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











Base de datos
Intervalo de año de publicación
1.
Biol Trace Elem Res ; 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39080232

RESUMEN

Two experiments were conducted to examine the impact of trace mineral (TM) source on in vitro and in vivo solubility characteristics. Experiment 1: Hydroxy TM (HTM) and sulfate TM (STM) sources of Cu, Mn, and Zn were incubated separately in water for 24 h. Immediately after mixing, initial pH of each solution was greater (P < 0.03) for HTM compared to STM for all elements. Final pH tended to be greater for Cu (P = 0.09) and Zn (P = 0.07) from HTM compared to STM. Water solubility of Cu, Mn, and Zn from STM was greater (P < 0.01) than HTM sources. Experiment 2: Eight steers fitted with rumen cannula were blocked by body weight and randomly assigned to treatments. Treatments consisted of 10 mg Cu, 40 mg Mn, and 60 mg Zn/kg DM from either STM or HTM sources. Steers were individually fed a cracked corn-corn silage-based diet. Treatments were top-dressed daily. Rumen contents were collected at 0, 2, and 4 h post-feeding on d 1 and 14. On d 15, strained ruminal fluid and particle-associated microorganisms were obtained. Zinc was more tightly bound (P = 0.01) to the digesta in HTM-supplemented steers compared to STM on d 14. These data indicate that TM source influences pH and solubility of Cu, Mn, and Zn in water and may affect rumen soluble Cu concentrations and binding strength of Zn to solid digesta.

2.
Transl Anim Sci ; 8: txae045, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38585172

RESUMEN

One hundred and eighty crossbred beef steers (406.0 ±â€…2.2 kg) were used to determine the impact of a novel direct-fed microbial (DFM) on growth performance, carcass characteristics, rumen fermentation characteristics, and immune response in finishing beef cattle. Steers were blocked by body weight (BW) and randomly assigned, within block, to 1 of 2 treatments (3 replicates/treatment: 30 steers/replicate). Treatments included: (1) no DFM (control) and (2) DFM supplementation at 50 mg ∙ animal-1 ∙ d-1 (BOVAMINE DEFEND Plus). All steers were fed a high-concentrate finishing diet and individual feed intake was recorded daily via the GrowSafe system. BWs were collected every 28 d. On day 55, 10 steers per pen were injected with ovalbumin (OVA). Jugular blood samples were collected from each steer on days 0, 7, 14, and 21 post injection. On day 112, the same steers were injected again with OVA and intramuscularly with a pig red blood cell solution. Jugular blood samples were collected from each steer on days 0, 7, 14, and 21 post injection. On day 124, rumen fluid was collected from 3 steers per treatment and used to estimate in vitro rumen fermentation characteristics. Equal numbers of steers per treatment were transported to a commercial abattoir on days 145, 167, and 185 of the experiment, harvested, and carcass data were collected. Initial BW was similar across treatments. On days 28 and 55, steers receiving DFM had heavier BW (P < 0.01) compared to controls. The average daily gain was greater in DFM-supplemented steers from days 0 to 28 (P < 0.01) and days 0 to 55 (P < 0.01) of the experiment compared to controls. Overall dry matter intake (DMI) was greater (P < 0.04) and overall feed efficiency was similar in DFM-supplemented steers compared to controls. Dressing percentage (P < 0.02) was greater in steers receiving DFM compared to controls. Antibody titers to injected antigens were similar across treatments. However, red blood cell superoxide dismutase activity was greater (P < 0.05) in DFM-supplemented steers compared to controls. In vitro molar proportions of isobutyric and butyric acid were greater (P < 0.01) and dry matter (DM) digestibility tended (P < 0.07) to be greater in rumen fluid obtained from steers supplemented with DFM. These data suggest that BOVAMINE DEFEND Plus supplementation improves growth performance during the initial period of the finishing phase, increases overall DMI and dressing percentage, and may impact antioxidant status in beef cattle.

3.
Cells ; 13(1)2023 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-38201243

RESUMEN

To better understand radiation-induced organ dysfunction at both high and low doses, it is critical to understand how endothelial cells (ECs) respond to radiation. The impact of irradiation (IR) on ECs varies depending on the dose administered. High doses can directly damage ECs, leading to EC impairment. In contrast, the effects of low doses on ECs are subtle but more complex. Low doses in this study refer to radiation exposure levels that are below those that cause immediate and necrotic damage. Mitochondria are the primary cellular components affected by IR, and this study explored their role in determining the effect of radiation on microvascular endothelial cells. Human dermal microvascular ECs (HMEC-1) were exposed to varying IR doses ranging from 0.1 Gy to 8 Gy (~0.4 Gy/min) in the AFRRI 60-Cobalt facility. Results indicated that high doses led to a dose-dependent reduction in cell survival, which can be attributed to factors such as DNA damage, oxidative stress, cell senescence, and mitochondrial dysfunction. However, low doses induced a small but significant increase in cell survival, and this was achieved without detectable DNA damage, oxidative stress, cell senescence, or mitochondrial dysfunction in HMEC-1. Moreover, the mitochondrial morphology was assessed, revealing that all doses increased the percentage of elongated mitochondria, with low doses (0.25 Gy and 0.5 Gy) having a greater effect than high doses. However, only high doses caused an increase in mitochondrial fragmentation/swelling. The study further revealed that low doses induced mitochondrial elongation, likely via an increase in mitochondrial fusion protein 1 (Mfn1), while high doses caused mitochondrial fragmentation via a decrease in optic atrophy protein 1 (Opa1). In conclusion, the study suggests, for the first time, that changes in mitochondrial morphology are likely involved in the mechanism for the radiation dose-dependent effect on the survival of microvascular endothelial cells. This research, by delineating the specific mechanisms through which radiation affects endothelial cells, offers invaluable insights into the potential impact of radiation exposure on cardiovascular health.


Asunto(s)
Enfermedades Mitocondriales , Traumatismos por Radiación , Humanos , Células Endoteliales , Supervivencia Celular , Mitocondrias , Senescencia Celular , Proteínas Mitocondriales
4.
Oxid Med Cell Longev ; 2020: 6401341, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33014275

RESUMEN

Human SARS-CoV-2 infection is characterized by a high mortality rate due to some patients developing a large innate immune response associated with a cytokine storm and acute respiratory distress syndrome (ARDS). This is characterized at the molecular level by decreased energy metabolism, altered redox state, oxidative damage, and cell death. Therapies that increase levels of (R)-beta-hydroxybutyrate (R-BHB), such as the ketogenic diet or consuming exogenous ketones, should restore altered energy metabolism and redox state. R-BHB activates anti-inflammatory GPR109A signaling and inhibits the NLRP3 inflammasome and histone deacetylases, while a ketogenic diet has been shown to protect mice from influenza virus infection through a protective γδ T cell response and by increasing electron transport chain gene expression to restore energy metabolism. During a virus-induced cytokine storm, metabolic flexibility is compromised due to increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) that damage, downregulate, or inactivate many enzymes of central metabolism including the pyruvate dehydrogenase complex (PDC). This leads to an energy and redox crisis that decreases B and T cell proliferation and results in increased cytokine production and cell death. It is hypothesized that a moderately high-fat diet together with exogenous ketone supplementation at the first signs of respiratory distress will increase mitochondrial metabolism by bypassing the block at PDC. R-BHB-mediated restoration of nucleotide coenzyme ratios and redox state should decrease ROS and RNS to blunt the innate immune response and the associated cytokine storm, allowing the proliferation of cells responsible for adaptive immunity. Limitations of the proposed therapy include the following: it is unknown if human immune and lung cell functions are enhanced by ketosis, the risk of ketoacidosis must be assessed prior to initiating treatment, and permissive dietary fat and carbohydrate levels for exogenous ketones to boost immune function are not yet established. The third limitation could be addressed by studies with influenza-infected mice. A clinical study is warranted where COVID-19 patients consume a permissive diet combined with ketone ester to raise blood ketone levels to 1 to 2 mM with measured outcomes of symptom severity, length of infection, and case fatality rate.


Asunto(s)
Infecciones por Coronavirus/terapia , Síndrome de Liberación de Citoquinas/terapia , Dieta Cetogénica/métodos , Cetonas/administración & dosificación , Neumonía Viral/terapia , Ácido 3-Hidroxibutírico/metabolismo , Inmunidad Adaptativa , Animales , Betacoronavirus , COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/metabolismo , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/metabolismo , Metabolismo Energético , Humanos , Inmunidad Innata , Cetonas/metabolismo , Oxidación-Reducción , Pandemias , Neumonía Viral/inmunología , Neumonía Viral/metabolismo , SARS-CoV-2
6.
Int J Radiat Biol ; 93(9): 907-919, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28749244

RESUMEN

PURPOSE: Phenylbutyrate (PB), a histone deacetylase inhibitor (HDACi) has demonstrated radiation protection in both in vitro and in vivo models. Studies previously demonstrated that PB and other HDAC inhibitors could inhibit radiation lethality in vivo by subcutaneous (s.c) injection. The objective of this study was to test the ability of oral PB treatment to protect against or to mitigate acute gamma radiation-induced lethality in vivo. MATERIALS AND METHODS: Human osteoblasts cells were used to evaluate radiation survival when PB was delivered pre- or post-radiation. A 30-day radiation lethality study was used to assess the radioprotective (pre-radiation) and radiomitigative (post-radiation) capability of PB. Possible mechanisms evaluated were antioxidant activity effects, HDAC inhibition, DNA damage, and hematological recovery. RESULTS: Treatment of HOS cells with PB 50 µM either before or after radiation increased radiation resistance as assessed by clonogenic survival. Western blot studies showed that PB treatment acetylated histones in vivo and ameliorated the radiation-induced reduction in acetylated histone-4 (H4). Pre-radiation oral administration of PB (10 mg/kg) provided radioprotection against gamma radiation (7-11.5 Gy) with a dose reduction factor of 1.25 (p = 0.001). PB oral administration post-radiation provided moderate radiation mitigation against gamma radiation (7-11.5 Gy) and demonstrated a dose reduction factor of 1.18 (p = 0.05). PB pre-radiation and post-radiation treatment was associated with significant elevations in neutrophils and platelets and attenuation of DNA damage. CONCLUSIONS: These results indicate that oral PB has potential as a radiation protector and a radiation mitigator and that potential mechanisms of action include attenuation of DNA damage, antioxidant activity, and bone marrow protection.


Asunto(s)
Daño del ADN/efectos de los fármacos , Rayos gamma , Osteoblastos/efectos de los fármacos , Osteoblastos/efectos de la radiación , Fenilbutiratos/farmacología , Traumatismos por Radiación/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Administración Oral , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Estudios de Factibilidad , Humanos , Dosificación Letal Mediana , Masculino , Ratones , Ratones Endogámicos DBA , Osteoblastos/citología , Osteoblastos/fisiología , Fenilbutiratos/efectos adversos , Dosis de Radiación , Traumatismos por Radiación/diagnóstico , Protectores contra Radiación/efectos adversos , Protectores contra Radiación/farmacología , Tasa de Supervivencia , Resultado del Tratamiento
7.
Toxicol Appl Pharmacol ; 331: 135-141, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28602947

RESUMEN

Depleted uranium (DU) is a radioactive heavy metal used primarily in military applications. Published data from our laboratory have demonstrated that DU exposure in vitro to immortalized human osteoblast cells (HOS) is both neoplastically transforming and genotoxic. In vivo studies have also demonstrated that DU is leukemogenic and genotoxic. DU possesses both a radiological (alpha particle) and chemical (metal) component but is generally considered a chemical biohazard. Studies have shown that alpha particle radiation does play a role in DU's toxic effects. Evidence has accumulated that non-irradiated cells in the vicinity of irradiated cells can have a response to ionization events. The purpose of this study was to determine if these "bystander effects" play a role in DU's toxic and neoplastic effects using HOS cells. We investigated the bystander responses between DU-exposed cells and non-exposed cells by co-culturing the two equal populations. Decreased cell survival and increased neoplastic transformation were observed in the non-DU exposed cells following 4 or 24h co-culture. In contrast Ni (II)- or Cr(VI)- exposed cells were unable to alter those biological effects in non-Ni(II) or non-Cr(VI) exposed co-cultured cells. Transfer experiments using medium from the DU-exposed and non-exposed co-cultured cells was able to cause adverse biological responses in cells; these results demonstrated that a factor (s) is secreted into the co-culture medium which is involved in this DU-associated bystander effect. This novel effect of DU exposure could have implications for radiation risk and for health risk assessment associated with DU exposure.


Asunto(s)
Efecto Espectador/efectos de los fármacos , Efecto Espectador/efectos de la radiación , Osteoblastos/efectos de los fármacos , Osteoblastos/efectos de la radiación , Exposición a la Radiación/efectos adversos , Uranio/toxicidad , Efecto Espectador/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/efectos de la radiación , Técnicas de Cocultivo/métodos , Humanos , Osteoblastos/fisiología , Nitrato de Uranilo/toxicidad
8.
Radiat Environ Biophys ; 50(4): 585-96, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21892632

RESUMEN

The histone deacetylase inhibitor (HDAC), phenylbutyrate (PB), is a novel anti-tumor agent. Studies have demonstrated that HDAC inhibitors can suppress cutaneous radiation syndrome and stimulate hematopoiesis. The objective of this study was to test the ability of PB treatment to protect against acute gamma-radiation-induced lethality in the DBA/2 mouse model. A 30-day radiation lethality study was used to assess radioprotective capability of PB. Mechanisms were evaluated using western blots, flow cytometry, and the single-cell gel electrophoresis assay. Western blot studies showed that PB treatment acetylated histones in vivo. For radiation protection studies, prophylactic administration of PB (24 h preradiation; 1-50 mg/kg) provided radioprotection against gamma radiation (8-9.5 Gy) and PB demonstrated a DRF of 1.31 (P = 0.001; 95% confidence interval: 1.27, 1.36). When PB (10 mg/kg) was administered post-radiation (4 h), it also provided significant radioprotection at 8.0 Gy radiation (P = 0.022). PB treatment before radiation was associated with significant elevations in neutrophils and platelets following radiation. Results from single-cell gel electrophoresis of peripheral blood leukocytes demonstrated that PB treatment before radiation can attenuate DNA damage and inhibit radiation-induced apoptosis. These results indicate that an HDAC inhibitor like PB has potential as a radiation protector and that mechanisms of action include attenuation of DNA damage and inhibition of apoptosis.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Fenilbutiratos/farmacología , Protectores contra Radiación/farmacología , Animales , Plaquetas/efectos de los fármacos , Plaquetas/efectos de la radiación , Daño del ADN , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Eritrocitos/efectos de la radiación , Hematopoyesis/efectos de los fármacos , Hematopoyesis/efectos de la radiación , Inhibidores de Histona Desacetilasas/toxicidad , Masculino , Ratones , Fenilbutiratos/toxicidad , Protectores contra Radiación/toxicidad , Tasa de Supervivencia , Pruebas de Toxicidad Aguda
9.
Health Phys ; 99(3): 371-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20699700

RESUMEN

Depleted uranium (DU) is an alpha particle emitter and radioactive heavy metal used in military applications. Due to internalization of DU during military operations and the ensuing chronic internal exposure to DU, there are concerns regarding its potential health effects. Preconceptional paternal irradiation has been implicated as a causal factor in childhood cancer and it has been suggested that this paternal exposure to radiation may play a role in the occurrence of leukemia and other cancers to offspring. Similarly, in vivo heavy metal studies have demonstrated that carcinogenic effects can occur in unexposed offspring. Using a transgenic mouse system employing a lambda shuttle vector allowing mutations (in the lacI gene) to be analyzed in vitro, we have investigated the possibility that chronic preconceptional paternal DU exposure can lead to transgenerational transmission of genomic instability. The mutation frequencies in vector recovered from the bone marrow cells of the F1 offspring of male parents exposed to low, medium, and high doses of internalized DU for 7 mo were evaluated and compared to control, tantalum, nickel, and gamma radiation F1 samples. Results demonstrate that as paternal DU-dose increased there was a trend towards higher mutation frequency in vector recovered from the DNA obtained from bone marrow of F1 progeny; medium and high dose DU exposure to P1 fathers resulted in a significant increase in mutation frequency in F1 offspring (3.57 +or - 0.37 and 4.81 + or - 0.43 x 10; p < 0.001) in comparison to control (2.28 + or - 0.31 x 10). The mutation frequencies from F1 offspring of low dose DU, Ta- or Ni-implanted fathers (2. 71 + or - 0.35, 2.38 + or - 0.35, and 2.93 + or - 0.39 x 10, respectively) were not significantly different than control levels (2.28 + or - 0.31 x 10). Offspring from Co (4 Gy) irradiated fathers did demonstrate an increased lacI mutation frequency (4.69 + or - 0.48 x 10) as had been shown previously. To evaluate the role of radiation involved in the observed DU effects, males were exposed to equal concentrations (50 mg U L) of either enriched uranium or DU in their drinking water for 2 mo prior to breeding. A comparison of these offspring indicated that there was a specific-activity dependent increase in offspring bone marrow mutation frequency. Taken together these uranyl nitrate data support earlier results in other model systems showing that radiation can play a role in DU-induced biological effects in vitro. However, since the lacI mutation model measures point mutations and cannot measure large deletions that are characteristic of radiation damage, the role of DU chemical effects in the observed offspring mutation frequency increase may also be significant. Regardless of the question of DU-radiation vs. DU-chemical effects, the data indicate that there exists a route for transgenerational transmission of factor(s) leading to genomic instability in F1 progeny from DU-exposed fathers.


Asunto(s)
Inestabilidad Genómica/genética , Inestabilidad Genómica/efectos de la radiación , Neoplasias Inducidas por Radiación/inducido químicamente , Exposición Paterna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/genética , Uranio/toxicidad , Partículas alfa , Animales , Médula Ósea/metabolismo , Médula Ósea/efectos de la radiación , Daño del ADN/genética , Relación Dosis-Respuesta en la Radiación , Femenino , Represoras Lac/genética , Leucemia/inducido químicamente , Leucemia/genética , Leucemia/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Uranio/metabolismo
10.
Biochimie ; 91(10): 1328-30, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19324073

RESUMEN

OBJECTIVES: The radioactive heavy metal depleted uranium (DU) is used in kinetic-energy penetrators in military applications. The objective of this study was to determine involvement of DNA methylation in DU-induced leukemia. METHODS: Methylation was measured by direct analysis of 5-methylcytosine content of spleen DNA in DU leukemic mice. RESULTS: Spleen hypomethylation occurred during DU-induced leukemogenesis (chronic internal DU exposure). Aberrant gene transcription was also detected. CONCLUSIONS: Epigenetic mechanisms are implicated in DU-induced leukemia. These data are evidence of aberrant DNA hypomethylation being associated with DU leukemogenesis.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Leucemia/inducido químicamente , Leucemia/genética , Uranio/toxicidad , Animales , Northern Blotting , Masculino , Ratones
11.
Rev Environ Health ; 22(1): 75-89, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17508699

RESUMEN

The use of depleted uranium in armor-penetrating munitions remains a source of controversy because of the numerous unanswered questions about its long-term health effects. Although no conclusive epidemiologic data have correlated DU exposure to specific health effects, studies using cultured cells and laboratory rodents continue to suggest the possibility of leukemogenic, genetic, reproductive, and neurological effects from chronic exposure. Until issues of concern are resolved with further research, the use of depleted uranium by the military will continue to be controversial.


Asunto(s)
Exposición a Riesgos Ambientales , Uranio/efectos adversos , Guerra , Animales , Humanos , Personal Militar , Neoplasias Inducidas por Radiación , Contaminantes Radiactivos/farmacocinética , Ratas , Ratas Sprague-Dawley , Uranio/farmacocinética
12.
Mol Cell Biochem ; 279(1-2): 97-104, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16283518

RESUMEN

Depleted uranium (DU) is a dense heavy metal used in military applications. During military conflicts, US military personnel have been wounded by DU shrapnel. The health effects of embedded DU are unknown. Published data from our laboratory demonstrated that DU exposure in vitro can transform immortalized human osteoblast cells (HOS) to the tumorigenic phenotype. Results from our laboratory have also shown that DU is genotoxic and mutagenic in cultured human cells. Internalized DU could be a carcinogenic risk and concurrent alpha particle and heavy metal toxic effects complicate this potential risk. Anecdotal reports have suggested that DU can cause leukemia. To better assess this risk, we have developed an in vivo leukemogenesis model. This model involves using murine hematopoietic cells (FDC-P1) that are dependent on stimulation by granulocyte-macrophage colony stimulating factor (GM-CSF) or interleukin 3 (IL-3) and injected into mice to produce myeloid leukemia. Although immortalized, these cells are not tumorigenic on subcutaneous inoculation in mice. Intravenous injection of FDC-P1 cells into DU-implanted DBA/2 mice was followed by the development of leukemias in 76% of all mice implanted with DU pellets. In contrast, only 12% of control mice developed leukemia. Karyotypic analysis confirmed that the leukemias originated from FDC-P1 cells. The growth properties of leukemic cells from bone marrow, spleen, and lymph node were assessed and indicate that the FDC-P1 cells had become transformed in vivo. The kidney, spleen, bone marrow, muscle, and urine showed significant elevations in tissue uranium levels prior to induction of leukemia. These results demonstrated that a DU altered in vivo environment may be involved in the pathogenesis of DU induced leukemia in an animal model.


Asunto(s)
Carcinógenos/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Células Madre Hematopoyéticas/efectos de los fármacos , Leucemia Experimental/inducido químicamente , Uranio/toxicidad , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Cariotipificación , Leucemia Experimental/genética , Leucemia Experimental/patología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos DBA , Modelos Animales , Células Progenitoras Mieloides/efectos de los fármacos , Bazo/metabolismo , Bazo/patología , Células Tumorales Cultivadas , Uranio/administración & dosificación , Irradiación Corporal Total
13.
Environ Health Perspect ; 113(6): 729-34, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15929896

RESUMEN

Continuing concern regarding the potential health and environmental effects of depleted uranium and lead has resulted in many countries adding tungsten alloy (WA)-based munitions to their battlefield arsenals as replacements for these metals. Because the alloys used in many munitions are relatively recent additions to the list of militarily relevant metals, very little is known about the health effects of these metals after internalization as embedded shrapnel. Previous work in this laboratory developed a rodent model system that mimicked shrapnel loads seen in wounded personnel from the 1991 Persian Gulf War. In the present study, we used that system and male F344 rats, implanted intramuscularly with pellets (1 mm times 2 mm cylinders) of weapons-grade WA, to simulate shrapnel wounds. Rats were implanted with 4 (low dose) or 20 pellets (high dose) of WA. Tantalum (20 pellets) and nickel (20 pellets) served as negative and positive controls, respectively. The high-dose WA-implanted rats (n = 46) developed extremely aggressive tumors surrounding the pellets within 4-5 months after implantation. The low-dose WA-implanted rats (n = 46) and nickel-implanted rats (n = 36) also developed tumors surrounding the pellets but at a slower rate. Rats implanted with tantalum (n = 46), an inert control metal, did not develop tumors. Tumor yield was 100% in both the low- and high-dose WA groups. The tumors, characterized as high-grade pleomorphic rhabdomyosarcomas by histopathology and immunohistochemical examination, rapidly metastasized to the lung and necessitated euthanasia of the animal. Significant hematologic changes, indicative of polycythemia, were also observed in the high-dose WA-implanted rats. These changes were apparent as early as 1 month postimplantation in the high-dose WA rats, well before any overt signs of tumor development. These results point out the need for further studies investigating the health effects of tungsten and tungsten-based alloys.


Asunto(s)
Aleaciones/toxicidad , Cuerpos Extraños , Neoplasias de los Músculos/inducido químicamente , Rabdomiosarcoma/inducido químicamente , Compuestos de Tungsteno/toxicidad , Aleaciones/administración & dosificación , Animales , Recuento de Células Sanguíneas , Hematócrito , Hemoglobinas/análisis , Riñón/efectos de los fármacos , Riñón/patología , Neoplasias Pulmonares/secundario , Masculino , Neoplasias de los Músculos/veterinaria , Músculo Esquelético , Tamaño de los Órganos , Ratas , Ratas Endogámicas F344 , Rabdomiosarcoma/veterinaria , Bazo/efectos de los fármacos , Bazo/patología , Compuestos de Tungsteno/administración & dosificación
14.
Mol Cell Biochem ; 255(1-2): 247-56, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14971665

RESUMEN

Depleted uranium (DU) and heavy-metal tungsten alloys (HMTAs) are dense heavy-metals used primarily in military applications. Chemically similar to natural uranium, but depleted of the higher activity 235U and 234U isotopes, DU is a low specific activity, high-density heavy metal. In contrast, the non-radioactive HMTAs are composed of a mixture of tungsten (91-93%), nickel (3-5%), and cobalt (2-4%) particles. The use of DU and HMTAs in military munitions could result in their internalization in humans. Limited data exist however, regarding the long-term health effects of internalized DU and HMTAs in humans. Both DU and HMTAs possess a tumorigenic transforming potential and are genotoxic and mutagenic in vitro. Using insoluble DU-UO2 and a reconstituted mixture of tungsten, nickel, cobalt (rWNiCo), we tested their ability to induce stress genes in thirteen different recombinant cell lines generated from human liver carcinoma cells (HepG2). The commercially available CAT-Tox (L) cellular assay consists of a panel of cell lines stably transfected with reporter genes consisting of a coding sequence for chloramphenicol acetyl transferase (CAT) under transcriptional control by mammalian stress gene regulatory sequences. DU, (5-50 microg/ml) produced a complex profile of activity demonstrating significant dose-dependent induction of the hMTIIA FOS, p53RE, Gadd153, Gadd45, NFkappaBRE, CRE, HSP70, RARE, and GRP78 promoters. The rWNiCo mixture (5-50 microg/ml) showed dose-related induction of the GSTYA, hMTIIA, p53RE, FOS, NFkappaBRE, HSP70, and CRE promoters. An examination of the pure metals, tungsten (W), nickel (Ni), and cobalt (Co), comprising the rWNiCo mixture, demonstrated that each metal exhibited a similar pattern of gene induction, but at a significantly decreased magnitude than that of the rWNiCo mixture. These data showed a synergistic activation of gene expression by the metals in the rWNiCo mixture. Our data show for the first time that DU and rWNiCo can activate gene expression through several signal transduction pathways that may be involved in the toxicity and tumorigenicity of both DU and HMTAs.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Compuestos de Tungsteno/toxicidad , Uranio/toxicidad , Carcinoma Hepatocelular/genética , Cobalto/toxicidad , Chaperón BiP del Retículo Endoplásmico , Perfilación de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Níquel/toxicidad , Análisis de Secuencia por Matrices de Oligonucleótidos , Activación Transcripcional , Células Tumorales Cultivadas
15.
J Environ Radioact ; 64(2-3): 247-59, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12500809

RESUMEN

It is known that radiation can induce a transmissible persistent destabilization of the genome. We have established an in vitro cellular model using HOS cells to investigate whether genomic instability plays a role in depleted uranium (DU)-induced effects. Transmissible genomic instability, manifested in the progeny of cells exposed to ionizing radiation, has been characterized by de novo chromosomal aberrations, gene mutations, and an enhanced death rate. Cell lethality and micronuclei formation were measured at various times after exposure to DU, Ni, or gamma radiation. Following a prompt, concentration-dependent acute response for both endpoints, there was de novo genomic instability in progeny cells. Delayed reproductive death was observed for many generations (36 days, 30 population doublings) following exposure to DU, Ni, or gamma radiation. While DU stimulated delayed production of micronuclei up to 36 days after exposure, levels in cells exposed to gamma-radiation or Ni returned to normal after 12 days. There was also a persistent increase in micronuclei in all clones isolated from cells that had been exposed to nontoxic concentrations of DU. While clones isolated from gamma-irradiated cells (at doses equitoxic to metal exposure) generally demonstrated an increase in micronuclei, most clonal progeny of Ni-exposed cells did not. These studies demonstrate that DU exposure in vitro results in genomic instability manifested as delayed reproductive death and micronuclei formation.


Asunto(s)
Muerte Celular , Daño del ADN , Osteoblastos/patología , Traumatismos por Radiación/fisiopatología , Uranio/efectos adversos , Técnicas de Cultivo de Célula , Células Clonales , Relación Dosis-Respuesta en la Radiación , Humanos , Metales Pesados/efectos adversos , Pruebas de Micronúcleos , Osteosarcoma/patología , Células Tumorales Cultivadas
16.
J Inorg Biochem ; 91(1): 246-52, 2002 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12121782

RESUMEN

Depleted uranium (DU) is a dense heavy metal used primarily in military applications. Published data from our laboratory have demonstrated that DU exposure in vitro to immortalized human osteoblast cells (HOS) is both neoplastically transforming and genotoxic. DU possesses both a radiological (alpha particle) and a chemical (metal) component. Since DU has a low-specific activity in comparison to natural uranium, it is not considered to be a significant radiological hazard. In the current study we demonstrate that DU can generate oxidative DNA damage and can also catalyze reactions that induce hydroxyl radicals in the absence of significant alpha particle decay. Experiments were conducted under conditions in which chemical generation of hydroxyl radicals was calculated to exceed the radiolytic generation by one million-fold. The data showed that markers of oxidative DNA base damage, thymine glycol and 8-deoxyguanosine could be induced from DU-catalyzed reactions of hydrogen peroxide and ascorbate similarly to those occurring in the presence of iron catalysts. DU was 6-fold more efficient than iron at catalyzing the oxidation of ascorbate at pH 7. These data not only demonstrate that DU at pH 7 can induced oxidative DNA damage in the absence of significant alpha particle decay, but also suggest that DU can induce carcinogenic lesions, e.g. oxidative DNA lesions, through interaction with a cellular oxygen species.


Asunto(s)
Partículas alfa , ADN/química , ADN/efectos de la radiación , Uranio/química , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Ácido Ascórbico/química , Catalasa/metabolismo , Bovinos , ADN/metabolismo , Daño del ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/química , Desoxiguanosina/metabolismo , Depuradores de Radicales Libres/metabolismo , Humanos , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/metabolismo , Hierro/química , Níquel/química , Oxidantes/química , Oxidantes/metabolismo , Oxidación-Reducción , Traumatismos por Radiación , Superóxido Dismutasa/metabolismo , Timina/análogos & derivados , Timina/química , Timina/metabolismo
17.
Mil Med ; 167(2 Suppl): 117-9, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11873491

RESUMEN

The health effects of embedded fragments of depleted uranium (DU) are being investigated to determine whether current surgical fragment-removal policies are appropriate for this metal. The authors studied rodents implanted with DU pellets as well as cultured human cells exposed to DU compounds. Results indicate that uranium from implanted DU fragments distributes to tissues distant from implantation sites, including bone, kidney, muscle, and liver. Despite levels of uranium in kidney that would be nephrotoxic after acute exposure, no histological or functional kidney toxicity was observed with embedded DU, indicating that the kidney adapts when exposed chronically. Nonetheless, further studies of the long-term health impact are needed. DU is mutagenic and transforms human osteoblastic cells into a tumorigenic phenotype. It alters neurophysiological parameters in rat hippocampus, crosses the placental barrier, and enters fetal tissue. Preliminary data also indicate decreased rodent litter size when animals are bred 6 months or longer after DU implantation.


Asunto(s)
Uranio , Animales , Humanos , Personal Militar , Ratas
18.
Mil Med ; 167(2 Suppl): 120-2, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11873492

RESUMEN

Limited data exist to permit an accurate assessment of risks for carcinogenesis and mutagenesis from embedded fragments or inhaled particulates of depleted uranium (DU). Ongoing studies have been designed to provide information about the carcinogenic potential of DU using in vitro and in vivo assessments of morphological transformation as well as cytogenetic, mutagenic, and oncogenic effects. For comparison, we also examined tungsten alloys used in military projectiles and the known carcinogen nickel. Quantitative and qualitative in vitro transformation studies were done to assess the carcinogenic potential of radiation and chemical hazards. Using a human osteosarcoma cell model, we demonstrated that soluble and insoluble DU compounds can transform cells to the tumorigenic phenotype, as characterized by morphological, biochemical, and oncogenic changes consistent with tumor cell behavior. Tungsten alloys and nickel were also shown to be neoplastic transforming agents, although at a frequency less than that of DU. Sister chromatid exchange, micronuclei, and alkaline filter elution assays showed DU and tungsten alloys were genotoxic. Exposure to a nontoxic, nontransforming dose of DU induced a small but statistically significant increase in the number of dicentrics formed in cells. These results suggest that long-term exposure to DU or tungsten alloys could be critical to the development of neoplastic disease in humans and that additional studies are needed.


Asunto(s)
Carcinógenos , Níquel , Tungsteno , Uranio , Heridas por Arma de Fuego , Humanos , Neoplasias Inducidas por Radiación
19.
Mil Med ; 167(2 Suppl): 16-9, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11873502

RESUMEN

A reliable, relatively easy method for diagnostic assessment of radiation exposure is needed to support the triage of radiation casualties and medical treatment decisions in military defense operations. Our strategy is to identify radiation-responsive DNA mutations and gene expression targets that can be analyzed using polymerase chain reaction (PCR) assays and an existing fluorescence-based nucleic acid analysis system designed for forward-deployable laboratory applications. Using an in vitro model system of human peripheral blood lymphocytes, we identified a candidate nucleic acid biomarker (i.e., gene expression target) that is responsive to ionizing radiation. In this report, we describe our preliminary Haras gene expression findings. A dose-dependent elevation in Haras gene expression levels was demonstrated using Northern-blot analysis 17 hours after exposure to a 250-kVp dose of X-rays (25-100 cGy, 1 Gy/minute); c-Haras expression levels at 100 cGy were ninefold higher than those of controls. An alternative protocol to quantify the Haras cDNA target, using the rapid, real-time reverse transcriptase fluorogenic 5'-nuclease PCR assay, is described, along with a preliminary characterization of the dynamic range for detection. Our research shows that the analysis of multitarget nucleic acid biomarkers, using the multiplex fluorogenic 5'-nuclease PCR assay, has beneficial applications in radiation epidemiology, radiation therapy, and biodosimetry.


Asunto(s)
Reacción en Cadena de la Polimerasa , Dosis de Radiación , Humanos , Linfocitos/efectos de la radiación
20.
Mil Med ; 167(2 Suppl): 54-6, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11873517

RESUMEN

Carcinogenesis is a multistage process involving dysregulation of signal transduction and cell cycle pathways. This dysregulation results in specific molecular and genetic alterations, including gene amplification, mutations, and chromosomal rearrangements. These aberrations can be measured to provide a novel means to assess carcinogenic risk or as targets for chemointervention. Recent human and in vivo studies have demonstrated that genetic alterations, such as oncogenes and oncoproteins, were observed in preneoplastic tissues or serum following exposure to chemical carcinogens or low-level radiation (LLR). Identification of preneoplastic changes following radiation exposure may provide information that will allow development of LLR chemopreventive strategies. Radiation carcinogenesis studies in vivo with a lung tumor model showed that a low-dose cobalt-60 radiation exposure induced persistent time-dependent genetic alterations, such as elevated ras expression. This radiation exposure also resulted in lung tumor formation in 26% of the irradiated animals at 232 days after irradiation. A significant and progressive increase in ras oncogene expression was measured using Northern blot analysis in 80% of the irradiated animals over the duration of the experiment. Pharmacological intervention strategies are being tested using buthionine-[S,R]-sulfoximine (BSO). BSO has been previously shown to down-regulate ras expression. Administration of BSO prevented radiation-induced changes in ras mRNA levels in this lung tumor model. Further studies are being conducted with an LLR-induced leukemia model in which detection of circulating levels of oncoproteins will be more feasible. Based on these preliminary results and on its clinical efficacy and low clinical toxicity, BSO warrants further study as an LLR chemopreventive agent. Furthermore, this strategy to target LLR-induced preneoplastic alterations may be an effective means of developing modulators of LLR-induced cancers.


Asunto(s)
Neoplasias Inducidas por Radiación/prevención & control , Animales , Radioisótopos de Cobalto , Ratones , Oncogenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA