radiación/farmacología , Animales , Apoptosis/efectos de la radiación , Sistema Hematopoyético/efectos de la radiación , Humanos , Células K562 , Masculino , Ratones , Ratones Endogámicos C57BL , Superóxido Dismutasa/metabolismo , Irradiación Corporal Total
18.
Radiat Res ; 191(4): 323-334, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30730284

RESUMEN

Intensive research is underway to find new agents that can successfully mitigate the acute effects of radiation exposure. This is primarily in response to potential counterthreats of radiological terrorism and nuclear accidents but there is some hope that they might also be of value for cancer patients treated with radiation therapy. Research into mitigation countermeasures typically employs classic animal models of acute radiation syndromes (ARS) that develop after whole-body irradiation (WBI). While agents are available that successfully mitigate ARS when given after radiation exposure, their success raises questions as to whether they simply delay lethality or unmask potentially lethal radiation pathologies that may appear later in time. Life shortening is a well-known consequence of WBI in humans and experimental animals, but it is not often examined in a mitigation setting and its causes, other than cancer, are not well-defined. This is in large part because delayed effects of acute radiation exposure (DEARE) do not follow the strict time-dose phenomena associated with ARS and present as a diverse range of symptoms and pathologies with low mortality rates that can be evaluated only with the use of large cohorts of subjects, as in this study. Here, we describe chronically increased mortality rates up to 660 days in large numbers of mice given LD70/30 doses of WBI. Systemic myeloid cell activation after WBI persists in some mice and is associated with late immunophenotypic changes and hematopoietic imbalance. Histopathological changes are largely of a chronic inflammatory nature and variable incidence, as are the clinical symptoms, including late diarrhea that correlates temporally with changes in the content of the microbiome. We also describe the acute and long-term consequences of mitigating hematopoietic ARS (H-ARS) lethality after LD70/30 doses of WBI in multiple cohorts of mice treated uniformly with radiation mitigators that have a common 4-nitro-phenylsulfonamide (NPS) pharmacophore. Effective NPS mitigators dramatically decrease ARS mortality. There is slightly increased subacute mortality, but the rate of late mortalities is slowed, allowing some mice to live a normal life span, which is not the case for WBI controls. The study has broad relevance to radiation late effects and their potential mitigation and epitomizes the complex interaction between radiation-damaged tissues and immune homeostasis.


Asunto(s)
Síndrome de Radiación Aguda/inmunología , Síndrome de Radiación Aguda/prevención & control , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/efectos de la radiación , Protectores contra Radiación/farmacología , Síndrome de Radiación Aguda/microbiología , Síndrome de Radiación Aguda/mortalidad , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/efectos de la radiación , Corazón/efectos de los fármacos , Corazón/efectos de la radiación , Masculino , Ratones , Neoplasias Inducidas por Radiación/inmunología , Neoplasias Inducidas por Radiación/microbiología , Neoplasias Inducidas por Radiación/mortalidad , Neoplasias Inducidas por Radiación/prevención & control , Sulfonamidas/farmacología , Análisis de Supervivencia
19.
PLoS One ; 14(2): e0210663, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30759098

RESUMEN

While exposure to radiation can be lifesaving in certain settings, it can also potentially result in long-lasting adverse effects, particularly to hematopoietic and immune cells. This study investigated hematopoietic recovery and immune function in rhesus macaques Cross-sectionally (at a single time point) 2 to 5 years after exposure to a single large dose (6.5 to 8.4 Gray) of total body radiation (TBI) derived from linear accelerator-derived photons (2 MeV, 80 cGy/minute) or Cobalt 60-derived gamma irradiation (60 cGy/min). Hematopoietic recovery was assessed through measurement of complete blood counts, lymphocyte subpopulation analysis, and thymus function assessment. Capacity to mount specific antibody responses against rabies, Streptococcus pneumoniae, and tetanus antigens was determined 2 years after TBI. Irradiated macaques showed increased white blood cells, decreased platelets, and decreased frequencies of peripheral blood T cells. Effects of prior radiation on production and export of new T cells by the thymus was dependent on age at the time of analysis, with evidence of interaction with radiation dose for CD8+ T cells. Irradiated and control animals mounted similar mean antibody responses to proteins from tetanus and rabies and to 10 of 11 serotype-specific pneumococcal polysaccharides. However, irradiated animals uniformly failed to make antibodies against polysaccharides from serotype 5 pneumococci, in contrast to the robust responses of non-irradiated controls. Trends toward decreased serum levels of anti-tetanus IgM and slower peak antibody responses to rabies were also observed. Taken together, these data show that dose-related changes in peripheral blood cells and immune responses to both novel and recall antigens can be detected 2 to 5 years after exposure to whole body radiation. Longer term follow-up data on this cohort and independent validation will be helpful to determine whether these changes persist or whether additional changes become evident with increasing time since radiation, particularly as animals begin to develop aging-related changes in immune function.


Asunto(s)
Rayos gamma/efectos adversos , Sistema Hematopoyético/efectos de la radiación , Inmunidad/efectos de la radiación , Irradiación Corporal Total/efectos adversos , Adulto , Animales , Formación de Anticuerpos/efectos de la radiación , Recuento de Células Sanguíneas , Relación Dosis-Respuesta en la Radiación , Hematopoyesis/efectos de la radiación , Humanos , Subgrupos Linfocitarios/efectos de la radiación , Macaca mulatta , Masculino , Traumatismos Experimentales por Radiación/etiología , Linfocitos T/efectos de la radiación , Timo/efectos de la radiación
20.
J Tradit Chin Med ; 39(3): 324-331, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-32186004

RESUMEN

OBJECTIVE: To investigate the radioprotective effect of tea polyphenols (TP 50) against radiation-induced organ and tissue damage. METHODS: Beagle dogs were exposed to a single acute dose of whole-body γ-radiation (3 Gy) and orally administered TP 50 (80 or 240 mg·kg-1·d-1) for 28 consecutive days. A hemogram was obtained from experimental dogs every other day for 42 d. At the end of the experiment, enzyme activities of the antioxidants superoxide-dismutase andglutathione peroxidase, serum levels of inflamma- tory cytokines (tumor necrosis factor-α, interleukin-1ß, and interleukin-6), colony-forming units of bone marrow hematopoietic progenitor cells, andorgan coefficients were measured. RESULTS: Dogs exposed to γ-radiation alone exhibited typical hematopoietic syndrome. In contrast, irradiated dogs that received TP 50 exhibited an improved blood profile with reduced leucopenia, thrombocytopenia (platelet counts), and reticulocyte levels. TP 50 also significantly elevated levels of the endogenous antioxidant enzyme superoxide-dismutase, reduced the increased levels of serum cytokine in response to radiation-induced toxicity, and increased colony-forming units of bone marrow hematopoietic progenitor cells. In addition, TP 50 repaired radiation-induced organ damage. CONCLUSION: The current findings suggest that oral administration of TP 50 to beagle dogs effectively alleviated hematopoietic bone marrow dam- age induced by γ-radiation.


Asunto(s)
Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/efectos de la radiación , Polifenoles/química , Polifenoles/farmacología , Té/química , Animales , Antioxidantes/metabolismo , Perros , Rayos gamma/efectos adversos , Interleucina-6/metabolismo , Masculino , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Irradiación Corporal Total/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA