Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proteomes ; 3(3): 249-265, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-28248270

RESUMEN

Damage to normal lung tissue is a limiting factor when ionizing radiation is used in clinical applications. In addition, radiation pneumonitis and fibrosis are a major cause of mortality following accidental radiation exposure in humans. Although clinical symptoms may not develop for months after radiation exposure, immediate events induced by radiation are believed to generate molecular and cellular cascades that proceed during a clinical latent period. Oxidative damage to DNA is considered a primary cause of radiation injury to cells. DNA can be repaired by highly efficient mechanisms while repair of oxidized proteins is limited. Oxidized proteins are often destined for degradation. We examined protein oxidation following 17 Gy (0.6 Gy/min) thoracic X-irradiation in C57BL/6J mice. Seventeen Gy thoracic irradiation resulted in 100% mortality of mice within 127-189 days postirradiation. Necropsy findings indicated that pneumonitis and pulmonary fibrosis were the leading cause of mortality. We investigated the oxidation of lung proteins at 24 h postirradiation following 17 Gy thoracic irradiation using 2-D gel electrophoresis and OxyBlot for the detection of protein carbonylation. Seven carbonylated proteins were identified using mass spectrometry: serum albumin, selenium binding protein-1, alpha antitrypsin, cytoplasmic actin-1, carbonic anhydrase-2, peroxiredoxin-6, and apolipoprotein A1. The carbonylation status of carbonic anhydrase-2, selenium binding protein, and peroxiredoxin-6 was higher in control lung tissue. Apolipoprotein A1 and serum albumin carbonylation were increased following X-irradiation, as confirmed by OxyBlot immunoprecipitation and Western blotting. Our findings indicate that the profile of specific protein oxidation in the lung is altered following radiation exposure.

2.
Int J Radiat Biol ; 90(12): 1169-78, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24899392

RESUMEN

PURPOSE: Protein oxidation in response to radiation results in DNA damage, endoplasmic reticulum stress/unfolded protein response, cell cycle arrest, cell death and senescence. The liver, a relatively radiosensitive organ, undergoes measurable alterations in metabolic functions following irradiation. Accordingly, we investigated radiation-induced changes in liver metabolism and alterations in protein oxidation. MATERIALS AND METHODS: C57BL/6 mice were sham irradiated or exposed to 8.5 Gy (60)Co (0.6 Gy/min) total body irradiation. Metabolites and metabolic enzymes in the blood and liver tissue were analyzed. Two-dimensional gel electrophoresis and OxyBlot™ were used to detect carbonylated proteins that were then identified by peptide mass fingerprinting. RESULTS: Analysis of serum metabolites revealed elevated glucose, bilirubin, lactate dehydrogenase (LDH), high-density lipoprotein, and aspartate aminotransferase within 24-72 h post irradiation. Liver tissue LDH and alkaline phosphatase activities were elevated 24-72 h post irradiation. OxyBlotting revealed that the hepatic proteome contains baseline protein carbonylation. Radiation exposure increased carbonylation of specific liver proteins including carbonic anhydrase 1, α-enolase, and regucalcin. CONCLUSIONS: 8.5 Gy irradiation resulted in distinct metabolic alterations in hepatic functions. Coincident with these changes, radiation induced the carbonylation of specific liver enzymes. The oxidation of liver enzymes may underlie some radiation-induced alterations in hepatic function.


Asunto(s)
Hígado/metabolismo , Hígado/efectos de la radiación , Proteínas/metabolismo , Animales , Biomarcadores/metabolismo , Femenino , Hígado/enzimología , Hígado/fisiología , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de la radiación , Carbonilación Proteica/efectos de la radiación
3.
Proteomes ; 2(3): 291-302, 2014 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-28250382

RESUMEN

The bone marrow is one of the most radio-sensitive tissues. Accidental ionizing radiation exposure can damage mature blood cells and hematopoietic progenitor/stem cells, and mortality can result from hematopoietic insufficiency and infection. Ionizing radiation induces alterations in gene and protein expression in hematopoietic tissue. Here we investigated radiation effects on protein carbonylation, a primary marker for protein oxidative damage. C57BL/6 mice were either sham irradiated or exposed to 7.5 Gy 60Co (0.6 Gy/min) total body irradiation. Bone marrow was obtained 24 h post-irradiation. Two dimensional (2-D) gel electrophoresis and Oxyblot immunodetection were used to discover carbonylated proteins, and peptide mass fingerprinting was performed for identification. 2D gels allowed the detection of 13 carbonylated proteins in the bone marrow; seven of these were identified, with two pairs of the same protein. Baseline levels of carbonylation were found in 78 kDa glucose-related protein, heat shock protein cognate 71 KDa, actin, chitinase-like protein 3 (CHI3L1), and carbonic anhydrase 2 (CAII). Radiation increased carbonylation in four proteins, including CHI3L1 and CAII, and induced carbonylation of one additional protein (not identified). Our findings indicate that the profile of specific protein carbonylation in bone marrow is substantially altered by ionizing radiation. Accordingly, protein oxidation may be a mechanism for reduced cell viability.

4.
Exp Hematol ; 39(3): 293-304, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21146580

RESUMEN

OBJECTIVE: Our laboratory reported that the angiotensin converting enzyme inhibitor captopril improves erythroid recovery from total body irradiation (TBI) in mice when administered after irradiation. However, captopril administered before TBI attenuates erythroid recovery. Here we investigate captopril and radiation regulation of erythropoietin (EPO) and thrombopoietin (TPO), key effectors of erythroid progenitor proliferation and differentiation. MATERIALS AND METHODS: C57BL/6 mice, nonirradiated or exposed to 7.5 Gy TBI ((60)Co, 0.6 Gy/min) were untreated or administered captopril. Plasma EPO and TPO levels were measured by enzyme-linked immunosorbent assay. Gene expression of EPO was determined by quantitative reverse transcription polymerase chain reaction. The hypoxia-inducible factors (HIF)-1α and -2α were measured by immunoblotting. RESULTS: In nonirradiated mice, continuous captopril administration in the water transiently reduced reticulocytes and red blood cells after 7 and 10 days, respectively. EPO plasma levels and gene expression were reduced below detectable limits after 2 days of captopril treatment, but recovered within 7 days. HIF-1α and HIF-2α were activated preceding reticulocyte and red blood cell recovery. TBI, which ablates early and late-stage erythroid progenitors, activated both HIFs and increased EPO and TPO. Captopril treatment postirradiation suppressed radiation-induced HIF activation and EPO expression. In contrast, captopril administration for 7 days before TBI resulted in earlier EPO induction and activation. Captopril treatment lowered TPO levels in nonirradiated mice, but had minimal effects on radiation-induced TPO. CONCLUSIONS: In nonirradiated mice, captopril biphasically regulates EPO via HIF activation. TBI ablates erythroid progenitors, resulting in hypoxia, HIF activation, and increased EPO expression that are modulated by captopril treatment. These data suggest that short-term suppression of radiation-induced EPO immediately after TBI is favorable for erythroid recovery.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Captopril/farmacología , Células Precursoras Eritroides/metabolismo , Eritropoyetina/biosíntesis , Rayos gamma/efectos adversos , Regulación de la Expresión Génica , Irradiación Corporal Total , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Ratones , Trombopoyetina/biosíntesis , Factores de Tiempo
5.
Exp Hematol ; 38(4): 270-81, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20116413

RESUMEN

OBJECTIVE: Angiotensin II (Ang II), a potent vasoconstrictor, affects the growth and development of hematopoietic cells. Mixed findings have been reported for the effects of angiotensin-converting enzyme (ACE) inhibitors on radiation-induced injury to the hematopoietic system. We investigated the consequences of different regimens of the ACE inhibitor captopril on radiation-induced hematopoietic injury. MATERIALS AND METHODS: C57BL/6 mice were either sham-irradiated or exposed to (60)Co total body irradiation (0.6 Gy/min). Captopril was provided in the water for different time periods relative to irradiation. RESULTS: In untreated mice, the survival rate from 7.5 Gy was 50% at 30 days postirradiation. Captopril treatment for 7 days prior to irradiation resulted in radiosensitization with 100% lethality and a rapid decline in mature blood cells. In contrast, captopril treatment beginning 1 hour postirradiation and continuing for 30 days resulted in 100% survival, with improved recovery of mature blood cells and multilineage hematopoietic progenitors. In nonirradiated control mice, captopril biphasically modulated Lin(-) marrow progenitor cell cycling. After 2 days, captopril suppressed G(0)-G(1) transition and a greater number of cells entered a quiescent state. However, after 7 days of captopril treatment Lin(-) progenitor cell cycling increased compared to untreated control mice. CONCLUSION: These findings suggest that ACE inhibition affects hematopoietic recovery following radiation by modulating the hematopoietic progenitor cell cycle. The timing of captopril treatment relative to radiation exposure differentially affects the viability and repopulation capacity of spared hematopoietic stem cells and, therefore, can result in either radiation protection or radiation sensitization.


Asunto(s)
Captopril/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Protección Radiológica/métodos , Irradiación Corporal Total , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Captopril/administración & dosificación , Femenino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Factores de Tiempo
6.
J Radiat Res ; 49(4): 361-72, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18434686

RESUMEN

The effects of genistein on 30-day survival and delayed lung injury were examined in C57BL/6J female mice. A single subcutaneous injection of vehicle (PEG-400) or genistein (200 mg/kg) was administered 24 h before total body irradiation (7.75 Gy (60)Co, 0.6 Gy/min). Experimental groups were: No treatment + Sham (NC), Vehicle + Sham (VC), Genistein + Sham (GC), Radiation only (NR), Vehicle + Radiation (VR), Genistein + Radiation (GR). Thirty-day survivals after 7.75 Gy were: NR 23%, VR 53%, and GR 92%, indicating significant protection from acute radiation injury by genistein. Genistein also mitigated radiation-induced weight loss on days 13-28 postirradiation. First generation lung fibroblasts were analyzed for micronuclei 24 h postirradiation. Fibroblasts from the lungs of GR-treated mice had significantly reduced micronuclei compared with NR mice. Collagen deposition was examined by histochemical staining. At 90 days postirradiation one half of the untreated and vehicle irradiated mice had focal distributions of small collagen-rich plaques in the lungs, whereas all of the genistein-treated animals had morphologically normal lungs. Radiation reduced the expression of COX-2, transforming growth factor-beta receptor (TGFbetaR) I and II at 90 days after irradiation. Genistein prevented the reduction in TGFbetaRI. However, by 180 days postirradiation, these proteins normalized in all groups. These results demonstrate that genistein protects against acute radiation-induced mortality in female mice and that GR-treated mice have reduced lung damage compared to NR or VR. These data suggest that genistein is protective against a range of radiation injuries.


Asunto(s)
Citocinas/análisis , Genisteína/administración & dosificación , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/prevención & control , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/prevención & control , Irradiación Corporal Total , Animales , Biomarcadores/metabolismo , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Femenino , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Dosis de Radiación , Protectores contra Radiación/administración & dosificación , Análisis de Supervivencia , Tasa de Supervivencia
7.
Front Biosci ; 11: 2093-105, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16720296

RESUMEN

Dysregulation of Wnt signaling is common in a variety of human malignancies. Activation of the canonical Wnt or beta-catenin pathway has been especially well documented in cancer, although other non-canonical Wnt signaling pathways also have been implicated in neoplasia. In most instances, constitutive signaling through the beta-catenin pathway involves activation of effector molecules or loss of tumor suppressor function downstream of Wnt binding to its cell surface receptors. Nonetheless, in recent years increasing evidence suggests that secreted Wnt antagonists act as tumor suppressors, with their expression often silenced by promoter hypermethylation. This implies that maximal constitutive signaling in cancer requires unimpaired Wnt stimulation at the cell surface as well as enhanced signal propagation within the cell. However, an understanding of the role secreted Wnt antagonists may play in cancer is complicated by the multiplicity of these proteins, their potential Wnt-independent activities and observations indicating that sometimes they may promote tumor growth. Just as the particular function of Wnt signaling in development and homeostasis varies with the setting, the impact of secreted Wnt antagonists on neoplasia depends on the molecular, cellular and tissue context.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/fisiopatología , Transducción de Señal , Proteínas Wnt/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteínas de la Membrana/fisiología , Neoplasias/prevención & control , Proteínas Represoras/fisiología
8.
Circ Res ; 98(10): 1331-9, 2006 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-16601226

RESUMEN

We have used global gene expression analysis to establish a comprehensive list of candidate genes in the developing vasculature during embryonic (ES) cell differentiation in vitro. A large set of genes, including growth factors, cell surface molecules, transcriptional factors, and members of several signal transduction pathways that are known to be involved in vasculogenesis or angiogenesis, were found to have expression patterns as expected. Some unknown or functionally uncharacterized genes were differentially regulated in flk1+ cells compared with flk1- cells, suggesting possible roles for these genes in vascular commitment. Particularly, multiple components of the Wnt signaling pathway were differentially regulated in flk1+ cells, including Wnt proteins, their receptors, downstream transcriptional factors, and other components belonging to this pathway. Activation of the Wnt signal was able to expand vascular progenitor populations whereas suppression of Wnt activity reduced flk1+ populations. Suppression of Wnt signaling also inhibited the formation of matured vascular capillary-like structures during late stages of embryoid body differentiation. These data indicate a requisite and ongoing role for Wnt activity during vascular development, and the gene expression profiles identify candidate components of this pathway that participate in vascular cell differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Embrión de Mamíferos/citología , Endotelio Vascular/citología , Perfilación de la Expresión Génica , Transducción de Señal/fisiología , Células Madre/citología , Proteínas Wnt/metabolismo , Animales , Linaje de la Célula , Células Cultivadas , Ratones , Células Madre/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
J Biol Chem ; 280(1): 777-86, 2005 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-15509575

RESUMEN

Wnts stimulate cell migration, although the mechanisms responsible for this effect are not fully understood. To investigate the pathways that mediate Wnt-dependent cell motility, we treated Chinese hamster ovary cells with Wnt-3a-conditioned medium and monitored changes in cell shape and movement. Wnt-3a induced cell spreading, formation of protrusive structures, reorganization of stress fibers and migration. Although Wnt-3a stabilized beta-catenin, two inhibitors of the beta-catenin/canonical pathway, Dickkopf-1 and a dominant-negative T cell factor construct, did not reduce motility. The small GTPase RhoA also was activated by Wnt-3a. In contrast to beta-catenin signaling, inhibition of Rho kinase partially blocked motility. Because Dishevelled (Dvl) proteins are effectors of both canonical and noncanonical Wnt signaling, we used immunofluorescent analysis and small interference RNA technology to evaluate the role of Dvl in cell motility. Specific knock-down of Dvl-2 expression markedly reduced Wnt-3a-dependent changes in cell shape and movement, suggesting that this Dvl isoform had a predominant role in mediating Wnt-3a-dependent motility in Chinese hamster ovary cells.


Asunto(s)
Forma de la Célula/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Proteínas/farmacología , Proteína de Unión al GTP rhoA/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células CHO , Cricetinae , Cricetulus , Medios de Cultivo Condicionados/farmacología , Proteínas Dishevelled , Activación Enzimática/efectos de los fármacos , Ratones , Fosfoproteínas , Proteínas/metabolismo , Transducción de Señal , Proteínas Wnt , Proteína Wnt3 , Proteína Wnt3A
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...