Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 31(6): 739-51, 2012 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21725368

RESUMEN

Glioblastomas are among the most incurable cancers. Our past findings indicated that glioblastoma cells, but not neurons or glia, require the transcription factor ATF5 (activating transcription factor 5) for survival. However, it was unknown whether interference with ATF5 function can prevent or promote regression/eradication of malignant gliomas in vivo. To address this issue, we created a mouse model by crossing a human glial fibrillary acidic protein (GFAP) promoter-tetracycline transactivator mouse line with tetracycline operon-dominant negative-ATF5 (d/n-ATF5) mice to establish bi-transgenic mice. In this model, d/n-ATF5 expression is controlled by doxycycline and the promoter for GFAP, a marker for stem/progenitor cells as well as gliomas. Endogenous gliomas were produced with high efficiency by retroviral delivery of platelet-derived growth factor (PDGF)-B and p53-short hairpin RNA (shRNA) in adult bi-transgenic mice in which expression of d/n-ATF5 was spatially and temporally regulated. Induction of d/n-ATF5 before delivery of PDGF-B/p53-shRNA virus greatly reduced the proportion of mice that formed tumors. Moreover, d/n-ATF5 induction after tumor formation led to regression/eradication of detectable gliomas without evident damage to normal brain cells in all 24 mice assessed.


Asunto(s)
Factores de Transcripción Activadores/metabolismo , Neoplasias Encefálicas/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Glioma/metabolismo , Factores de Transcripción Activadores/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Doxiciclina/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Glioma/genética , Glioma/patología , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Toxicol In Vitro ; 25(8): 1895-905, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21703343

RESUMEN

Environmental particulate matter (PM) exposure has been correlated with pathogenesis of acute airway inflammatory disease such as asthma and COPD. PM size and concentration have been studied extensively, but the additional effects of particulate components such as biological material, transition metals, and polycyclic aromatic hydrocarbons could also impact initial disease pathogenesis. In this study, we compared urban ambient particulate matter (APM) collected from Fresno, California with wildfire (WF) particulate matter collected from Escalon, California on early transcriptional responses in human bronchial epithelial cells (HBE). Global gene expression profiling of APM treated HBE activated genes related to xenobiotic metabolism (CYP 1B1), endogenous ROS generation and response genes (DUOX1, SOD2, PTGS2) and pro-inflammatory responses associated with asthma or COPD such as IL-1α, IL-1ß, IL-8, and CCL20. WF PM treatments also induced a pro-inflammatory gene response, but elicited a more robust xenobiotic metabolism and oxidative stress response. Inhibitor studies targeting endotoxin, ROS, and trace metals, found endotoxin inhibition had modest selective inhibition of inflammation while inhibition of hydrogen peroxide and transition metals had broad effects suggesting additional interactions with xenobiotic metabolism pathways. APM induced a greater inflammatory response while WF PM had more marked metabolism and ROS related responses.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Material Particulado/toxicidad , Adulto , Bronquios/citología , California , Células Cultivadas , Ciudades , Citocinas/genética , Endotoxinas/toxicidad , Células Epiteliales/metabolismo , Femenino , Incendios , Perfilación de la Expresión Génica , Humanos , Inflamación/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Xenobióticos/toxicidad
3.
Toxicol In Vitro ; 24(7): 1993-2002, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20801209

RESUMEN

In urban areas, a correlation between exposure to particulate matter (PM) from air pollution and increased cardiovascular morbidity and mortality has been observed. Components of PM include bacterial contaminants, transition metals, salts, polycyclic aromatic hydrocarbons (PAH), and carbonaceous material, which could interact with various cell types to produce systemic responses when inhaled. We examined the effects of PM collected from Fresno, California on activation of human monocytes and their interaction with vascular endothelium, a key event in atherogenesis. PM exposure increased cytokine expression and secretion from monocytes and enhanced monocyte adhesion to human aortic endothelial cells, both of which were attenuated by neutralizing endotoxin. PM also increased monocyte CYP1a1 expression, and inhibition of the aryl hydrocarbon receptor reduced the CYP1a1 and inflammatory responses. PM-treated monocytes accumulated intracellular reactive oxygen species (ROS), and antioxidants attenuated inflammatory and xenobiotic responses. Finally, supernatants from PM-treated pulmonary microvascular endothelial cells induced monocyte inflammatory responses that were not a consequence of endotoxin transfer. These results suggest that certain components of urban PM, namely endotoxin and PAH, activate circulating monocytes directly or indirectly by first stimulating other cells such as pulmonary endothelial cells, providing several mechanisms by which PM inhalation could induce pulmonary and/or systemic inflammation.


Asunto(s)
Contaminantes Atmosféricos/efectos adversos , Endotoxinas/toxicidad , Monocitos/efectos de los fármacos , Hidrocarburos Policíclicos Aromáticos/toxicidad , Contaminantes Atmosféricos/química , Contaminación del Aire/efectos adversos , Aorta/citología , Aorta/efectos de los fármacos , California , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Células Cultivadas , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotoxinas/aislamiento & purificación , Humanos , Monocitos/metabolismo , Material Particulado/química , Material Particulado/toxicidad , Hidrocarburos Policíclicos Aromáticos/aislamiento & purificación , Especies Reactivas de Oxígeno/metabolismo
4.
Inhal Toxicol ; 22(4): 267-76, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20064101

RESUMEN

Increasingly, evidence suggests a role for a systemic procoagulant state in the pathogenesis of cardiac dysfunction subsequent to inhalation of airborne particulate matter. The authors evaluated blood cell parameters and markers of platelet activation in mice exposed to concentrated ambient particulate matter (CAPs) from the San Joaquin Valley of California, a region with severe particulate matter (PM) pollution episodes. The authors exposed mice to an average of 88.5 microg/m(3) of CAPs in a size range less than 2.5 microm for 6 h/day for 5 days per week for 2 weeks. Platelets were analyzed by flow cytometry for relative size, shape, aggregation, fibrinogen binding, P-selectin, and lysosomal-associated membrane protein-1 (LAMP-1) expression. Serum cytokines were analyzed by bead-based immunologic assays. CAPs-exposed mice had elevations in macrophage inflammatory protein (MIP)-1 alpha, MIP-1 beta, interleukin (IL)-6, IL-10, tumor necrosis factor alpha (TNFalpha), macrophage colony-stimulating factor (M-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), platelet-derived growth factor (PDGF)-bb, and RANTES (regulated upon activation, normally T-expressed, and presumably secreted). Platelets were the only peripheral blood cells that were significantly elevated in number in CAPs-exposed mice. Flow cytometric analysis of unstimulated platelets from CAPs-exposed mice indicated size and shape changes, and platelets from CAPs-exposed animals had a 54% increase in fibrinogen binding indicative of platelet priming. Stimulation of platelets by thrombin resulted in up-regulation of LAMP-1 expression in CAPs-exposed animals and an increased microparticle population relative to control animals. These findings demonstrate a systemic proinflammatory and procoagulant response to inhalation of environmentally derived fine and ultrafine PM and suggests a role for platelet activation in the cardiovascular and respiratory effects of particulate air pollution.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Citocinas/metabolismo , Material Particulado/toxicidad , Activación Plaquetaria/efectos de los fármacos , Contaminantes Atmosféricos/análisis , Animales , Recuento de Células Sanguíneas , California , Exposición a Riesgos Ambientales , Monitoreo del Ambiente , Fibrinógeno/metabolismo , Citometría de Flujo , Mediadores de Inflamación/metabolismo , Exposición por Inhalación , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de la Partícula , Material Particulado/análisis , Hidrocarburos Policíclicos Aromáticos/análisis
5.
Vascul Pharmacol ; 46(6): 439-48, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17336165

RESUMEN

The mechanistic relationship between the widely used monocrotaline model of primary pulmonary hypertension and altered TGFbeta family signaling due to genetic defects in the Bone Morphogenetic Protein type II receptor in affected humans has not been investigated. In this study we use fluorescent microscopy to demonstrate nuclear translocation of Smad 4 in human pulmonary arterial endothelial cell (HPAEC) cultures treated with monocrotaline pyrrole (MCTP), Bone Morphogenetic Protein (BMP) and TGFbeta. While MCTP induced transient nuclear accumulation of phosphorylated Smad 1 (P-Smad 1) and phosphorylated Smad 2 (P-Smad 2), only expression of P-Smad 1 was significantly altered in western blots. P-Smad 1 expression significantly increased 30 min following treatment with MCTP correlating with P-Smad 1 and Smad 4 nuclear translocation. Although a modest, but significant decrease in P-Smad 1 expression occurred 1 h after treatment, expression was significantly increased at 72 h. Evaluation of components of the signal and response pathway at 72 h showed decreased expression of the BMP type II receptor (BMPrII), no change in TGFbeta Activin Receptor-like Kinase 1 (Alk 1), no change in Smad 4 but increase in the inhibitory Smad 6, decrease in the alternate BMP signaling pathway p38(MAPK) but no change in the psmad1 response element ID 1. Our results suggest transient activation of Smad signaling pathways in initial MCTP endothelial cell toxicity, and a persistent dysregulation of BMP signaling. Electron microscopy of cell membrane caveoli revealed a dramatic decrease in these structures after 72 h. Loss of these structural elements, noted for their sequestration and inhibition of receptor activity, may contribute to prolonged alterations in BMP signaling.


Asunto(s)
Alquilantes/toxicidad , Núcleo Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Monocrotalina/análogos & derivados , Arteria Pulmonar/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Caveolas/efectos de los fármacos , Caveolas/ultraestructura , Núcleo Celular/metabolismo , Células Cultivadas , Preescolar , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Femenino , Humanos , Monocrotalina/toxicidad , Fosforilación , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Proteínas Smad/genética , Proteína Smad1/metabolismo , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , Proteína smad6/metabolismo , Factores de Tiempo , Transfección , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Vascul Pharmacol ; 44(1): 50-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16271518

RESUMEN

Polymorphic mutations in the Bone Morphogenetic Protein type II receptor (BMPrII) gene have been implicated in the development of familial primary pulmonary hypertension (PPH) however, the role BMPrII mutations play in the development of PH has not yet been elucidated. Endothelial caveolae are an important domain of hemodynamics containing eNOS, the serotonin transporter, and endothelin receptors. In this study we show by standard immunohistochemistry (IHC) that BMPrII is widely distributed in the vasculature of the rat lung, and more specifically distributed to both apical and basal membranes of the arteriolar endothelium by fluorescent IHC. We also examined compartmentalization of BMPrII in lipid fractions of plasma membranes isolated by silica based extraction from human pulmonary artery endothelial cells and rat lung endothelium. Density gradient centrifugation demonstrated BMPrII in separate caveolin-1 (cav-1) and non-cav-1 lipid rich fractions. Electron microscopy co-localized cav-1 and BMPrII in flask shaped membrane fragments. Three-dimensional fluorescence microscopy demonstrated BMPrII in discrete membrane foci, a portion of which were co-localized with cav-1, as well as in Golgi. Our findings indicate that BMPrII is located within lipid-dense fractions of pulmonary endothelial cell membranes with a portion present in caveolae suggesting potential dynamic regulatory structural relationships.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/análisis , Caveolas/metabolismo , Endotelio Vascular/metabolismo , Pulmón/irrigación sanguínea , Adulto , Animales , Caveolina 1/análisis , Línea Celular , Preescolar , Endotelio Vascular/ultraestructura , Femenino , Aparato de Golgi/metabolismo , Humanos , Pulmón/ultraestructura , Masculino , Microdominios de Membrana/metabolismo , Ratas , Ratas Sprague-Dawley
7.
Aquat Toxicol ; 73(3): 221-9, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15935862

RESUMEN

Four cytosolic glutathione S-transferase (GST) classes were isolated and characterized from juvenile winter run Chinook salmon (Oncorhynchus tshawytscha) liver. Two techniques were used: (1) gel electrophoresis/immunoblotting against a polyclonal striped bass GST antibody and (2) high-pressure liquid chromatography (HPLC). Nanospray liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to elucidate peptide sequences and the proteins were identified as pi, theta, mu and alpha, by searching against the NCBI non-redundant database (nrDB). Catalytic activity of the cytosolic GSTs towards 1-chloro-2,4-dinitrobenzene (CDNB) and ethacrynic acid (ETHA) were determined to be 0.3+/-0.05 U/mg cytosolic protein and 0.06+/-0.02 U/mg cytosolic protein, respectively.


Asunto(s)
Citosol/enzimología , Glutatión Transferasa/aislamiento & purificación , Salmón/metabolismo , Secuencia de Aminoácidos , Animales , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Biología Computacional , Dinitroclorobenceno/metabolismo , Ácido Etacrínico/metabolismo , Glutatión Transferasa/genética , Immunoblotting , Espectrometría de Masas , Datos de Secuencia Molecular , Alineación de Secuencia
8.
Aquat Toxicol ; 71(3): 203-14, 2005 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-15670627

RESUMEN

Glutathione S-transferases (GSTs) are a family of detoxification enzymes that catalyze the conjugation of glutathione (GSH) to electrophiles, thus preventing toxicity. This study characterized the cytosolic GST classes of juvenile white sturgeon (Acipenser transmontanus) liver, using two methods of isolation. The first, which employed affinity chromatography, electrophoresis and immunoblotting against a polyclonal striped bass GST antibody, yielded two cytosolic GSTs. The GSTs were identified by nanospray liquid chromatography-tandem mass spectrometry (LC-MS/MS), peptide mass mapping and MS/MS sequencing, as well as de novo MS/MS sequencing as GST classes pi and mu using the Mascot search engine and the NCBI non-redundant database (nrDB) for both methods. The molecular masses were determined to be 23,548 +/- 23 and 26,027 +/- 23 Da, respectively, using linear matrix assisted laser desorption ionization time of flight (MALDI-TOF) mass spectrometry. The second method of isolation, which used affinity chromatography and high-pressure liquid chromatography (HPLC), yielded pi, mu, and possibly two alpha isoforms by MALDI-TOF-TOF, again searching against the NCBI nrDB. The alpha isoforms were determined to have molecular masses of 25,528 +/- 23 and 25,348 +/- 23 Da by electrospray ionization source (ESI)-MS. Overall, it appears that the HPLC method is more sensitive than immunoblotting with the current antibody. Activity of the cytosolic GSTs was evaluated using the substrate 1-chloro-2,4-dinitrobenzene (CDNB) and found to be 2.4 +/- 0.6 U/mg cytosolic protein, and 0.41 +/- 0.05 U/mg cytosolic protein using ethacrynic acid (ETHA).


Asunto(s)
Citosol/química , Peces/metabolismo , Glutatión Transferasa/química , Secuencia de Aminoácidos , Animales , Cromatografía de Afinidad , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Biología Computacional , Electroforesis , Glutatión Transferasa/genética , Immunoblotting , Espectrometría de Masas , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
9.
J Lipid Res ; 41(11): 1790-800, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11060348

RESUMEN

We present a sensitive tracer method, suitable for in vivo human research, that uses beta-[(14)C]carotene coupled with accelerator mass spectrometry (AMS) detection. Using this approach, the concentration-time course of a physiological (306 microgram 200 nCi) oral dose of beta-[(14)C]carotene was determined for 209 days in plasma. Analytes included beta-[(14)C]carotene, [(14)C]retinyl esters, [(14)C]retinol, and several [(14)C]retinoic acids. There was a 5.5-h lag between dosing and the appearance of (14)C in plasma. Labeled beta-carotene and [(14)C]retinyl esters rose and displayed several maxima with virtually identical kinetic profiles over the first 24-h period; elevated [(14)C]retinyl ester concentrations were sustained in the plasma compartment for >21 h postdosing. The appearance of [(14)C]retinol in plasma was also delayed 5.5 h postdosing and its concentration rose linearly for 28 h before declining. Cumulative urine and stool were collected for 17 and 10 days, respectively, and 57.4% of the dose was recovered in the stool within 48 h postdosing. The stool was the major excretion route for the absorbed dose. The turnover times (1/k(el)) for beta-carotene and retinol were 58 and 302 days, respectively. Area under the curve analysis of the plasma response curves suggested a molar vitamin A value of 0.53 for beta-carotene, with a minimum of 62% of the absorbed beta-carotene being cleaved to vitamin A.In summary, AMS is an excellent tool for defining the in vivo metabolic behavior of beta-carotene and related compounds at physiological concentrations. Further, our data suggest that retinyl esters derived from beta-carotene may undergo hepatic resecretion with VLDL in a process similar to that observed for beta-carotene.


Asunto(s)
beta Caroteno/farmacocinética , Adulto , Disponibilidad Biológica , Dióxido de Carbono , Radioisótopos de Carbono , Heces/química , Humanos , Marcaje Isotópico/métodos , Cinética , Masculino , Fotosíntesis , Spinacia oleracea , Tretinoina/sangre , Vitamina A/sangre , beta Caroteno/sangre , beta Caroteno/orina
10.
Toxicol Appl Pharmacol ; 166(2): 69-80, 2000 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10896848

RESUMEN

Monocrotaline pyrrole (MCTP) causes cyto- and karyomegaly and persistent cell cycle arrest in the G2 stage of the cell cycle in cultured bovine pulmonary artery endothelial cells. To better characterize the cell cycle regulatory mechanisms of this process as well as determine whether this process would occur in cells of human origin, we treated human pulmonary artery endothelial cell (HPAEC) cultures with MCTP and determined, by flow cytometry, the expression of cyclin B1 and p53 in conjunction with DNA content. We also validated by Western blots that the persistence of cdc2 in its inactivated phosphorylated state, previously described in bovine cell cultures, occurred in HPAEC. Alterations in p53, cyclin A, cyclin B1, and cdc25c expression were also examined in Western blots of treated HPAEC extracts. The response of HPAEC to MCTP was compared with that of adriamycin and nocodazole, agents known to cause cell cycle alterations. Results of these experiments demonstrate that HPAEC treated with MCTP develop a population of cells in G2 that has increased cyclin B1 expression. These cells express increased amounts of cdc2 but not cdc25c. The ratio of inactive triphosphorylated cdc2 to the active monophosphorylated form increased moderately from control cultures in contrast to predominance of the active form in nocodazole-treated cultures. In addition, a second population of cells expressing cyclin B1 had continued incorporation of BrdU and DNA content consistent with 8 N chromosomes. A similar 8 N cell population was evident in nocodazole-treated cells but these cells had both cyclin B1 positive and negative components. Compared with adriamycin, a known inducer of p53, MCTP-treated HPAEC expressed p53 only at high concentrations and p53 expression was not coordinated with G2 arrest or polyploidy. We conclude that HPAEC treated with low concentrations of MCTP develop G2 arrest in association with persistent cyclin B1 expression, failure to completely activate cdc2, and continued DNA synthesis through a pathway that is unrelated to altered expression of p53.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Monocrotalina/análogos & derivados , Arteria Pulmonar/efectos de los fármacos , Adulto , Animales , Western Blotting , Bovinos , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Niño , Ciclina A/metabolismo , Ciclina B/metabolismo , Ciclina B1 , ADN/análisis , Doxorrubicina/farmacología , Endotelio Vascular/metabolismo , Femenino , Citometría de Flujo , Fase G2/efectos de los fármacos , Fase G2/fisiología , Humanos , Masculino , Monocrotalina/toxicidad , Nocodazol/farmacología , Arteria Pulmonar/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Fosfatasas cdc25/metabolismo
11.
J Biol Chem ; 275(37): 29091-9, 2000 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-10875930

RESUMEN

A single administration of monocrotaline to rats results in pathologic alterations in the lung and heart similar to human pulmonary hypertension. In order to produce these lesions, monocrotaline is oxidized to monocrotaline pyrrole in the liver followed by hematogenous transport to the lung where it injures pulmonary endothelium. In this study, we determined specific endothelial targets for (14)C-monocrotaline pyrrole using two-dimensional gel electrophoresis and autoradiographic detection of protein metabolite adducts. Selective labeling of specific proteins was observed. Labeled proteins were digested with trypsin, and the resulting peptides were analyzed using matrix-assisted laser desorption ionization mass spectrometry. The results were searched against sequence data bases to identify the adducted proteins. Five abundant adducted proteins were identified as galectin-1, protein-disulfide isomerase, probable protein-disulfide isomerase (ER60), beta- or gamma-cytoplasmic actin, and cytoskeletal tropomyosin (TM30-NM). With the exception of actin, the proteins identified in this study have never been identified as potential targets for pyrroles, and the majority of these proteins have either received no or minimal attention as targets for other electrophilic compounds. The known functions of these proteins are discussed in terms of their potential for explaining the pulmonary toxicity of monocrotaline.


Asunto(s)
Endotelio Vascular/metabolismo , Hemaglutininas/metabolismo , Monocrotalina/análogos & derivados , Proteína Disulfuro Isomerasas/metabolismo , Arteria Pulmonar/metabolismo , Actinas/metabolismo , Adulto , Secuencia de Aminoácidos , Autorradiografía , Células Cultivadas , ADN/metabolismo , Femenino , Galectina 1 , Humanos , Espectrometría de Masas , Datos de Secuencia Molecular , Monocrotalina/metabolismo , Mapeo Peptídico , Tropomiosina/metabolismo
12.
Toxicol Appl Pharmacol ; 152(1): 138-44, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9772209

RESUMEN

One of the earliest morphologic changes evident in the monocrotaline (MCT) model of pulmonary hypertension in rats is microvascular leak. Whether this represents a direct effect of MCT metabolites or is secondary to inflammatory and thrombotic changes remains uncertain. To determine whether MCT directly affects endothelial cell permeability barrier function, we characterized the interaction of the reactive pyrrole intermediate of MCT (MCTP) with endothelial cell actin and characterized its effects on thrombin-mediated signal transduction and monolayer permeability. Bovine pulmonary endothelial cells (BPAEC) treated with MCTP had altered distribution of filamentous actin evident by fluorescence microscopy. Correlative Western blots and autoradiography of actin isolated from BPAEC treated with 14C-MCTP showed comigration of actin and MCTP-derived 14C. MCTP treatment did not alter cellular free Ca2+ concentrations nor did it interfere with thrombin-mediated intracellular Ca2+ signal. Pretreatment with MCTP significantly augmented the thrombin-mediated transudation of Evan's blue albumin in BPAEC monolayers apparently by increasing the size of intercellular gaps. We conclude that MCTP directly interacts with actin to alter its polymerization state but does not significantly affect endothelial cell response to contractile stimulus. Our results suggest that MCTP may affect endothelial cell barrier function through alterations in intracellular junctions.


Asunto(s)
Actinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Monocrotalina/análogos & derivados , Arteria Pulmonar/efectos de los fármacos , Trombina/farmacología , Animales , Autorradiografía , Western Blotting , Calcio/metabolismo , Bovinos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Monocrotalina/farmacología , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Albúmina Sérica/metabolismo , Transducción de Señal/efectos de los fármacos
13.
Toxicol Appl Pharmacol ; 151(2): 236-44, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9707500

RESUMEN

In the monocrotaline (MCT) model of pulmonary hypertension, the pulmonary vascular endothelium is the likely early target of the reactive metabolite monocrotaline pyrrole (MCTP). Incubation of cultured bovine pulmonary arterial endothelial cells (BPAEC) with MCTP results in covalent binding to DNA, cell cycle arrest, and delayed but progressive cell death. The mode of cell death in MCTP-induced endothelial damage has not been addressed previously. Since DNA damage is frequently associated with apoptosis, the presence or absence of apoptosis in adherent BPAEC was determined by several techniques, including morphologic and terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling. Two concentrations of MCTP (5 and 34.5 microgram/ml) along with a vehicle control were examined with each assay. Both concentrations of MCTP induced increasing numbers of cells to undergo apoptosis over time beginning as early as 6 h after exposure to MCTP in the high concentration group. Control and vehicle control cells exhibited small amounts of apoptosis (1-2%), which did not change over the duration of the experiment. Additionally, cell membrane integrity was assessed over time by either exposure to membrane-impermeant dyes or measuring LDH release. By either method, BPAEC had increased membrane permeability after about 48 h of either low or high concentration MCTP exposure. We conclude that both a low or high concentration of MCTP causes cell death in BPAEC by inducing apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Monocrotalina/análogos & derivados , Arteria Pulmonar/efectos de los fármacos , Animales , Bovinos , Células Cultivadas , ADN Nucleotidilexotransferasa/efectos de los fármacos , ADN Nucleotidilexotransferasa/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , L-Lactato Deshidrogenasa/efectos de los fármacos , L-Lactato Deshidrogenasa/metabolismo , Microscopía Fluorescente , Monocrotalina/administración & dosificación , Monocrotalina/farmacología , Factores de Tiempo
14.
Am J Respir Cell Mol Biol ; 19(1): 129-42, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9651189

RESUMEN

Monocrotaline pyrrole (MCTP), a metabolite of the pyrrolizidine alkaloid monocrotaline, is thought to initiate damage to pulmonary endothelial cells resulting in delayed but progressive pulmonary interstitial edema, vascular wall remodeling, and increasing pulmonary hypertension. MCTP was previously shown to inhibit pulmonary endothelial cell proliferation and cause cell-cycle arrest in vitro. To determine the persistence of arrest and better characterize the cell-cycle stage at which it occurs, bovine pulmonary artery endothelial cells (BPAEC) under differing growth conditions were exposed to low (5 microg/ml) or high (34.5 microg/ml) concentrations of MCTP for varying times. Flow cytometric cell-cycle analysis was coupled with Western blot and biochemical analysis of cdc2 kinase and measurements of cell size. MCTP treatment induced a G2 + M phase arrest in 48-h exposed confluent BPAEC that persisted for at least 28 d and was associated with continued cellular enlargement. A short-duration MCTP exposure of confluent (low and high concentration) and log phase (high concentration) BPAEC caused persistent cell-cycle arrest for 1 wk, whereas a low-concentration exposure in log phase cells resulted in cell-cycle arrest with reversal 96 h after exposure. Western blot examination revealed that by 24 h of MCTP exposure, the phosphorylation state of cdc2 was consistent with the inactive form of the kinase (confirmed by biochemical assay); this alteration persisted through at least 96 h of exposure. We conclude that MCTP induces a progressive irreversible endothelial cell dysfunction leading to inactivation of cdc2 kinase and irreversible cell-cycle arrest at the G2 checkpoint.


Asunto(s)
Proteína Quinasa CDC2/antagonistas & inhibidores , Endotelio Vascular/citología , Fase G2/efectos de los fármacos , Mitosis/efectos de los fármacos , Monocrotalina/análogos & derivados , Animales , Western Blotting , Proteína Quinasa CDC2/metabolismo , Bovinos , Tamaño de la Célula , Células Cultivadas , Relación Dosis-Respuesta a Droga , Endotelio Vascular/enzimología , Monocrotalina/farmacología , Fosforilación , Arteria Pulmonar , Factores de Tiempo
15.
Res Commun Mol Pathol Pharmacol ; 99(1): 53-68, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9523355

RESUMEN

After being dehydrogenated by cytochrome P450 enzymes in the liver, monocrotaline's highly-reactive pyrrole metabolite, dehydromonocrotaline, is believed to interact with pulmonary artery endothelial cells to initiate a pulmonary vascular toxicity resembling pulmonary hypertension. Glutathione, an abundant antioxidant in pulmonary artery endothelial cells, has been shown to react with and detoxify the pyrrolic metabolites derived from monocrotaline in the liver. Using high-performance liquid chromatography with electrochemical detection, glutathione levels were measured in a time- and dose-dependent manner in human pulmonary artery endothelial cells following treatment with dehydromonocrotaline, dehydroretronecine and N-ethylmaleimide and bovine pulmonary artery endothelial cells after treatment with dehydromonocrotaline. The bovine cells had 40% less glutathione than the human in the control groups. Bovine pulmonary artery endothelial glutathione levels were depleted 20% more than the human at 15 minutes when treated with 100 microM dehydromonocrotaline. 15 microM N-ethylmaleimide caused an 80% depletion of glutathione compared to a 30% depletion with 15 microM dehydromonocrotaline in human pulmonary artery endothelial cells.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Glutatión/metabolismo , Monocrotalina/análogos & derivados , Arteria Pulmonar/efectos de los fármacos , Análisis de Varianza , Animales , Bovinos , Células Cultivadas , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Etilmaleimida/toxicidad , Humanos , Monocrotalina/toxicidad , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Factores de Tiempo
16.
J Biochem Mol Toxicol ; 12(3): 157-66, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9522275

RESUMEN

The metabolism and covalent binding of 14C-monocrotaline in Sprague-Dawley (SD) rat liver microsomes was investigated using the inducers dexamethasone, clotrimazole, pregnenolone-16 alpha-carbonitrile, and phenobarbital. Monocrotaline is a pyrrolizidine alkaloid (PA) that causes a syndrome in rats that is a model for human primary pulmonary hypertension. It has been documented that bioactivation of PAs (dehydrogenation to reactive pyrroles) in the liver by cytochromes P450 is required for their toxicity. Covalent binding of these reactive pyrroles to tissue macromolecules has been hypothesized to correspond to PA toxicosis. We correlated metabolism and total microsomal covalent binding of 14C-monocrotaline with cytochrome P450 3A using the aforementioned inducers, troleandomycin (a cytochrome P450 3A inhibitor), erythromycin N-demethylase assay of cytochrome P450 3A activity, and Western blots employing anti-rat cytochrome P450 3A antibodies. In addition, autoradiography of membranes electroblotted from SDS-PAGE demonstrated the formation of radiolabeled adducts with specific protein(s). The most intensely radiolabeled protein bands have an apparent molecular weight of approximately 52 kDa, which was similar to the molecular weight detected by anti-rat cytochrome P450 3A antibodies in the Western blots. No radiolabeled proteins were detected in microsomes pretreated with troleandomycin.


Asunto(s)
Hidrocarburo de Aril Hidroxilasas , Sistema Enzimático del Citocromo P-450/metabolismo , Microsomas Hepáticos/enzimología , Monocrotalina/metabolismo , Oxidorreductasas N-Desmetilantes/metabolismo , Animales , Sitios de Unión , Radioisótopos de Carbono/metabolismo , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/biosíntesis , Inducción Enzimática , Masculino , Microsomas Hepáticos/metabolismo , Oxidorreductasas N-Desmetilantes/biosíntesis , Ratas , Ratas Sprague-Dawley
17.
Chem Res Toxicol ; 10(6): 694-701, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9208177

RESUMEN

The association of radiolabeled monocrotaline pyrrole (DHM) with red blood cell (RBCs) ghosts, globins, and heme was examined to determine their role in the transport and stabilization of this hepatic produced putative toxic metabolite of the pyrrolizidine alkaloid monocrotaline (MCT). Rats were administered 5 mg of DHM/kg, i.v., and RBCs and plasma were harvested at 4 and 24 h. Extensive washing of the RBCs with isotonic phosphate buffer did not decrease the amount of radioactivity associated with the cells. The level of DHM equivalents recovered in the RBCs did not decrease between 4 and 24 h, while the plasma levels, which were 29- and 75-fold lower, respectively, decreased from 5.0 to 2.2 nmol of DHM equiv/g of plasma. Globin chains were found to contain 383 and 453 pmol of DHM equiv/mg of protein, respectively. Rats receiving 10 mg of DHM/kg, i.v., with RBCs collected at 2 h, had approximately double the level of radioactivity associated with their RBCs in addition to 2 times the amount of adducts on the globin chains. Globins and ghosts plus heme (2 h) contained 69% and 2% of the radioactivity, respectively. Globin chains treated with an acidic ethanol solution containing AgNO3 resulted in the removal of 31% of the associated radioactivity. GC/ MS and TLC separation of AgNO3-displaced material revealed the presence of the ethyl ether derivatives of 7-hydroxy-1-(hydroxymethyl)-6,7-dihydro-5H-pyrrolizine. The HPLC separation of globin chains revealed that the majority of radioactivity coeluted with the beta-chains. In conclusion, this study found that the administration of radiolabeled DHM resulted in extensive radioactive labeling of RBCs; similar findings have been reported for [14C]MCT.


Asunto(s)
Eritrocitos/metabolismo , Monocrotalina/análogos & derivados , Animales , Transporte Biológico , Cromatografía Líquida de Alta Presión , Eritrocitos/química , Eritrocitos/citología , Globinas/química , Hemo/química , Inyecciones Intravenosas , Masculino , Membranas/química , Monocrotalina/química , Monocrotalina/farmacocinética , Fragmentos de Péptidos/química , Ratas , Ratas Sprague-Dawley , Distribución Tisular
18.
Comp Biochem Physiol B Biochem Mol Biol ; 117(1): 115-23, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9180020

RESUMEN

The metabolism and distribution of 14C-monocrotaline in Fisher 344 (F344) rats was compared with that in Sprague-Dawley (SD) rats. In vitro microsomal preparations, in situ isolated perfused livers and in vivo excretion and distribution studies were used to discern any differences between these two strains. These strains have previously been shown to differ in their susceptibility to monocrotaline-induced pulmonary hypertension. Hepatic phase I metabolism appears to be similar in both strains with N-oxidation and dehydrogenation to the reactive pyrroles as the major pathways. During the liver perfusions, SD rats generated more monocrotalic acid than F344 rats, but the microsome and excretion studies demonstrated no significant differences in the amount of monocrotalic acid. Monocrotalic acid is a stable byproducer of dehydromonocrotaline reacting with cellular nucleophiles and indicates the amount of monocrotaline dehydrogenation when carboxylesterase activity is negligible. These data suggest that the differences in strain susceptibility to pulmonary vascular toxicity is most likely due to differences in their response to the toxic metabolites.


Asunto(s)
Hígado/metabolismo , Microsomas Hepáticos/metabolismo , Monocrotalina/metabolismo , Animales , Bilis/química , Biotransformación , Carboxilesterasa , Hidrolasas de Éster Carboxílico/metabolismo , Cromatografía Líquida de Alta Presión , Heces/química , Glutatión/metabolismo , Hipertensión Pulmonar/inducido químicamente , Masculino , Monocrotalina/análogos & derivados , Monocrotalina/análisis , Monocrotalina/toxicidad , Perfusión , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Especificidad de la Especie , Distribución Tisular
19.
Toxicol Appl Pharmacol ; 143(1): 196-204, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9073608

RESUMEN

Metabolites of the pyrrolizidine alkaloid monocrotaline cause progressive development of pulmonary hypertension in rats. The putative reactive intermediate monocrotaline pyrrole (MCTP) has been shown to cause cytotoxicity, hypertrophy, decreased proliferation, and altered synthetic capability in cultured pulmonary endothelial cells. We compared effects of monocrotaline (MCT) at 60 micrograms/ml (0.185 mM) with previously identified metabolites, MCTP 10 micrograms/ml (0.031 mM) and glutathione-conjugated dihydropyrrolizine (GSH-DHP) 60 micrograms/ml (0.135 mM), in cultured bovine pulmonary artery endothelial cells (BPAECs). To determine whether endothelial metabolism might contribute to the mechanism of this toxicity, we used markers of cytotoxicity (LDH release), synthetic activity (PGI2 synthesis), hypertrophy (planimetry), cell density (cell count/area), and Evans blue albumin (EBA) transudation as a marker for loss of fluid barrier integrity. We found changes in all endothelial markers with MCTP only. MCTP caused increased LDH release by 48 hr, augmented PGI2 synthesis by 96 hr, and resulted in hypertrophy and decreased cell density by 48 hr that persisted at least 21 days. There was increased EBA transudation at 24 hr posttreatment. We concluded that, based on markers of endothelial damage, BPAECs showed no apparent ability to metabolize MCT to a reactive intermediate nor to further metabolize GSH-DHP to a toxic species. We also concluded that MCTP can cause a direct effect on fluid barrier integrity of endothelial cell monolayers in the absence of inflammation.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Monocrotalina/toxicidad , Venenos/toxicidad , 6-Cetoprostaglandina F1 alfa/biosíntesis , Animales , Biomarcadores , Bovinos , Recuento de Células/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , L-Lactato Deshidrogenasa/efectos de los fármacos , L-Lactato Deshidrogenasa/metabolismo , Monocrotalina/metabolismo , Permeabilidad/efectos de los fármacos , Venenos/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología
20.
Toxicol Appl Pharmacol ; 141(1): 319-29, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8917705

RESUMEN

In the monocrotaline (MCT) rat model of pulmonary hypertension, the pulmonary vascular endothelium is thought to be the early target of the bifunctionally reactive metabolite monocrotaline pyrrole (MCTP). In previous studies, bovine pulmonary arterial endothelial cells (BPAEC) exposed to MCTP exhibited inhibition of proliferation. Since other compounds that crosslink DNA lead to cell cycle alterations, we utilized BPAEC to correlate the effects of MCTP on the cell cycle with the extent of covalent binding of [14C]MCTP to BPAEC DNA. Dose response (0.0 to 50.0 micrograms MCTP/ ml) and 96-hr time course (5 micrograms MCTP/ml low dose or 34.5 micrograms MCTP/ml high dose) studies were carried out followed by flow cytometric cell cycle analysis. High concentrations of MCTP caused cell cycle arrest in S phase, beginning by 24 hr, while an S phase delay was observed at low concentrations, but progressed to a G2 + M phase arrest by 48 hr. Covalent DNA binding (34.5 micrograms/ml of [14C]MCTP incubated with BPAEC) occurred within 1 hr and progressively increased through 96 hr. In conclusion, covalent binding of MCTP to DNA is associated with cell cycle arrest; however, the position of cell cycle arrest is dependent on dose, with an S phase arrest at high concentrations and a G2 + M phase arrest at low concentrations of MCTP. The mechanism by which MCTP induces proliferative inhibition could be cell cycle arrest.


Asunto(s)
Ciclo Celular/efectos de los fármacos , ADN/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Monocrotalina/análogos & derivados , Animales , Bovinos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Monocrotalina/metabolismo , Monocrotalina/farmacología , Arteria Pulmonar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...