Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Health Phys ; 121(4): 352-371, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34546217

RESUMEN

ABSTRACT: High-dose radiation exposure results in hematopoietic and gastrointestinal acute radiation syndromes followed by delayed effects of acute radiation exposure, which encompasses multiple organs, including heart, kidney, and lung. Here we sought to further characterize the natural history of radiation-induced heart injury via determination of differential protein and metabolite expression in the heart. We quantitatively profiled the proteome and metabolome of left and right ventricle from non-human primates following 12 Gy partial body irradiation with 2.5% bone marrow sparing over a time period of 3 wk. Global proteome profiling identified more than 2,200 unique proteins, with 220 and 286 in the left and right ventricles, respectively, showing significant responses across at least three time points compared to baseline levels. High-throughput targeted metabolomics analyzed a total of 229 metabolites and metabolite combinations, with 18 and 22 in the left and right ventricles, respectively, showing significant responses compared to baseline levels. Bioinformatic analysis performed on metabolomic and proteomic data revealed pathways related to inflammation, energy metabolism, and myocardial remodeling were dysregulated. Additionally, we observed dysregulation of the retinoid homeostasis pathway, including significant post-radiation decreases in retinoic acid, an active metabolite of vitamin A. Significant differences between left and right ventricles in the pathology of radiation-induced injury were identified. This multi-omic study characterizes the natural history and molecular mechanisms of radiation-induced heart injury in NHP exposed to PBI with minimal bone marrow sparing.


Asunto(s)
Síndrome de Radiación Aguda , Médula Ósea , Primates , Proteómica , Traumatismos por Radiación , Síndrome de Radiación Aguda/patología , Animales , Médula Ósea/efectos de la radiación , Dosis de Radiación , Traumatismos por Radiación/metabolismo
2.
Int J Radiat Biol ; 97(4): 464-473, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33464146

RESUMEN

PURPOSE: Baicalein (an anti-ferroptosis drug) was recently reported to synergistically improve the survival rate of mice following a high dose of total body irradiation with anti-apoptosis and anti-necroptosis drugs. At the same time, our group has demonstrated that ferrostatin-1, a ferroptosis inhibitor, improves the survival rate of a mouse model of hematopoietic acute radiation syndrome to 60% for 150 days (p < .001). These phenomena suggest that ferroptosis inhibition can mitigate radiation damage. In this study, we continued to study the mechanisms by which ferrostatin-1 alleviated radiation-induced ferroptosis and subsequent hematopoietic acute radiation syndrome. MATERIALS AND METHODS: Male ICR mice (8-10 weeks old) were exposed to doses of 0, 8, or 10 Gy irradiated from a 137Cs source. Ferrostatin-1 was intraperitoneally injected into mice 72 h post-irradiation. Bone marrow mononuclear cells (BMMCs) and peripheral blood cells were counted. The changes in iron-related parameters, lipid metabolic enzymes, lipid peroxidation repair molecules (glutathione peroxidase 4, glutathione, and coenzyme Q10), and inflammatory factors (TNF-α, IL-6, and IL-1ß) were evaluated using biochemical or antibody techniques. RESULTS: Ferrostatin-1 increased the number of red and white blood cells, lymphocytes, and monocytes in the peripheral blood after total body irradiation in mice by mitigating the ferroptosis of BMMCs. Total body irradiation induced ferroptosis in BMMCs by increasing the iron and lipid peroxidation levels and depleting the acyl-CoA synthetase long-chain family member 4 (ASCL4), lipoxygenase 15, glutathione peroxidase 4, and glutathione levels. Ferroptotic BMMCs did not release TNF-α, IL-6, or IL-1ß at the early stage of radiation exposure. Ferrostatin-1 mitigated the lipid peroxidation of radiation-induced ferroptosis by attenuating increases in levels of hemosiderin and liable iron pool and decreases in levels of ASCL4 and glutathione peroxidase 4. CONCLUSIONS: The onset of total body irradiation-induced ferroptosis in BMMCs involved changes in iron, lipid metabolic enzymes, and anti-lipid peroxidation molecules. Ferrostatin-1 could be a potential radiation mitigation agent by acting on these targets.


Asunto(s)
Síndrome de Radiación Aguda/patología , Ciclohexilaminas/farmacología , Hematopoyesis/efectos de los fármacos , Fenilendiaminas/farmacología , Animales , Ferroptosis/efectos de los fármacos , Ferroptosis/efectos de la radiación , Hematopoyesis/efectos de la radiación , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos ICR
3.
Exp Hematol ; 84: 54-66, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32240658

RESUMEN

Exposure to high-dose total body irradiation (TBI) can result in hematopoietic acute radiation syndrome (H-ARS), characterized by leukopenia, anemia, and coagulopathy. Death from H-ARS occurs from hematopoietic insufficiency and opportunistic infections. Following radiation exposure, red blood cells (RBCs) undergo hemolysis from radiation-induced hemoglobin denaturation, causing the release of iron. Free iron can have multiple detrimental biological effects, including suppression of hematopoiesis. We investigated the impact of radiation-induced iron release on the bone marrow following TBI and the potential impact of the ACE inhibitor captopril, which improves survival from H-ARS. C57BL/6J mice were exposed to 7.9 Gy, 60Co irradiation, 0.6 Gy/min (LD70-90/30). RBCs and reticulocytes were significantly reduced within 7 days of TBI, with the RBC nadir at 14-21 days. Iron accumulation in the bone marrow correlated with the time course of RBC hemolysis, with an ∼10-fold increase in bone marrow iron at 14-21 days post-irradiation, primarily within the cytoplasm of macrophages. Iron accumulation in the bone marrow was associated with increased expression of genes for iron binding and transport proteins, including transferrin, transferrin receptor 1, ferroportin, and integrin αMß2. Expression of the gene encoding Nrf2, a transcription factor activated by oxidative stress, also increased at 21 days post-irradiation. Captopril did not alter iron accumulation in the bone marrow or expression of iron storage genes, but did suppress Nrf2 expression. Our study suggests that following TBI, iron is deposited in tissues not normally associated with iron storage, which may be a secondary mechanism of radiation-induced tissue injury.


Asunto(s)
Síndrome de Radiación Aguda/metabolismo , Médula Ósea/metabolismo , Rayos gamma/efectos adversos , Hematopoyesis/efectos de la radiación , Hierro/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Síndrome de Radiación Aguda/genética , Síndrome de Radiación Aguda/patología , Animales , Médula Ósea/patología , Captopril/farmacología , Eritrocitos/metabolismo , Eritrocitos/patología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Ratones , Ratones Transgénicos , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Traumatismos Experimentales por Radiación/genética , Traumatismos Experimentales por Radiación/patología
4.
Am J Physiol Gastrointest Liver Physiol ; 318(3): G439-G450, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31961718

RESUMEN

Methionine is an essential amino acid needed for a variety of processes in living organisms. Ionizing radiation depletes tissue methionine concentrations and leads to the loss of DNA methylation and decreased synthesis of glutathione. In this study, we aimed to investigate the effects of methionine dietary supplementation in CBA/CaJ mice after exposure to doses ranging from 3 to 8.5 Gy of 137Cs of total body irradiation. We report that mice fed a methionine-supplemented diet (MSD; 19.5 vs. 6.5 mg/kg in a methionine-adequate diet, MAD) developed acute radiation toxicity at doses as low as 3 Gy. Partial body irradiation performed with hindlimb shielding resulted in a 50% mortality rate in MSD-fed mice exposed to 8.5 Gy, suggesting prevalence of radiation-induced gastrointestinal syndrome in the development of acute radiation toxicity. Analysis of the intestinal microbiome demonstrated shifts in the gut ecology, observed along with the development of leaky gut syndrome and bacterial translocation into the liver. Normal gut physiology impairment was facilitated by alterations in the one-carbon metabolism pathway and was exhibited as decreases in circulating citrulline levels mirrored by decreased intestinal mucosal surface area and the number of surviving crypts. In conclusion, we demonstrate that a relevant excess of methionine dietary intake exacerbates the detrimental effects of exposure to ionizing radiation in the small intestine.NEW & NOTEWORTHY Methionine supplementation, instead of an anticipated health-promoting effect, sensitizes mice to gastrointestinal radiation syndrome. Mechanistically, excess of methionine negatively affects intestinal ecology, leading to a cascade of physiological, biochemical, and molecular alterations that impair normal gut response to a clinically relevant genotoxic stressor. These findings speak toward increasing the role of registered dietitians during cancer therapy and the necessity of a solid scientific background behind the sales of dietary supplements and claims regarding their benefits.


Asunto(s)
Síndrome de Radiación Aguda/etiología , Suplementos Dietéticos/toxicidad , Intestino Delgado/efectos de los fármacos , Metionina/toxicidad , Traumatismos Experimentales por Radiación/etiología , Síndrome de Radiación Aguda/metabolismo , Síndrome de Radiación Aguda/microbiología , Síndrome de Radiación Aguda/patología , Animales , Metilación de ADN/efectos de los fármacos , Disbiosis , Metabolismo Energético/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Intestino Delgado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Dosis de Radiación , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/microbiología , Traumatismos Experimentales por Radiación/patología , Factores de Riesgo , Irradiación Corporal Total
5.
Health Phys ; 111(2): 85-92, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27356050

RESUMEN

This article reviews studies of progenitor mobilization with gamma-tocotrienol (GT3), a tocol under advanced development as a radiation countermeasure for acute radiation syndrome (ARS). GT3 protects mice against high doses of ionizing radiation and induces high levels of granulocyte colony-stimulating factor (G-CSF). GT3-induced G-CSF in conjunction with AMD3100 (a chemokine receptor antagonist clinically used to improve the yield of mobilized progenitors) mobilizes progenitors; these mobilized progenitors mitigate injury when infused to mice exposed to acute, high-dose ionizing radiation. The administration of a G-CSF antibody to GT3-injected donor mice abrogated the radiomitigative efficacy of blood or peripheral blood mononuclear cells (PBMC) in irradiated recipient mice. The efficacy of GT3-injected donor mice blood or PBMC was comparable to a recently published article involving blood or mononuclear cells obtained from mice injected with G-CSF. The injected progenitors were found to localize in various tissues of irradiated hosts. The authors demonstrate the efficacy of a bridging therapy in a preclinical animal model that allows the lymphohematopoietic system of severely immunocompromised mice to recover. This suggests that GT3 is a highly effective agent for radioprotection and mobilizing progenitors with significant therapeutic potential. Therefore, GT3 may be considered for further translational development and ultimately for use in humans.


Asunto(s)
Síndrome de Radiación Aguda/patología , Síndrome de Radiación Aguda/terapia , Cromanos/administración & dosificación , Movilización de Célula Madre Hematopoyética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Vitamina E/análogos & derivados , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Células Madre Hematopoyéticas/citología , Masculino , Ratones , Protectores contra Radiación/administración & dosificación , Resultado del Tratamiento , Vitamina E/administración & dosificación
6.
Molecules ; 20(6): 11017-33, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-26083036

RESUMEN

Accidental exposure to uranium is a matter of concern, as U(VI) is nephrotoxic in both human and animal models, and its toxicity is associated to chemical toxicity instead of radioactivity. We synthesized different PAMAM G4 and G5 derivatives in order to prove their interaction with uranium and their effect on the viability of red blood cells in vitro. Furthermore, we prove the effectiveness of the selected dendrimers in an animal model of acute uranium intoxication. The dendrimer PAMAM G4-Lys-Fmoc-Cbz demonstrated the ability to chelate the uranyl ion in vivo, improving the biochemical and histopathologic features caused by acute intoxication with uranium.


Asunto(s)
Síndrome de Radiación Aguda/tratamiento farmacológico , Quelantes/administración & dosificación , Nanopartículas , Exposición a la Radiación/efectos adversos , Uranio/efectos adversos , Síndrome de Radiación Aguda/sangre , Síndrome de Radiación Aguda/patología , Animales , Biomarcadores , Quelantes/química , Dendrímeros/administración & dosificación , Dendrímeros/química , Modelos Animales de Enfermedad , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Hemólisis/efectos de los fármacos , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Riñón/efectos de la radiación , Masculino , Ratones , Nylons/química
7.
Radiat Res ; 181(5): 464-70, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24754562

RESUMEN

Metformin, a biguanide drug used in the treatment of type II diabetes, was evaluated alone and in combination with amifostine, captopril, MESNA or N-acetyl-cysteine (NAC) for its ability to protect when administered 24 h after irradiation. Mouse embryo fibroblasts (MEF), human microvascular endothelial cells (HMEC) and SA-NH mouse sarcoma cells were exposed to 4 Gy in vitro. C3H mice were exposed to 7 Gy and evaluated utilizing an endogenous spleen colony assay system. Amifostine and WR1065, administered 30 min prior to irradiation, were used as positive controls. Treatment of MEF, HMEC and SA-NH cells with metformin elevated survival levels by 1.4-, 1.5- and 1.3-fold compared to 1.9-, 1.8- and 1.6-fold for these same cells treated with WR1065, respectively. Metformin (250 mg/kg) was effective in protecting splenic cells from a 7 Gy dose in vivo (protection factor = 1.8). Amifostine (400 mg/kg), administered 30 min prior to irradiation resulted in a 2.6-fold survival elevation, while metformin administered 24 h after irradiation in combination with NAC (400 mg/kg), MESNA (300 mg/kg) or captopril (200 mg/kg) enhanced survival by 2.6-, 2.8- and 2.4-fold, respectively. Each of these agents has been approved by the FDA for human use and each has a well characterized human safety profile. Metformin alone or in combination with selected sulfhydryl agents possesses radioprotective properties when administered 24 h after radiation exposure comparable to that observed for amifostine administered 30 min prior to irradiation making it a potentially useful agent for radiation countermeasures use.


Asunto(s)
Síndrome de Radiación Aguda/prevención & control , Metformina/uso terapéutico , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Compuestos de Sulfhidrilo/uso terapéutico , Acetilcisteína/administración & dosificación , Acetilcisteína/uso terapéutico , Síndrome de Radiación Aguda/patología , Amifostina/administración & dosificación , Amifostina/farmacología , Amifostina/uso terapéutico , Animales , Captopril/administración & dosificación , Captopril/farmacología , Captopril/uso terapéutico , Línea Celular Transformada , Línea Celular Tumoral , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Depuradores de Radicales Libres/administración & dosificación , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Humanos , Mesna/administración & dosificación , Mesna/farmacología , Mesna/uso terapéutico , Metformina/administración & dosificación , Metformina/farmacología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/patología , Protectores contra Radiación/administración & dosificación , Protectores contra Radiación/farmacología , Sarcoma/patología , Compuestos de Sulfhidrilo/administración & dosificación , Compuestos de Sulfhidrilo/farmacología
8.
Radiat Res ; 181(5): 445-51, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24720754

RESUMEN

Exposure to a nuclear accident or radiological attack can cause death from acute radiation syndrome (ARS), which results from radiation injury to vital organs such as the hematopoietic system. However, the U.S. Food and Drug Administration (FDA) has not approved any medical countermeasures for this specific purpose. With growing concern over nuclear terrorism, there is an urgent need to develop small molecule deliverables that mitigate mortality from ARS. One emerging modulator of hematopoietic stem/progenitor cell (HSPC) activity is glycogen synthase kinase-3 (GSK-3). The inhibition of GSK-3 has been shown to augment hematopoietic repopulation in mouse models of bone marrow transplantation. In this study, we performed an in vitro screen using irradiated bone marrow mononuclear cells (BM-MNCs) to test the effects of four GSK-3 inhibitors: CHIR99021; 6-Bromoindirubin-3'-oxime (BIO); SB415286; and SB216763. This screen showed that SB216763 significantly increased the frequency of c-Kit(+) Lin(-) Sca1(+) (KLS) cells and hematopoietic colony-forming cells in irradiated BM-MNCs. Importantly, administration of a single dose of SB216763 to C57BL/6J mice by subcutaneous injection 24 h after total-body irradiation significantly improved hematopoietic recovery and mitigated hematopoietic ARS. Collectively, our results demonstrate that the GSK-3 inhibitor SB216763 is an effective medical countermeasure against acute radiation injury of the hematopoietic system.


Asunto(s)
Síndrome de Radiación Aguda/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Células Madre Hematopoyéticas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Síndrome de Radiación Aguda/enzimología , Síndrome de Radiación Aguda/patología , Aminofenoles/farmacología , Aminofenoles/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Médula Ósea/patología , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Evaluación Preclínica de Medicamentos , Glucógeno Sintasa Quinasa 3/fisiología , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/efectos de la radiación , Indoles/farmacología , Indoles/uso terapéutico , Inyecciones Subcutáneas , Maleimidas/farmacología , Maleimidas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Oximas/farmacología , Oximas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Piridinas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Traumatismos Experimentales por Radiación/enzimología , Traumatismos Experimentales por Radiación/patología , Irradiación Corporal Total/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA