Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 205
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurochem Res ; 41(10): 2559-2572, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27321306

RESUMEN

Long-term exposure to inorganic arsenic (iAs) through drinking water has been associated with cognitive impairment in children and adults; however, the related pathogenic mechanisms have not been completely described. Increased or chronic inflammation in the brain is linked to impaired cognition and neurodegeneration; iAs induces strong inflammatory responses in several cells, but this effect has been poorly evaluated in central nervous system (CNS) cells. Because astrocytes are the most abundant cells in the CNS and play a critical role in brain homeostasis, including regulation of the inflammatory response, any functional impairment in them can be deleterious for the brain. We propose that iAs could induce cognitive impairment through inflammatory response activation in astrocytes. In the present work, rat cortical astrocytes were acutely exposed in vitro to the monomethylated metabolite of iAs (MMAIII), which accumulates in glial cells without compromising cell viability. MMAIII LD50 in astrocytes was 10.52 µM, however, exposure to sub-toxic MMAIII concentrations (50-1000 nM) significantly increased IL-1ß, IL-6, TNF-α, COX-2, and MIF-1 gene expression. These effects were consistent with amyloid precursor protein (APP) and ß-secretase (BACE-1) increased gene expression, mainly for those MMAIII concentrations that also induced TNF-α over-expression. Other effects of MMAIII on cortical astrocytes included increased proliferative and metabolic activity. All tested MMAIII concentrations led to an inhibition of intracellular lactate dehydrogenase (LDH) activity. Results suggest that MMAIII induces important metabolic and functional changes in astrocytes that may affect brain homeostasis and that inflammation may play a major role in cognitive impairment-related pathogenicity in As-exposed populations.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Ácido Aspártico Endopeptidasas/metabolismo , Astrocitos/efectos de los fármacos , Citocinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Compuestos Organometálicos/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Homeostasis/efectos de los fármacos , Ratas Wistar
2.
Toxicol Appl Pharmacol ; 258(1): 10-8, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22015448

RESUMEN

The association between chronic human exposure to arsenicals and bladder cancer development is well recognized; however, the underlying molecular mechanisms have not been fully determined. We propose that inflammatory responses can play a pathogenic role in arsenic-related bladder carcinogenesis. In previous studies, it was demonstrated that chronic exposure to 50 nM monomethylarsenous acid [MMA(III)] leads to malignant transformation of an immortalized model of urothelial cells (UROtsa), with only 3 mo of exposure necessary to trigger the transformation-related changes. In the three-month window of exposure, the cells over-expressed pro-inflammatory cytokines (IL-1ß, IL-6 and IL-8), consistent with the sustained activation of NFKß and AP1/c-jun, ERK2, and STAT3. IL-8 was over-expressed within hours after exposure to MMA(III), and sustained over-expression was observed during chronic exposure. In this study, we profiled IL-8 expression in UROtsa cells exposed to 50 nM MMA(III) for 1 to 5 mo. IL-8 expression was increased mainly in cells after 3 mo MMA(III) exposure, and its production was also found increased in tumors derived from these cells after heterotransplantation in SCID mice. UROtsa cells do express both receptors, CXCR1 and CXCR2, suggesting that autocrine cell activation could be important in cell transformation. Supporting this observation and consistent with IL-8 over-expression, CXCR1 internalization was significantly increased after three months of exposure to MMA(III). The expression of MMP-9, cyclin D1, bcl-2, and VGEF was significantly increased in cells exposed to MMA(III) for 3 mo, but these mitogen-activated kinases were significantly decreased after IL-8 gene silencing, together with a decrease in cell proliferation rate and in anchorage-independent colony formation. These results suggest a relevant role of IL-8 in MMA(III)-induced UROtsa cell transformation.


Asunto(s)
Transformación Celular Neoplásica/inducido químicamente , Interleucina-8/fisiología , Compuestos Organometálicos/toxicidad , Neoplasias de la Vejiga Urinaria/inducido químicamente , Animales , Proliferación Celular , Células Cultivadas , Humanos , Masculino , Ratones , Ratones SCID , Invasividad Neoplásica , Receptores de Interleucina-8A/análisis , Receptores de Interleucina-8B/análisis , Neoplasias de la Vejiga Urinaria/patología , Urotelio
3.
Toxicol Appl Pharmacol ; 257(1): 1-13, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21925530

RESUMEN

Exposure of human bladder urothelial cells (UROtsa) to 50 nM of the arsenic metabolite, monomethylarsonous acid (MMA(III)), for 12 weeks results in irreversible malignant transformation. The ability of continuous, low-level MMA(III) exposure to cause an increase in genotoxic potential by inhibiting repair processes necessary to maintain genomic stability is unknown. Following genomic insult within cellular systems poly(ADP-ribose) polymerase-1 (PARP-1), a zinc finger protein, is rapidly activated and recruited to sites of DNA strand breaks. When UROtsa cells are continuously exposed to 50 nM MMA(III), PARP-1 activity does not increase despite the increase in MMA(III)-induced DNA single-strand breaks through 12 weeks of exposure. When UROtsa cells are removed from continuous MMA(III) exposure (2 weeks), PARP-1 activity increases coinciding with a subsequent decrease in DNA damage levels. Paradoxically, PARP-1 mRNA expression and protein levels are elevated in the presence of continuous MMA(III) indicating a possible mechanism to compensate for the inhibition of PARP-1 activity in the presence of MMA(III). The zinc finger domains of PARP-1 contain vicinal sulfhydryl groups which may act as a potential site for MMA(III) to bind, displace zinc ion, and render PARP-1 inactive. Mass spectrometry analysis demonstrates the ability of MMA(III) to bind a synthetic peptide representing the zinc-finger domain of PARP-1, and displace zinc from the peptide in a dose-dependent manner. In the presence of continuous MMA(III) exposure, continuous 4-week zinc supplementation restored PARP-1 activity levels and reduced the genotoxicity associated with MMA(III). Zinc supplementation did not produce an overall increase in PARP-1 protein levels, decrease the levels of MMA(III)-induced reactive oxygen species, or alter Cu-Zn superoxide dismutase levels. Overall, these results present two potential interdependent mechanisms in which MMA(III) may increase the susceptibility of UROtsa cells to genotoxic insult and/or malignant transformation: elevated levels of MMA(III)-induced DNA damage through the production of reactive oxygen species, and the direct MMA(III)-induced inhibition of PARP-1.


Asunto(s)
Carcinógenos/toxicidad , Transformación Celular Neoplásica/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Compuestos Organometálicos/toxicidad , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Especies Reactivas de Oxígeno/metabolismo , Vejiga Urinaria/efectos de los fármacos , Urotelio/efectos de los fármacos , Western Blotting , Línea Celular , Transformación Celular Neoplásica/metabolismo , Cloruros/farmacología , Ensayo Cometa , Citometría de Flujo , Humanos , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Vejiga Urinaria/citología , Neoplasias de la Vejiga Urinaria/inducido químicamente , Urotelio/citología , Compuestos de Zinc/farmacología
5.
Toxicol Appl Pharmacol ; 241(2): 202-9, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19699219

RESUMEN

Malignant transformation was demonstrated in UROtsa cells following 52-weeks of exposure to 50 nM monomethylarsonous acid (MMA(III)); the result was the malignantly transformed cell line, URO-MSC. URO-MSC cells were used to study the induction of DNA damage and the alteration of DNA repair enzymes in both the presence of MMA(III) [URO-MSC(+)] and after subsequent removal of MMA(III) [URO-MSC(-)] following chronic, low-level exposure. In the presence of MMA(III), URO-MSC(+) cells demonstrated a sustained increase in DNA damage following 12-weeks of exposure; in particular, a significant increase in DNA single-strand breaks at 12-weeks of exposure consistently elevated through 52 weeks. The persistence of DNA damage in URO-MSC cells was assessed after a 2-week removal of MMA(III). URO-MSC(-) cells demonstrated a decrease in DNA damage compared to URO-MSC(+); however, DNA damage in URO-MSC(-) remained significantly elevated when compared to untreated UROtsa and increased in a time-dependent manner. Reactive oxygen species (ROS) were demonstrated to be a critical component in the generation of DNA damage determined through the incubation of ROS scavengers with URO-MSC cells. Poly (ADP-ribose) polymerase (PARP) is a key repair enzyme in DNA single-strand break repair. URO-MSC(+) resulted in a slight increase in PARP activity after 36-weeks of MMA(III) exposure, suggesting the presence of MMA(III) is inhibiting the increase in PARP activity. In support, PARP activity in URO-MSC(-) increased significantly, coinciding with a subsequent decrease in DNA damage demonstrated in URO-MSC(-) compared to URO-MSC(+). These data demonstrate that chronic, low-level exposure of UROtsa cells to 50 nM MMA(III) results in: the induction of DNA damage that remains elevated upon removal of MMA(III); increased levels of ROS that play a role in MMA(III) induced-DNA damage; and decreased PARP activity in the presence of MMA(III).


Asunto(s)
Carcinógenos/toxicidad , Daño del ADN , Compuestos Organometálicos/toxicidad , Vejiga Urinaria/efectos de los fármacos , Carcinógenos/administración & dosificación , Células Cultivadas , Ensayo Cometa , Reparación del ADN/efectos de los fármacos , ADN de Cadena Simple/efectos de los fármacos , ADN de Cadena Simple/metabolismo , Esquema de Medicación , Humanos , Compuestos Organometálicos/administración & dosificación , Poli(ADP-Ribosa) Polimerasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Fluorescencia , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/inducido químicamente , Neoplasias de la Vejiga Urinaria/patología
6.
Toxicology ; 255(1-2): 107-14, 2009 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19014992

RESUMEN

UROtsa cells exposed to 50 nM monomethylarsonous acid [MMA(III)] for 52 wk (MSC52) achieved hyperproliferation, anchorage independent growth, and enhanced tumorgenicity. MMA(III) has been shown to induce reactive oxygen species (ROS), which can lead to activation of signaling cascades causing stress-related proliferation of cells and even cellular transformation. Previous research established the acute activation of MAPK signaling cascade by ROS produced by MMA(III) as well as chronic up regulation of COX-2 and EGFR in MSC52 cells. To determine if ROS played a role in the chronic pathway perturbations by acting as secondary messengers, activation of Ras was determined in UROtsa cells [exposed to MMA(III) for 0-52 wk] and found to be increased through 52 wk most dramatically after 20 wk of exposure. Ras has been shown to cause an increase in O2(-) and be activated by increases in O2(-), making ROS important to study in the transformation process. COX-2 upregulation in MSC52 cells was confirmed by real time RT-PCR. By utilizing both antioxidants or specific COX inhibitors, it was shown that COX-2 upregulation was dependent on ROS, specifically, O2(-). In addition, because previous research established the importance of MAPK activation in phenotypic changes associated with transformation in MSC52 cells, it was hypothesized that ROS play a role in maintaining phenotypic characteristics of the malignant transformation of MSC52 cells. Several studies have demonstrated that cancer cells have lowered superoxide dismutase (MnSOD) activity and protein levels. Increasing levels of MnSOD have been shown to suppress the malignant phenotype of cells. SOD was added to MSC52 cells resulting in slower proliferation rates (doubling time=42h vs. 31h). ROS scavengers of OH also slowed proliferation rates of MSC52 cells. To further substantiate the importance of ROS in these properties of transformation in MSC52 cells, anchorage independent growth was assessed after the addition of antioxidants, both enzymatic and non-enzymatic. Scavengers of OH, and O2(-) blocked the colony formation of MSC52 cells. These data support the role for the involvement of ROS in properties of transformation of UROtsa cells exposed to MMA(III).


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Compuestos Organometálicos/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Adenosina Trifosfato/metabolismo , Antioxidantes/farmacología , Western Blotting , Caspasa 3/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Genes ras , Humanos , Ácidos Nucleicos/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Toxicology ; 248(2-3): 67-76, 2008 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-18456381

RESUMEN

Arsenical-induced carcinogenesis in human bladder has been established through epidemiological evidence, and UROtsa cells, a normal, immortalized cell culture model of human urothelium, have proven to be a good model for the bladder epithelium. This cell line does not form tumors when injected into immuno-compromised mice nor does it have anchorage-independent growth. UROtsa can be easily manipulated for acute studies related to arsenical exposure. They have been shown to be sensitive to all arsenicals, in particular, the trivalent species, arsenite and monomethylarsonous acid. UROtsa cells have also opened the area of cellular signaling alterations following subcytotoxic exposure to arsenicals in both the acute and long-term time points. In addition, UROtsa cells were shown to be malignantly transformed following low-level exposure to both As(III) and MMA(III) providing additional models for studying arsenical-induced carcinogenesis of the bladder. These transformed cell lines allow researchers the ability to investigate the process of urothelial tumorigenesis at multiple time points of arsenical exposure. Overall, UROtsa cells are an effective model for cellular insult following arsenical exposure.


Asunto(s)
Arsenicales/efectos adversos , Carcinógenos/toxicidad , Carcinoma de Células Transicionales/inducido químicamente , Neoplasias de la Vejiga Urinaria/inducido químicamente , Urotelio/citología , Animales , Carcinoma de Células Transicionales/patología , Línea Celular Transformada , Transformación Celular Neoplásica , Humanos , Modelos Biológicos , Neoplasias de la Vejiga Urinaria/patología
8.
Toxicology ; 250(1): 47-54, 2008 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-18588940

RESUMEN

Arsenicals are known to induce ROS, which can lead to DNA damage, oxidative stress, and carcinogenesis. A human urothelial cell line, UROtsa, was used to study the effects of arsenicals on the human bladder. Arsenite [As(III)] and monomethylarsonous acid [MMA(III)] induce oxidative stress in UROtsa cells after exposure to concentrations as low as 1 microM and 50 nM, respectively. Previous research has implicated ROS as signaling molecules in the MAPK signaling pathway. As(III) and MMA(III) have been shown to increase phosphorylation of key proteins in the MAPK signaling cascade downstream of ErbB2. Both Src phosphorylation (p-Src) and cyclooxygenase-2 (COX-2) are induced after exposure to 50 nM MMA(III) and 1 microM As(III). These data suggest that ROS production is a plausible mechanism for the signaling alterations seen in UROtsa cells after acute arsenical exposure. To determine importance of ROS in the MAPK cascade and its downstream induction of p-Src and COX-2, specific ROS antioxidants (both enzymatic and non-enzymatic) were used concomitantly with arsenicals. COX-2 protein and mRNA was shown to be much more influenced by altering the levels of ROS in cells, particularly after MMA(III) treatment. The antioxidant enzyme superoxide dismutase (SOD) effectively blocked both As(III)-and MMA(III)- associated COX-2 induction. The generation of ROS and subsequent altered signaling did lead to changes in protein levels of SOD, which were detected after treatment with either 1 microM As(III) or 50 nM MMA(III). These data suggest that the generation of ROS by arsenicals may be a mechanism leading to the altered cellular signaling seen after low-level arsenical exposure.


Asunto(s)
Arsenitos/toxicidad , Compuestos Organometálicos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Vejiga Urinaria/efectos de los fármacos , Línea Celular , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Humanos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Vejiga Urinaria/metabolismo
9.
Toxicol In Vitro ; 21(5): 956-61, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17376647

RESUMEN

Precision-cut tissue slices mimic specific organ toxicity because normal cellular heterogeneity and organ architecture are retained. To optimize the use of the smaller tissues of the mouse and to establish easy assays for tissue viability, a tissue chip based system was used to generate large numbers of samples from a single organ. Iodoacetamide (IAM) was used as a model toxicant and assays for intracellular potassium (normalized to DNA content) were used to establish viability and toxicant susceptibility. Thereafter, assays that were more rapid and specific were pursued. Lysates from tissues incubated in 6-carboxyfluorescein fluoresced proportionately to concentrations of IAM, indicating disruption of cellular membranes. Similarly, FURA-2, a probe applied to lysates to measure calcium levels, fluoresced proportionately to IAM dosage. Monobromobimane, a fluorescent sulfhydryl probe, displayed a decrease in fluorescent intensity at higher IAM challenge-a finding confirmed with an absorbance assay with Ellman's reagent. Importantly, the number of samples per organ/mouse was increased at least threefold and a significant time reduction per analysis was realized.


Asunto(s)
Técnicas de Cultivo de Tejidos/instrumentación , Técnicas de Cultivo de Tejidos/métodos , Toxicología/métodos , Alquilantes/toxicidad , Animales , Compuestos Bicíclicos con Puentes , Supervivencia Celular/efectos de los fármacos , Ácido Ditionitrobenzoico , Fluoresceínas , Colorantes Fluorescentes , Fura-2 , Yodoacetamida/toxicidad , Ratones , Ratones Endogámicos C57BL , Microtomía , Potasio/metabolismo , Reactivos de Sulfhidrilo , Sales de Tetrazolio , Tiazoles
10.
Toxicol Lett ; 280: 92-98, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28823542

RESUMEN

UROtsa cells have been accepted as a model to study carcinogenicity mechanisms of arsenic-associated human bladder cancer. In vitro continuous exposure to monomethylarsonous acid (MMAIII), leads UROtsa cells to commit to malignant transformation. In this process, NF-κß-associated inflammatory response seems to play an important role since this transcription factor activates some minutes after cells are exposed in vitro to MMAIII and keeps activated during the cellular malignant transformation. It is known that a slight decrease in the protein phosphatase and tensin homologue (PTEN) gene expression is enough for some cells to become malignantly transformed. Interestingly, this tumor suppressor has been proven to be negatively regulated by NF-κß through binding to its gene promoter. Based on these observations we propose that NF-κß may be involved in arsenic associated carcinogenesis through the negative regulation of PTEN gene expression. Changes in PTEN expression and the binding of p50 NF-κß subunit to PTEN promoter were evaluated in UROtsa cells exposed for 4, 12, 20, or 24 wk to 50nM MMAIII. Results showed that MMAIII induced a significant decrease in PTEN expression around 20 wk exposure to MMAIII,which correlated with increased binding of p50 subunit to the PTEN promoter. Consistent with these results, ChIP assays also showed a significant decrease in H3 acetylation (H3ac) but an increase in the repression marks H3k9me3 and H327me3 in PTEN promoter when compared with not treated cells. These results suggest that the activation of NF-κß by MMAIII may participate in UROtsa cells malignant transformation through the negative regulation of PTEN expression involving p50 homodimers-mediated chromatin remodeling around the PTEN promoter.


Asunto(s)
Histonas/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Compuestos Organometálicos/toxicidad , Fosfohidrolasa PTEN/metabolismo , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Regulación hacia Abajo , Regulación de la Expresión Génica/fisiología , Histonas/genética , Humanos , Metilación , Subunidad p50 de NF-kappa B/genética , Fosfohidrolasa PTEN/genética , Regiones Promotoras Genéticas
11.
Clin Pharmacol Ther ; 37(5): 544-8, 1985 May.
Artículo en Inglés | MEDLINE | ID: mdl-3987178

RESUMEN

The hypothesis that the H2-receptor blockers cimetidine and ranitidine have different effects on the disposition of lidocaine, a microsomally metabolized drug dependent on hepatic blood flow for elimination, was tested. Six normal men received lidocaine infusions (2 mg/kg over 10 minutes) and lidocaine levels were determined by HPLC. Lidocaine kinetics were studied in the untreated state (O) and in a double-blind, double-dummy design after 2 days of placebo (P), cimetidine (C, 300 mg every 6 hours by mouth), or ranitidine (R, 160 mg every 12 hours by mouth). Model-independent kinetics were estimated by the statistical moment theory. The steady-state volume of distribution was lower after cimetidine (means +/- SD: O, 156 +/- 39 L; P, 156 +/- 48 L; C, 123 +/- 20 L; and R, 174 +/- 38 L). A trend toward decreased lidocaine clearance after cimetidine was also noted (O, 1011 +/- 140 ml/min; P, 1087 +/- 227 ml/min; C, 886 +/- 214 ml/min; and R, 1143 +/- 225 ml/min). Elimination rate constants were of the same order in all four treatments. Only higher levels of alpha 1-acid glycoprotein appeared to limit the lidocaine steady-state volume of distribution. Cimetidine and ranitidine have distinctly different effects on lidocaine kinetics in normal subjects. The absence of ranitidine effects on the disposition of lidocaine, a high-extraction, high-clearance drug, suggests that H2-receptor blockade may not decrease hepatic blood flow, and that cimetidine impairs drug elimination only by inhibition of hepatic microsomal enzymes. Such interactions are not likely to occur with ranitidine.


Asunto(s)
Cimetidina/farmacología , Lidocaína/metabolismo , Ranitidina/farmacología , Administración Oral , Adulto , Análisis de Varianza , Cromatografía Líquida de Alta Presión , Método Doble Ciego , Interacciones Farmacológicas , Semivida , Humanos , Infusiones Parenterales , Cinética , Lidocaína/sangre , Masculino , Orosomucoide
12.
Environ Health Perspect ; 101(4): 302-8, 1993 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8275986

RESUMEN

We optimized proton-induced X-ray emission (PIXE) for tissue analysis in a toxicity-disposition study. We used cultured rabbit renal slices as the biological system to demonstrate the use of PIXE analysis. The renal slices were exposed to HgCl2, CdCl2, K2Cr2O7, or NaAsO2 alone or in a mixture. The PIXE analysis provides information on concentrations of elements above atomic number 11, and it is the only analytical technique that can determine 20-30 elements nondestructively in a single, small sample (approximately 5 mg) with detection limits of 1-5 ppm (dry weight). The renal slices are thin targets that yield X-ray emission spectra with low backgrounds and high elemental sensitivities. The nondestructive nature of PIXE and the ability to simultaneously measure uptake of multiple metals and endogenous elements are unique to this methodology.


Asunto(s)
Riñón/química , Animales , Riñón/metabolismo , Masculino , Técnicas de Cultivo de Órganos , Conejos , Manejo de Especímenes/métodos , Espectrometría por Rayos X/instrumentación
13.
Environ Health Perspect ; 103 Suppl 1: 77-80, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7621806

RESUMEN

The uptake and accumulation of metals occurs in the kidney, which is a key site for interaction between metal nephrotoxicants. The uptake/accumulation and interaction of CdCl2, HgCl2, K2Cr2O7, and NaAsO2 was examined in precision-cut rabbit renal cortical slices. Slices were incubated with 10(-6) to 10(-3) M of a single metal toxicant or combinations of metal toxicants for 12 hr in DME-F12 media. Slices were blotted and sandwiched between two mylar films stretched across XRF sample cups. Quantitation of the metal in the slices was performed by proton-induced X-ray emission analysis (PIXE). The uptake of the metals was rapid, often reaching a maximum between 3 to 6 hr; the accumulation of Hg was highest, followed in order by Cd, Cr, and As. When two metals were present together, substantial alterations were observed in the uptake of the metals in the slices. HgCl2 hindered the uptake of K2Cr2O7, NaAsO2, CdCl2 (in this order), whereas these metals facilitated the uptake of HgCl2. However, a decreased uptake of both metals was often noted after exposure to other combinations of metals. PIXE analysis of metal content in slices is attractive since all elements (atomic number > 20) can be determined simultaneously. This information will be particularly useful in studying potential toxic interactions.


Asunto(s)
Corteza Renal/metabolismo , Metales/metabolismo , Animales , Técnicas In Vitro , Masculino , Conejos , Espectrometría por Rayos X
14.
Environ Health Perspect ; 103 Suppl 1: 81-4, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7621807

RESUMEN

The mechanism of renal uptake of nephrotoxic heavy metals such as HgCl2 and NaAsO2 is not clear. The metals are known to react with endogenous sulfhydryls such as glutathione (GSH), so metal-GSH conjugates may be delivered to the kidney. To study this possibility, renal cortical slices from male New Zealand white rabbits were incubated with 10(-4) M HgCl2 or 10(-3) M NaAsO2 +/- stoichiometric amounts (1-3x) of GSH; or synthetic metal-GSH conjugates [10(-4) M Hg(SG)2 or 10(-3) M As(SG)3]. Incubations were performed at 37 degrees C in DME-F12 buffer (95/5 O2/CO2) for 8 hr. Hg(SG)2 reduced slice K+/DNA content, as an indicator of viability, significantly less than HgCl2. As(SG)3 exhibited a 2-hr delay in K+/DNA content reduction compared to NaAsO2. This delay in toxicity was not correlated to changes in uptake. Arsenic and mercury accumulation, determined by proton-induced X-ray emission, were also identical between the metal salts and the metal-GSH conjugates. Exogenous GSH decreased HgCl2 cytotoxicity and was correlated to a decrease in Hg accumulation in the slice. Exogenous GSH had limited if any protective effects against cytotoxicity by NaAsO2 and a decrease in As accumulation was not observed. Complex metal-GSH interactions appear to exist and impact on the uptake and toxicity of these metals.


Asunto(s)
Arsenitos/toxicidad , Glutatión/farmacología , Corteza Renal/efectos de los fármacos , Cloruro de Mercurio/toxicidad , Compuestos de Sodio/toxicidad , Reactivos de Sulfhidrilo/toxicidad , Animales , Arsenitos/metabolismo , Técnicas In Vitro , Corteza Renal/metabolismo , Masculino , Cloruro de Mercurio/metabolismo , Conejos , Compuestos de Sodio/metabolismo , Reactivos de Sulfhidrilo/metabolismo
15.
Toxicol Sci ; 61(2): 283-94, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11353137

RESUMEN

Arsenic is a known human carcinogen. These studies were designed to examine the impact of low arsenite concentrations on immediate early gene expression in precision-cut rat lung slices. Precision-cut lung slices are a versatile in-vitro system for toxicity studies, as they preserve the architecture and cellular heterogeneity of the lung. Since 0.1-100 microM arsenite did not compromise slice viability at 4 hours, effects of arsenite on the expression of c-jun/AP-1, NFkappaB, HSP 32, HSP 72, HSP 60, and HSP 90 were studied, using these concentrations of arsenite at 4 h. Nuclear c-jun was increased by 10 and 100 microM arsenite, while NFkappaB was not affected. Gel-shift assays indicated that 10 microM arsenite resulted in an enhanced DNA-binding activity of both AP-1 and NFkappaB. Confocal microscopic analysis of AP-1 indicated nuclear localization of this transcription factor, mainly in type-II epithelial cells and alveolar macrophages. Nuclear localization of NFkappaB was lower than that observed for AP-1, while most of the NFkappaB was localized to cytoplasm of type-II epithelial cells and alveolar macrophages. HSP 32 was increased by 1.0 and 10 microM arsenite, while HSP 72 was increased by only 100 microM arsenite. HSP 60 and HSP 90 were not changed by arsenite. These studies indicate that noncytotoxic concentrations of arsenite are capable of affecting signal transduction pathways and gene expression in the lung.


Asunto(s)
Arsenitos/farmacología , Proteínas de Unión al ADN/metabolismo , Proteínas de Choque Térmico/biosíntesis , Pulmón/efectos de los fármacos , Pulmón/metabolismo , FN-kappa B/metabolismo , Compuestos de Sodio/farmacología , Factor de Transcripción AP-1/metabolismo , Animales , Arsenitos/toxicidad , Western Blotting , Supervivencia Celular , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Genes jun/efectos de los fármacos , Técnicas In Vitro , Pulmón/patología , Macrófagos Alveolares/metabolismo , Masculino , Microscopía Confocal , Potasio/análisis , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Compuestos de Sodio/toxicidad , Factores de Tiempo
16.
Toxicol Sci ; 51(1): 80-6, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10496679

RESUMEN

The impact of xenobiotics on intercellular adhesion, a fundamental biological process regulating most, if not all, cellular pathways, has been sparsely investigated. Cell-cell adhesion is regulated in the epithelium primarily by the E-cadherin/catenin complex. To characterize the impact of oxidative stress on the E-cadherin/catenin complex, precision-cut mouse liver slices were challenged with two model compounds for the generation of oxidative stress, diamide (DA; 25-250 microM) or t-butylhydroperoxide (tBHP; 5-50 microM), for 6 h. At the concentrations used, neither compound elicited cytotoxicity, as assessed by intracellular K+ content and leakage of lactate dehydrogenase into the culture media. However, a 25% reduction in non-protein sulfhydryl levels, an indication of oxidative perturbation, was seen in liver slices treated with DA or tBHP. Total protein expression of E-cadherin, beta-, or alpha-catenin was not affected by challenge with DA or tBHP. A decrease of beta-catenin in the SDS-soluble fraction of slices, an indicator of the formation of the adhesion complex, was observed. Additionally, a decrease in beta-catenin interactions with E-cadherin and alpha-catenin, as assessed by immunoprecipitation and Western blot analysis, was seen. Disruption of the E-cadherin/catenin complex by tBHP, but not DA, correlated with enhanced tyrosine phosphorylation of beta-catenin. These results suggest that noncytotoxic oxidative stress disrupts the E-cadherin/catenin cell adhesion complex in precision-cut mouse liver slices.


Asunto(s)
Cadherinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Diamida/farmacología , Estrés Oxidativo/efectos de los fármacos , Transactivadores , terc-Butilhidroperóxido/farmacología , Animales , Western Blotting , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Técnicas In Vitro , L-Lactato Deshidrogenasa/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Fosfotirosina/metabolismo , Potasio/metabolismo , alfa Catenina , beta Catenina
17.
Toxicol Sci ; 50(1): 98-105, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10445758

RESUMEN

Although the kidney represents a target for the accumulation and toxicity of arsenic, little is known about the molecular targets of arsenic in this organ. Therefore, these studies were designed to examine the molecular impact of arsenite [As(III)] and arsenate [As(V)] at low (nanomolar) concentrations. Precision-cut rabbit renal cortical slices were challenged with As(III) or As(V) for up to 8 h. Neither form of the metal induced overt cytotoxicity as assessed by intracellular K+ levels over this time period at concentrations from 0.01-10 microM. In addition, no alterations in the expression of Hsp 60, 70, or 90 were observed. However, induction of heme oxygenase-1 (Hsp 32) was seen following a 4-h challenge with As(III), but not with As(V). As(III) and As(V) induced DNA binding of AP-1 at 2- and 4-h exposure; following a 6-h exposure there was no difference. Although no alteration in the DNA binding activity of ATF-2 was induced by As(III) or As(V), both forms enhanced the DNA binding activity of Elk-1. Enhanced DNA binding activity of AP-1 and Elk-1 correlated with increased gene expression of c-fos, but not c-jun, at 2 h. c-myc gene expression was also induced by As(III) and As(V), albeit at a later time point (6 h). These results suggest that acute arsenic challenge, by either As(III) or As(V), is associated with discrete alterations in the activity of signaling pathways and gene expression in renal tissue.


Asunto(s)
Intoxicación por Arsénico , Aductos de ADN/metabolismo , Expresión Génica/efectos de los fármacos , Riñón/efectos de los fármacos , Factores de Transcripción/metabolismo , Animales , Arseniatos/toxicidad , Arsenitos/toxicidad , Western Blotting , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Electroforesis , Técnicas In Vitro , Masculino , Potasio/análisis , Conejos , Teratógenos/toxicidad , Factores de Tiempo
18.
Toxicology ; 89(1): 35-47, 1994 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-8178321

RESUMEN

The anesthetic halothane is bioactivated by the liver cytochrome P450 system to the reactive intermediate, trifluoroacetyl chloride, which can acylate liver protein. Cytosolic glutathione-S-transferase (GST) was identified as a major target for protein adduct formation in guinea pig liver slices exposed to halothane. To determine if GST is also a target in vivo, male Hartley guinea pigs were exposed to 1% halothane in 40% O2 for 4 h. At 10 h post exposure, livers were removed and microsomal and cytosolic fractions prepared. Past studies have shown these conditions resulted in maximal covalent binding of halothane intermediates to hepatic protein. Protein was isolated by ethanol precipitation and washed with trichloroacetic acid to remove unbound metabolites. Cytosolic GST was isolated by gel filtration and S-hexyl-glutathione affinity chromatography to electrophoretic purity. Protein adducts were quantified using a covalently bound fluorine assay. Covalent binding of a halothane intermediate to cytosolic and microsomal protein was determined as 2.0 +/- 0.4 and 13.2 +/- 2.3 nmol F/mg protein, respectively. Liver glutathione depletion by buthionine sulfoximine pretreatment produced an increase in covalent binding only to cytosolic proteins (3.3 +/- 0.4 nmol F/mg protein). Adduct formation to cytosolic GST was determined to be 4.7 +/- 1.6 nmol F/mg protein. Glutathione-S-transferase is a target for covalent modification in the liver following an inhalation exposure to halothane.


Asunto(s)
Glutatión Transferasa/metabolismo , Halotano/metabolismo , Hígado/enzimología , Administración por Inhalación , Animales , Biotransformación , Cromatografía de Afinidad , Citosol/enzimología , Glutatión/fisiología , Glutatión Transferasa/efectos de los fármacos , Cobayas , Halotano/toxicidad , Masculino , Microsomas Hepáticos/enzimología , Unión Proteica
19.
Toxicology ; 120(2): 119-32, 1997 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-9184199

RESUMEN

The anesthetic, halothane, is bioactivated by the liver cytochrome P450 system to trifluoroacetyl-chloride, which can readily acylate liver protein. Covalent binding of the trifluoroacetyl moiety may result in hapten formation leading to the induction of an immune response and ultimately halothane hepatitis. In this study the presence of trifluoroacetylated-protein adducts in Kupffer cells was investigated to learn how the immune system might come in contact with the proteins. Guinea pigs were exposed to 1.0% halothane, 40% oxygen for 4 h. Kupffer cells were isolated on days 1 through 9 post-exposure, by liver perfusion and purification by elutriation. Using gel electrophoresis and Western blotting techniques, it has been demonstrated that Kupffer cells obtained from halothane-treated guinea pigs, do carry trifluoroacetyl-protein adducts as recognized by an anti-trifluoroacetyl-rabbit serum albumin antibody. Apparent molecular weights of polypeptides bound by trifluoroacetyl were of a wide range, 25-152 kDa. Bands were most prominent in the larger Kupffer cells with more appearing at lower molecular weights. Trifluoroacetyl-protein adducts were not detected in lung, spleen, lymph node or peripheral blood macrophages. This work suggests a role for Kupffer cells in the presentation of altered proteins in the liver to cells of the immune system.


Asunto(s)
Anestésicos por Inhalación/toxicidad , Células Presentadoras de Antígenos/efectos de los fármacos , Halotano/toxicidad , Macrófagos del Hígado/efectos de los fármacos , Ácido Trifluoroacético/metabolismo , Acetilación , Anestésicos por Inhalación/administración & dosificación , Animales , Formación de Anticuerpos , Complejo Antígeno-Anticuerpo , Células Presentadoras de Antígenos/citología , Western Blotting , Separación Celular , Supervivencia Celular/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/metabolismo , Electroforesis en Gel de Poliacrilamida , Cobayas , Halotano/administración & dosificación , Macrófagos del Hígado/citología , Ganglios Linfáticos/citología , Ganglios Linfáticos/efectos de los fármacos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos Alveolares/citología , Macrófagos Alveolares/efectos de los fármacos , Masculino , Peso Molecular , Conejos , Albúmina Sérica/inmunología , Organismos Libres de Patógenos Específicos , Bazo/citología , Bazo/efectos de los fármacos , Ácido Trifluoroacético/inmunología
20.
Toxicology ; 26(3-4): 285-94, 1983.
Artículo en Inglés | MEDLINE | ID: mdl-6857701

RESUMEN

Addition of halogenated vinyl cysteine conjugates to isolated rabbit kidney tubule suspensions resulted in a decrease in the active transport of para-aminohippuric acid (PAH) and tetraethylammonium bromide (TEA). At 10(-5) M vinyl cysteine conjugate, tubule to medium accumulation ratios (T/M) were similar to those of controls while at 10(-3) M the T/M values decreased to 1, indicating complete inhibition of active accumulation of PAH or TEA. The decreased active transport was not caused by inhibition of mitochondrial oxidation since incubations in the presence of 10(-3) M halogenated vinyl cysteine did not inhibit tubule O2 utilization or production of 14CO2 from [14C]glucose or [14C]succinate. A mechanism is proposed whereby toxicity may result from covalent binding of an active intermediate, produced by enzyme cleavage, to membrane associated nucleophilic groups thereby decreasing active transport.


Asunto(s)
Hidrocarburos Halogenados/toxicidad , Túbulos Renales Proximales/efectos de los fármacos , Compuestos de Vinilo/toxicidad , Animales , Transporte Biológico Activo/efectos de los fármacos , Glucosa/metabolismo , Técnicas In Vitro , Túbulos Renales Proximales/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Conejos , Succinatos/metabolismo , Ácido Succínico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA