Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 610
Filtrar
1.
Toxicol Appl Pharmacol ; 429: 115695, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34419493

RESUMEN

M3258 is the first selective inhibitor of the immunoproteasome subunit LMP7 (Large multifunctional protease 7) in early clinical development with the potential to improve therapeutic utility in patients of multiple myeloma (MM) or other hematological malignancies. Safety pharmacology studies with M3258 did not reveal any functional impairments of the cardiovascular system in several in vitro tests employing human cardiomyocytes and cardiac ion channels (including hERG), guinea pig heart refractory period and force contraction, and rat aortic contraction as well as in cardiovascular function tests in dogs. Following single dose M3258 administration to rats, no changes were observed on respiratory function by using whole body plethysmography, nor did it change (neuro)behavioral parameters in a battery of tests. Based on pivotal 4-week toxicity studies with daily oral dosing of M3258, the identified key target organs of toxicity were limited to the lympho-hematopoietic system in rats and dogs, and to the intestine with its local lymphoid tissues in dogs only. Importantly, the stomach, nervous system, heart, lungs, and kidneys, that may be part of clinically relevant toxicities as reported for pan-proteasome inhibitors, were spared with M3258. Therefore, it is anticipated that by targeting highly selective and potent inhibition of LMP7, the resulting favorable safety profile of M3258 together with the maintained potent anti-tumor activity as previously reported in mouse MM xenograft models, may translate into an improved benefit-risk profile in MM patients.


Asunto(s)
Ácidos Borónicos/toxicidad , Furanos/toxicidad , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/toxicidad , Administración Oral , Animales , Ácidos Borónicos/administración & dosificación , Células Cultivadas , Perros , Femenino , Furanos/administración & dosificación , Cobayas , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/patología , Humanos , Intestinos/efectos de los fármacos , Intestinos/patología , Sistema Linfático/efectos de los fármacos , Sistema Linfático/patología , Masculino , Inhibidores de Proteasoma/administración & dosificación , Ratas Wistar , Medición de Riesgo , Especificidad de la Especie , Pruebas de Toxicidad
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
J Radiat Res ; 60(4): 432-441, 2019 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-31165150

RESUMEN

Exposure to ionizing radiation leads to severe damages in radiosensitive organs and induces acute radiation syndrome, including effects on the hematopoietic system and gastrointestinal system. In this study, the radioprotective ability of KMRC011, a novel toll-like receptor 5 (TLR5) agonist, was investigated in C57BL6/N mice exposed to lethal total-body gamma-irradiation. In a 30-day survival study, KMRC011-treated mice had a significantly improved survival rate compared with control after 11 Gy total-body irradiation (TBI), and it was found that the radioprotective activity of KMRC011 depended on its dosage and repeated treatment. In a 5-day short-term study, we demonstrated that KMRC011 treatment stimulated cell proliferation and had an anti-apoptotic effect. Furthermore, KMRC011 increased the expressions of genes related to DNA repair, such as Rad21, Gadd45b, Sod2 and Irg1, in the small intestine of lethally irradiated mice. Interestingly, downregulation of NF-κB p65 in the mouse intestine by KMRC011 treatment was observed. This data indicated that KMRC011 exerted a radioprotective activity partially by regulating NF-κB signaling. Finally, peak expression levels of G-CSF, IL-6, IFN-γ, TNF-α and IP-10 induced by KMRC011 treatment were different depending on the route of administration and type of cytokine. These cytokines could be used as candidate biomarkers for the evaluation of KMRC011 clinical efficacy. Our data indicated that KMRC011 has radioprotective activity in lethally irradiated mice and may be developed as a therapeutic agent for radioprotection.


Asunto(s)
Síndrome de Radiación Aguda/prevención & control , Fragmentos de Péptidos/farmacología , Protectores contra Radiación/farmacología , Receptor Toll-Like 5/agonistas , Irradiación Corporal Total , Animales , Apoptosis/efectos de los fármacos , Médula Ósea/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL10/metabolismo , Rayos gamma , Sistema Hematopoyético/efectos de los fármacos , Hidroliasas/metabolismo , Interferón gamma/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Péptidos/farmacología , Protección Radiológica , Tolerancia a Radiación/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
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
10.
Free Radic Biol Med ; 131: 382-392, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30578918

RESUMEN

Reactive oxygen species (ROS) play a critical role in total body irradiation (TBI)-induced hematopoietic system injury. However, the mechanisms involved in ROS production in hematopoietic stem cells (HSCs) post TBI need to be further explored. In this study, we demonstrated that hematopoietic system injury in mice radiated with TBI was effectively alleviated when the blood circulation environment was changed via the injection of serum from non-radiated mice. Serum injection increased the survival of radiated mice and ameliorated TBI-induced hematopoietic system injury through attenuating myeloid skew, increasing HSC frequency, and promoting the reconstitution of radiated HSCs. Serum injection also decreased ROS levels in HSCs and regulated oxidative stress-related proteins. A serum proteome sequence array showed that proteins related to tissue injury and oxidative stress were regulated, and a serum-derived exosome microRNA sequence assay showed that the PI3K-Akt and Hippo signaling pathways were affected in radiated mice injected with serum from non-radiated mice. Furthermore, a significant increase in cell viability and a decrease in ROS were observed in radiated lineage-c-kit+ cells treated with serum-derived exosomes. Similarly, an improvement in the impaired differentiation of HSCs was observed in radiated mice injected with serum-derived exosomes. Taken together, our observations suggest that serum from non-radiated mice alleviates HSC injury in radiated mice by improving the systemic environment after radiation, and exosomes contribute to this radioprotective effect as important serum active component.


Asunto(s)
Proteínas Sanguíneas/farmacología , Exosomas/trasplante , Células Madre Hematopoyéticas/efectos de los fármacos , Sistema Hematopoyético/efectos de los fármacos , Protectores contra Radiación/farmacología , Suero/fisiología , Animales , Proteínas Sanguíneas/química , Proteínas Sanguíneas/aislamiento & purificación , Relación Dosis-Respuesta en la Radiación , Exosomas/química , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Sistema Hematopoyético/citología , Sistema Hematopoyético/efectos de la radiación , Vía de Señalización Hippo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Suero/química , Transducción de Señal , Análisis de Supervivencia , Irradiación Corporal Total
11.
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
12.
Int J Mol Sci ; 19(5)2018 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-29883417

RESUMEN

Ionizing radiation (IR) acts as an external stimulating factor, when it acts on the body, it will activate NF- κ B and cause the up-regulation of inducible nitric oxide synthase (iNOS) and induce a large amount of nitric oxide (NO) production. NO and other reactive nitrogen and oxygen species (RNS and ROS) can cause damage to biological molecules and affect their physiological functions. Our study investigated the protective role of 2-amino-5,6-dihydro-4H-1,3-thiazine hydrobromide (2-ADT) and 2-acetylamino-5,6-dihydro-4H-1,3-thiazine hydrobromide (2-AADT), two nitric oxide synthase inhibitors, against radiation-induced hematopoietic and intestinal injury in mice. Pretreatment with 2-ADT and 2-AADT improved the survival of mice exposed to a lethal dose of radiation, especially, the survival rate of the 2-ADT 20 mg/kg group was significantly higher than that of the vehicle group (p < 0.001). Our findings indicated that the radioprotective actions of 2-ADT and 2-AADT are achieved via accelerating hematopoietic system recovery, decreasing oxidative and nitrosative stress by enhancing the antioxidant defense system and reducing NO as well as peroxynitrite (ONOO − ) content, and mitigating the radiation-induced DNA damage evaluated by comet assay. These results suggest that 2-ADT and 2-AADT may have great application potential in ameliorating the damages of radiotherapy.


Asunto(s)
Sistema Hematopoyético/lesiones , Intestinos/lesiones , Traumatismos por Radiación/prevención & control , Protectores contra Radiación/uso terapéutico , Tiazinas/uso terapéutico , Animales , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/metabolismo , Sistema Hematopoyético/efectos de la radiación , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/efectos de la radiación , Ratones , Óxido Nítrico/sangre , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Traumatismos por Radiación/sangre , Traumatismos por Radiación/metabolismo , Protectores contra Radiación/química , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tiazinas/química
13.
Health Phys ; 115(1): 65-76, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29787432

RESUMEN

Interleukin-11 was developed to reduce chemotherapy-induced thrombocytopenia; however, its clinical use was limited by severe adverse effects in humans. PEGylated interleukin-11 (BBT-059), developed by Bolder Biotechnology, Inc., exhibited a longer half-life in rodents and induced longer-lasting increases in hematopoietic cells than interleukin-11. A single dose of 1.2 mg kg of BBT-059, administered subcutaneously to CD2F1 mice (12-14 wk, male) was found to be safe in a 14 d toxicity study. The drug demonstrated its efficacy both as a prophylactic countermeasure and a mitigator in CD2F1 mice exposed to Co gamma total-body irradiation. A single dose of 0.3 mg kg, administered either 24 h pre-, 4 h post-, or 24 h postirradiation increased the survival of mice to 70-100% from lethal doses of radiation. Preadministration (-24 h) of the drug conferred a significantly (p < 0.05) higher survival compared to 24 h post-total-body irradiation. There was significantly accelerated recovery from radiation-induced peripheral blood neutropenia and thrombocytopenia in animals pretreated with BBT-059. The drug also increased bone marrow cellularity and megakaryocytes and accelerated multilineage hematopoietic recovery. In addition, BBT-059 inhibited the induction of radiation-induced hematopoietic biomarkers, thrombopoietin, erythropoietin, and Flt-3 ligand. These results indicate that BBT-059 is a promising radiation countermeasure, demonstrating its potential to be used both pre- and postirradiation for hematopoietic acute radiation syndrome with a broad window for medical management in a radiological or nuclear event.


Asunto(s)
Síndrome de Radiación Aguda/tratamiento farmacológico , Sistema Hematopoyético/efectos de los fármacos , Interleucina-11/administración & dosificación , Polietilenglicoles/química , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Irradiación Corporal Total/efectos adversos , Síndrome de Radiación Aguda/etiología , Animales , Relación Dosis-Respuesta en la Radiación , Sistema Hematopoyético/patología , Sistema Hematopoyético/efectos de la radiación , Interleucina-11/química , Masculino , Ratones , Traumatismos Experimentales por Radiación/etiología
14.
Ecotoxicol Environ Saf ; 152: 121-131, 2018 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-29407778

RESUMEN

Silver nanoparticles (Ag NPs) are known for their antibacterial properties and are used in a growing number of nano-enabled products, with inevitable concerns for releases to the environment. Nanoparticles may also be antigenic and toxic to the haematopoietic system, but the immunotoxic effect of Ag NPs on non-target species such as fishes is poorly understood. This study aimed to assess the effect of Ag NP exposure via the water on the haematopoietic system of rainbow trout, Oncorhynchus mykiss, and to determine whether or not the hazard from Ag NPs was different from that of AgNO3. Fish were exposed for 7 days to a control (dechlorinated Plymouth freshwater), dispersant control, 1µgl-1 Ag as AgNO3 or 100µgl-1 Ag NPs. Animals were sampled on days 0, 4 and 7 for haematology, tissue trace metal concentration, biochemistry for evidence of oxidative stress/inflammation in the spleen and histopathology of the blood cells and spleen. The Ag NP treatment significantly increased the haematocrit, but the haematological changes were within the normal physiological range of the animal. Thrombocytes in spleen prints at day 4, and melanomacrophage deposits at day 7 in the spleen, of Ag NP exposed-fish displayed significant increases compared to all the other treatments within the time point. A dialysis experiment confirmed that dissolution rates were very low and any pathology observed is likely from the NP form rather than dissolved metal released from it. Overall, the data showed subtle differences in the effects of Ag NPs compared to AgNO3 on the haematopoietic system. The lack of pathology in the circulating blood cells and melanomacrophage deposits in the spleen suggests a compensatory physiological effort by the spleen to maintain normal circulating haematology during Ag NP exposure.


Asunto(s)
Sistema Hematopoyético/efectos de los fármacos , Nanopartículas del Metal/toxicidad , Oncorhynchus mykiss/sangre , Nitrato de Plata/toxicidad , Plata/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Sistema Hematopoyético/patología , Modelos Teóricos , Estrés Oxidativo/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/patología
15.
Environ Mol Mutagen ; 59(1): 79-90, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28766757

RESUMEN

Ionizing radiation exposure is harmful and at high doses can lead to acute hematopoietic radiation syndrome. Therefore, agents that can protect hematopoietic system are important for development of radioprotector. Sesamol is a potential molecule for development of radioprotector due to its strong free radical scavenging and antioxidant properties. In the present study, sesamol was evaluated for its role in DNA damage and repair in hematopoietic system of γ-irradiated CB57BL/6 mice and compared with amifostine. C57BL/6 male mice were administered with sesamol 20 mg/kg (i.p.) followed by 2 Gy whole body irradiation (WBI) at 30 min. Mice were sacrificed at 0.5, 3, 24 h postirradiation; bone marrow, splenocytes, and peripheral blood lymphocytes were isolated to measure DNA damages and repair using alkaline comet,γ-H2AXand micronucleus assays. An increase in % of tail DNA was observed in all organs of WBI mice. Whereas in pre-administered sesamol reduced %DNA in tail (P ≤ 0.05). Sesamol has also reduced formation of radiation induced γ-H2AX foci after 0.5 h in these organs and further lowered to respective control values at 24 h of WBI. Similar reduction of % DNA in tail and γ-H2AX foci were observed with amifostine (P ≤ 0.05). Analysis of mnPCE frequency at 24 h has revealed similar extent of protection by sesamol and amifostine. Interestingly, both sesamol and amifostine, alone and with radiation, also increased the granulocytes count significantly compared to the control (P ≤ 0.05). These findings suggest that sesamol has strong potential to protect hematopoietic system by lowering radiation induced DNA damages and can prevent acute hematopoietic syndrome in mice. Environ. Mol. Mutagen. 59:79-90, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Benzodioxoles/farmacología , Daño del ADN/efectos de los fármacos , Rayos gamma/efectos adversos , Sistema Hematopoyético/efectos de los fármacos , Fenoles/farmacología , Protectores contra Radiación/farmacología , Irradiación Corporal Total/efectos adversos , Animales , Antioxidantes/farmacología , ADN/efectos de los fármacos , Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Pruebas de Micronúcleos/métodos
16.
Toxicol Pathol ; 45(7): 879-883, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28990496

RESUMEN

New medullary bone formation has been observed in rats administered a variety of antineoplastic compounds. Similar effects reported in rats administered granulocyte colony-stimulating factor (G-CSF) were attributed to exaggerated pharmacology of G-CSF as a cytokine and growth factor, resulting in stromal proliferation in addition to the intended hematopoietic effects. Similar phenomena of marrow stromal change are reported among other species in association with various growth factors. Case study summaries of test item-related histopathologic changes in bone marrow, reflecting trabecular and/or endosteal new bone formation, are presented. In each of these cases, it was concluded that the new medullary bone and stromal proliferation did not reflect a primary target-related toxicity; rather, the mesenchymal changes were attributed to nonspecific, secondary effects of cytokines elaborated in response to primary cytotoxic effects on hematopoietic cells with subsequent impact on circulating blood cells. The common features associated with marrow stromal changes in the case studies, as well as with a variety of pharmacologic compounds across several species described in the literature, are hematologic effects and/or changes in growth factor levels and cytokine expression.


Asunto(s)
Antineoplásicos/efectos adversos , Médula Ósea/efectos de los fármacos , Citocinas/metabolismo , Sistema Hematopoyético/efectos de los fármacos , Animales , Antineoplásicos/administración & dosificación , Médula Ósea/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/efectos adversos , Sistema Hematopoyético/metabolismo , Hemoglobinas/metabolismo , Humanos , Masculino , Ratones , Modelos Animales , Neutrófilos/metabolismo , Ratas
17.
Cell Physiol Biochem ; 43(2): 457-464, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28922655

RESUMEN

BACKGROUND/AIMS: The hematopoietic system is vulnerable to ionizing radiation and is often severely damaged by radiation. Molecules affecting radioresistance include Toll-like receptor 2. We investigated whether Zymosan-A, a novel TLR2 agonist, can protect the hematopoietic system from radiation-induced damage after total body irradiation. METHODS: Mice were exposed to total body radiation after treatment with Zymosan-A or normal saline, and their survival was recorded. Tissue damage was evaluated by hematoxylin-eosin staining. The number of nucleated cells in bone marrow was determined by flow cytometry. Cell viability and apoptosis assay were determined by CCK-8 assay and flow cytometry assay. Enzyme-linked immunosorbent assay was used to detect the level of cytokines. RESULTS: Zymosan-A protected mice from radiation-induced death and prevented radiation-induced hematopoietic system damage. Zymosan-A also promoted cell viability and inhibited cell apoptosis caused by radiation, induced radioprotective effects via TLR2, upregulated IL-6, IL-11, IL-12, and TNF-α in vivo. CONCLUSION: Zymosan-A can provide protection against radiation-induced hematopoietic system damage by targeting the TLR2 signaling pathway. Thus, Zymosan-A can be potentially effective radioprotectant.


Asunto(s)
Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/efectos de la radiación , Protectores contra Radiación/farmacología , Receptor Toll-Like 2/metabolismo , Zimosan/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular , Sistema Hematopoyético/patología , Masculino , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
18.
Sci Rep ; 7(1): 9777, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28852188

RESUMEN

Concern over potential exposures of ionizing radiation (IR) to large populations has emphasized the need for rapid and reliable methods of biodosimetry to determine absorbed dose and required triage. Lipidomics has emerged as a powerful technique for large-scale lipid identification and quantification. Indirect effects from IR exposure generate reactive oxygen species (ROS) through water hydrolysis and may subsequently damage cellular lipids. Thus, rapid identification of specific affected lipid molecules represents possible targets for biodosimetry. The current study addresses temporal changes in the serum lipidome from 4 h to 28 d in nonhuman primates (NHPs) with radiation-induced hematopoietic syndrome (6.5 Gy exposure, LD50/30). Statistical analyses revealed a highly dynamic temporal response in the serum lipidome after IR exposure. Marked lipidomic perturbations occurred within 24 h post-irradiation along with increases in cytokine levels and C-reactive protein. Decreases were observed in di- and triacylglycerides, sphingomyelins (SMs), lysophosphatidylcholines (LysoPCs), and esterified sterols. Conversely, free fatty acids and monoacylglycerides significantly increased. Decreased levels of SMs and increased levels of LysoPCs may be important markers for biodosimetry ~2 d-3 d post-irradiation. The biphasic and dynamic response to the serum lipidome post-irradiation emphasize the importance of determining the temporal long-term response of possible radiation markers.


Asunto(s)
Enfermedades Hematológicas/etiología , Enfermedades Hematológicas/metabolismo , Metabolismo de los Lípidos , Metaboloma , Metabolómica , Traumatismos por Radiación/metabolismo , Radiación Ionizante , Algoritmos , Animales , Biomarcadores , Biología Computacional/métodos , Modelos Animales de Enfermedad , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/metabolismo , Metabolómica/métodos , Primates , Espectrometría de Masas en Tándem
19.
Nat Commun ; 8: 15990, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28748922

RESUMEN

An outstanding question in animal development, tissue homeostasis and disease is how cell populations adapt to sensory inputs. During Drosophila larval development, hematopoietic sites are in direct contact with sensory neuron clusters of the peripheral nervous system (PNS), and blood cells (hemocytes) require the PNS for their survival and recruitment to these microenvironments, known as Hematopoietic Pockets. Here we report that Activin-ß, a TGF-ß family ligand, is expressed by sensory neurons of the PNS and regulates the proliferation and adhesion of hemocytes. These hemocyte responses depend on PNS activity, as shown by agonist treatment and transient silencing of sensory neurons. Activin-ß has a key role in this regulation, which is apparent from reporter expression and mutant analyses. This mechanism of local sensory neurons controlling blood cell adaptation invites evolutionary parallels with vertebrate hematopoietic progenitors and the independent myeloid system of tissue macrophages, whose regulation by local microenvironments remain undefined.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Hematopoyesis , Sistema Hematopoyético/metabolismo , Hemocitos/metabolismo , Subunidades beta de Inhibinas/metabolismo , Larva/crecimiento & desarrollo , Células Receptoras Sensoriales/metabolismo , Animales , Carbacol/farmacología , Supervivencia Celular , Microambiente Celular , Agonistas Colinérgicos/farmacología , Proteínas de Drosophila/efectos de los fármacos , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/metabolismo , Sistema Hematopoyético/efectos de los fármacos , Hemocitos/efectos de los fármacos , Larva/efectos de los fármacos , Larva/metabolismo , Sistema Nervioso Periférico/efectos de los fármacos , Sistema Nervioso Periférico/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos
20.
Int J Mol Sci ; 18(7)2017 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-28657605

RESUMEN

In recent times, cytokines and hematopoietic growth factors have been at the center of attention for many researchers trying to establish pharmacological therapeutic procedures for the treatment of radiation accident victims. Two granulocyte colony-stimulating factor-based radiation countermeasures have been approved for the treatment of the hematopoietic acute radiation syndrome. However, at the same time, many different substances with varying effects have been tested in animal studies as potential radioprotectors and mitigators of radiation damage. A wide spectrum of these substances has been studied, comprising various immunomodulators, prostaglandins, inhibitors of prostaglandin synthesis, agonists of adenosine cell receptors, herbal extracts, flavonoids, vitamins, and others. These agents are often effective, relatively non-toxic, and cheap. This review summarizes the results of animal experiments, which show the potential for some of these untraditional or new radiation countermeasures to become a part of therapeutic procedures applicable in patients with the acute radiation syndrome. The authors consider ß-glucan, 5-AED (5-androstenediol), meloxicam, γ-tocotrienol, genistein, IB-MECA (N6-(3-iodobezyl)adenosine-5'-N-methyluronamide), Ex-RAD (4-carboxystyryl-4-chlorobenzylsulfone), and entolimod the most promising agents, with regards to their contingent use in clinical practice.


Asunto(s)
Síndrome de Radiación Aguda/prevención & control , Citocinas/metabolismo , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/metabolismo , Protectores contra Radiación/uso terapéutico , Síndrome de Radiación Aguda/tratamiento farmacológico , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...