Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Carcinog ; 50(11): 884-900, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21400612

RESUMEN

This study critically examined the role of PPARß/δ in colon cancer models. Expression of PPARß/δ mRNA and protein was lower and expression of CYCLIN D1 protein higher in human colon adenocarcinomas compared to matched non-transformed tissue. Similar results were observed in colon tumors from Apc(+/Min-FCCC) mice compared to control tissue. Dietary administration of sulindac to Apc(+/Min-FCCC) mice had no influence on expression of PPARß/δ in normal colon tissue or colon tumors. Cleaved poly (ADP-ribose) polymerase (PARP) was either increased or unchanged, while expression of 14-3-3ε was not influenced in human colon cancer cell lines cultured with the PPARß/δ ligand GW0742 under conditions known to increase apoptosis. While DLD1 cells exhibited fewer early apoptotic cells after ligand activation of PPARß/δ following treatment with hydrogen peroxide, this change was associated with an increase in late apoptotic/necrotic cells, but not an increase in viable cells. Stable over-expression of PPARß/δ in human colon cancer cell lines enhanced ligand activation of PPARß/δ and inhibition of clonogenicity in HT29 cells. These studies are the most quantitative to date to demonstrate that expression of PPARß/δ is lower in human and Apc(+/Min-FCCC) mouse colon tumors than in corresponding normal tissue, consistent with the finding that increasing expression and activation of PPARß/δ in human colon cancer cell lines inhibits clonogenicity. Because ligand-induced attenuation of early apoptosis can be associated with more late, apoptotic/necrotic cells, but not more viable cells, these studies illustrate why more comprehensive analysis of PPARß/δ-dependent modulation of apoptosis is required in the future.


Asunto(s)
Adenocarcinoma/genética , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , PPAR delta/genética , PPAR-beta/genética , Adenocarcinoma/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR delta/metabolismo , PPAR-beta/metabolismo
2.
Toxicology ; 276(1): 79-84, 2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20637823

RESUMEN

Recent work indicates that PPARalpha is required for perfluorooctanoic acid (PFOA)-induced postnatal lethality resulting from prenatal exposure. The present study tested the hypothesis that relatively modest activation of PPARalpha during prenatal development will cause postnatal lethality, similar to that observed with PFOA, a relatively low affinity PPARalpha agonist. Female wild-type and Pparalpha-null mice were mated overnight with males of the same genotype. The presence of a copulatory plug on the morning after mating was indicative of pregnancy and considered gestation day (GD) 0. Plugged female mice were fed either a control diet or one containing clofibrate (0.5%) or Wy-14,643 (0.005%) until GD18 or until parturition. Mice were examined on GD18 or on postnatal day (PND) 20 following the prenatal exposure period. Dietary administration of clofibrate or Wy-14,643 did not affect maternal weight or weight gain, the average number of implantations, the percentage of litter loss, the average number of live/dead fetuses, average crown-rump length, or the average fetal weight on GD18 in either genotype. An increase in relative maternal liver weight and elevated expression of PPARalpha target genes in maternal and fetal livers on GD18 were observed, indicative of PPARalpha-dependent changes in both the maternal and fetal compartments. However, no defects in postnatal development were observed by either clofibrate or Wy-14,643 in either genotype by PND20. These results demonstrate that relatively low level activation of PPARalpha by clofibrate or Wy-14,643 during prenatal development does not cause postnatal lethality.


Asunto(s)
Clofibrato/farmacología , PPAR alfa/agonistas , Proliferadores de Peroxisomas/farmacología , Pirimidinas/farmacología , Animales , Femenino , Genotipo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , PPAR alfa/genética , Embarazo , Efectos Tardíos de la Exposición Prenatal
3.
Mol Pharmacol ; 78(3): 419-30, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20516370

RESUMEN

The availability of high-affinity agonists for peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) has led to significant advances in our understanding of the functional role of PPARbeta/delta. In this study, a new PPARbeta/delta antagonist, 4-chloro-N-(2-{[5-trifluoromethyl)-2-pyridyl]sulfonyl}ethyl)benzamide (GSK3787), was characterized using in vivo and in vitro models. Orally administered GSK3787 caused antagonism of 4-[2-(3-fluoro-4-trifluoromethyl-phenyl)-4-methyl-thiazol-5-ylmethylsulfanyl]-2-methyl-phenoxy}-acetic acid (GW0742)-induced up-regulation of Angptl4 and Adrp mRNA expression in wild-type mouse colon but not in Pparbeta/delta-null mouse colon. Chromatin immunoprecipitation (ChIP) analysis indicates that this correlated with reduced promoter occupancy of PPARbeta/delta on the Angptl4 and Adrp genes. Reporter assays demonstrated antagonism of PPARbeta/delta activity and weak antagonism and agonism of PPARgamma activity but no effect on PPARalpha activity. Time-resolved fluorescence resonance energy transfer assays confirmed the ability of GSK3787 to modulate the association of both PPARbeta/delta and PPARgamma coregulator peptides in response to ligand activation, consistent with reporter assays. In vivo and in vitro analysis indicates that the efficacy of GSK3787 to modulate PPARgamma activity is markedly lower than the efficacy of GSK3787 to act as a PPARbeta/delta antagonist. GSK3787 antagonized GW0742-induced expression of Angptl4 in mouse fibroblasts, mouse keratinocytes, and human cancer cell lines. Cell proliferation was unchanged in response to either GW0742 or GSK3787 in human cancer cell lines. Results from these studies demonstrate that GSK3787 can antagonize PPARbeta/delta in vivo, thus providing a new strategy to delineate the functional role of a receptor with great potential as a therapeutic target for the treatment and prevention of disease.


Asunto(s)
Receptores Activados del Proliferador del Peroxisoma/metabolismo , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Células/metabolismo , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Ratones , Ratones Noqueados , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR alfa/farmacología , PPAR gamma/genética , PPAR gamma/metabolismo , PPAR gamma/farmacología , PPAR-beta/genética , PPAR-beta/metabolismo , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/farmacología , Tiazoles , Activación Transcripcional , Regulación hacia Arriba/efectos de los fármacos
4.
Toxicol Sci ; 110(1): 204-11, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19359353

RESUMEN

Perfluorobutyrate (PFBA) is a short chain perfluoroalkyl carboxylate that is structurally similar to perfluorooctanoate. Administration of PFBA can cause peroxisome proliferation, induction of peroxisomal fatty acid oxidation and hepatomegaly, suggesting that PFBA activates the nuclear receptor, peroxisome proliferator-activated receptor-alpha (PPAR-alpha). In this study, the role of PPAR-alpha in mediating the effects of PFBA was examined using PPAR-alpha null mice and a mouse line expressing the human PPAR-alpha in the absence of mouse PPAR-alpha (PPAR-alpha humanized mice). PFBA caused upregulation of known PPAR-alpha target genes that modulate lipid metabolism in wild-type and PPAR-alpha humanized mice, and this effect was not found in PPAR-alpha null mice. Increased liver weight and hepatocyte hypertrophy were also found in wild-type and humanized PPAR-alpha mice treated with PFBA, but not in PPAR-alpha null mice. Interestingly, hepatocyte focal necrosis with inflammatory cell infiltrate was only found in wild-type mice administered PFBA; this effect was markedly diminished in both PPAR-alpha null and PPAR-alpha humanized mice. Results from these studies demonstrate that PFBA can modulate gene expression and cause mild hepatomegaly and hepatocyte hypertrophy through a mechanism that requires PPAR-alpha and that these effects do not exhibit a species difference. In contrast, the PPAR-alpha-dependent increase in PFBA-induced hepatocyte focal necrosis with inflammatory cell infiltrate was mediated by the mouse PPAR-alpha but not the human PPAR-alpha. Collectively, these findings demonstrate that PFBA can activate both the mouse and human PPAR-alpha, but there is a species difference in the hepatotoxic response to this chemical.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Fluorocarburos/toxicidad , Hígado/efectos de los fármacos , PPAR alfa/fisiología , Alanina Transaminasa/sangre , Animales , Peso Corporal/efectos de los fármacos , Fluorocarburos/farmacocinética , Humanos , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Tamaño de los Órganos/efectos de los fármacos , Pirimidinas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Toxicol Sci ; 105(2): 418-28, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18622026

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) beta/delta-null mice exhibit exacerbated hepatotoxicity in response to administration of carbon tetrachloride (CCl(4)). To determine whether ligand activation of the receptor protects against chemical toxicity in the liver, wild-type and PPARbeta/delta-null mice were administered CCl(4) with or without coadministration of the highly specific PPARbeta/delta ligand GW0742. Biomarkers of liver toxicity, including serum alanine aminotransferase (ALT) and hepatic tumor necrosis factor (TNF) alpha mRNA, were significantly higher in CCl(4)-treated PPARbeta/delta-null mice compared to wild-type mice. Hepatic expression of TNF-like weak inducer of apoptosis receptor (TWEAKr) and S100 calcium-binding protein A6 (S100A6/calcyclin), genes involved in nuclear factor kappa B signaling, was higher in the CCl(4)-treated PPARbeta/delta-null mice compared to wild-type mice. GW0742 treatment resulted in reduced serum ALT concentration and lower expression of CCl(4)-induced TNF-alpha, S100A6, monocyte chemoattractant protein-1 (MCP1), and TWEAKr in wild-type mice, and these effects were not observed in PPARbeta/delta-null mice. Expression of TNF-alpha was higher in PPARbeta/delta-null primary hepatocytes in response to interleukin-1beta treatment compared to wild-type hepatocytes, but GW0742 did not significantly modulate TNF-alpha expression in hepatocytes from either genotype. While PPARbeta/delta-null hepatic stellate exhibited higher rates of proliferation compared to wild-type cells, GW0742 did not affect alpha-smooth muscle actin expression in these cells. Combined, these findings demonstrate that ligand activation of PPARbeta/delta protects against chemically induced hepatotoxicity by downregulating expression of proinflammatory genes. Hepatocytes and hepatic stellate cells do not appear to directly mediate the inhibitory effects of ligand activation of PPARbeta/delta in liver, suggesting the involvement of paracrine and autocrine events mediated by hepatic cells.


Asunto(s)
Mediadores de Inflamación/metabolismo , Hepatopatías/prevención & control , Hígado/efectos de los fármacos , PPAR delta/agonistas , PPAR-beta/agonistas , Sustancias Protectoras/farmacología , Tiazolidinas/farmacología , Animales , Biomarcadores/metabolismo , Tetracloruro de Carbono , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Ligandos , Hígado/enzimología , Hígado/patología , Hepatopatías/genética , Hepatopatías/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR delta/genética , PPAR delta/metabolismo , PPAR-beta/genética , PPAR-beta/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
Toxicol Appl Pharmacol ; 230(3): 338-45, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18436273

RESUMEN

Administration of a low priming dose of 2-butoxyethanol (BE, 500 mg/kg, p.o.) 7 days prior to a larger LD(90) dose (1500 mg BE/kg, p.o.) offers protection against the lethal dose-induced hemolysis and death in female Sprague Dawley rats because of prompt and efficient replacement of red blood cells (RBCs) with new resilient RBCs. The objective of the present work was to analyze the altered proteome of RBCs upon priming with BE in order to identify the potential anti-hemolytic survival proteins induced in the primed rat RBCs (P-RBCs) as opposed to vehicle-treated RBCs (V-RBCs). The RBCs from the two groups were fractionated into membrane and cytosolic fractions. The cytosolic fractions were further fractionated for proteomic analysis into 3 fractions. The fractions were labeled with Cy3 and Cy5 fluorescent dyes and subjected to 2-dimensional differential gel electrophoresis (DIGE) to analyze the protein profiles. Seven membrane and 8 cytosolic proteins were found to be significantly increased (> or =2.5 fold) in P-RBCs as compared to V-RBCs. The identified proteins can be classified into antioxidant, membrane skeleton, protein turnover, lipid raft, and energy metabolism components. Increased levels of the proteins from antioxidant and membrane skeleton groups were confirmed by Western blot analysis. The study provides the first report on protein profiling of rat RBCs as well as on alteration of the proteome upon exposure to a priming dose of hemotoxicant. Further studies are needed to prove the protective role of the identified proteins and will initiate the field of survival/protective/anti-hemolytic proteins in RBCs.


Asunto(s)
Proteínas Sanguíneas/análisis , Eritrocitos/efectos de los fármacos , Glicoles de Etileno/toxicidad , Proteoma , Animales , Catalasa/sangre , Citosol/química , Metabolismo Energético , Eritrocitos/química , Femenino , Glutatión/metabolismo , Hematócrito , Microdominios de Membrana/química , Proteínas de la Membrana/análisis , Ratas , Ratas Sprague-Dawley
7.
Toxicol Appl Pharmacol ; 225(1): 102-12, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17850838

RESUMEN

Protection against a high dose of a toxicant by prior exposure to another toxicant is called heteroprotection. Our objective was to establish a heteroprotection model in RBCs. Female Sprague Dawley rats treated with an LD90 dose of 2-butoxyethanol (BE, 1500 mg/kg in water, 5 ml/kg po) 14 days after priming with 0.9% NaCl suffered 90% mortality by 15 days, whereas all rats receiving the LD90 dose of BE 14 days after priming with phenylhydrazine (PHZ, 125 mg/kg in 0.9% NaCl, 3 ml/kg po) survived. Hematocrit decreased from normal 45% to 24% by day 3 after PHZ priming and improved thereafter. Increasing the time interval between the priming and LD90 dose to 21 days abolished the heteroprotection. RBCs obtained on days 7 and 14 after PHZ priming unlike those on day 21 were resilient to the hemotoxic metabolite of BE, butoxyacetic acid (BAA). Unaltered hepatic alcohol and aldehyde dehydrogenase activities upon PHZ priming suggested that bioactivation of BE to BAA was unaffected. Lower renal (6 and 12 h) and hepatic (12 h) BAA levels and 3 fold higher excretion of BAA in PHZ-primed rat urine suggested a protective role of toxicokinetics. Higher erythropoietin, reticulocytes, and resiliency of PHZ-primed rat RBCs indicated that newly formed RBCs are resilient to hemolytic BAA. The antioxidant levels in the PHZ-primed rat RBCs did not indicate a protective role in heteroprotection. In conclusion, the resistance of PHZ-primed rats against BE-induced hemotoxicity and lethality is mediated by a combination of altered toxicokinetics, robust erythropoiesis, and resiliency of new RBCs.


Asunto(s)
Eritrocitos/efectos de los fármacos , Glicoles de Etileno/toxicidad , Hemólisis/efectos de los fármacos , Oxidantes/farmacología , Fenilhidrazinas/farmacología , Aldehído Deshidrogenasa/efectos de los fármacos , Aldehído Deshidrogenasa/metabolismo , Animales , Antioxidantes/metabolismo , Eritrocitos/metabolismo , Eritropoyesis/efectos de los fármacos , Glicoles de Etileno/farmacocinética , Femenino , Glicolatos/farmacocinética , Glicolatos/toxicidad , Hematócrito , Ratas , Ratas Sprague-Dawley , Reticulocitos/efectos de los fármacos , Reticulocitos/metabolismo , Factores de Tiempo
8.
Curr Protoc Toxicol ; Chapter 14: Unit14.6, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-23045139

RESUMEN

Many hepatotoxicants like acetaminophen, chloroform, carbon tetrachloride, halothane, and thioacetamide cause hepatotoxicity through covalent binding of their reactive metabolites to proteins. The covalent binding to proteins may lead to dysfunction of critical proteins such as enzymes, transporters, receptors, and regulatory molecules. Because most reactive metabolites covalently bind to tissue macromolecules and tend to be unstable, they can not be isolated, and direct quantitation of the formation of reactive metabolites is not possible. Measuring their covalent binding to proteins offers a convenient way to estimate the amount of reactive metabolite formation. Such estimates have been used to quantify the bioactivation-based injury due to such hepatotoxicants. There are various methods by which covalent binding may be measured. This unit describes a protocol in which a radiolabeled compound can be utilized to measure covalent binding. Alternate protocols involve immunoblotting and immunohistochemistry. The time and method of measuring covalent binding play an important role in the evaluation.


Asunto(s)
Tetracloruro de Carbono/metabolismo , Tetracloruro de Carbono/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hígado/metabolismo , Animales , Biotransformación , Radioisótopos de Carbono , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Immunoblotting , Inmunohistoquímica , Hígado/efectos de los fármacos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Unión Proteica , Ensayo de Unión Radioligante , Ratas
9.
Hepatology ; 44(2): 379-88, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16871587

RESUMEN

Acute liver failure induced by hepatotoxic drugs results from rapid progression of injury. Substantial research has shown that timely liver regeneration can prevent progression of injury leading to a favorable prognosis. However, the mechanism by which compensatory regeneration prevents progression of injury is not known. We have recently reported that calpain released from necrotic hepatocytes mediates progression of liver injury even after the hepatotoxic drug is cleared from the body. By examining expression of calpastatin (CAST), an endogenous inhibitor of calpain in three liver cell division models known to be resistant to hepatotoxicity, we tested the hypothesis that increased CAST in the dividing hepatocytes affords resistance against progression of injury. Liver regeneration that follows CCl(4)-induced liver injury, 70% partial hepatectomy, and postnatal liver development were used. In all three models, CAST was upregulated in the dividing/newly divided hepatocytes and declined to normal levels with the cessation of cell proliferation. To test whether CAST overexpression confers resistance against hepatotoxicity, CAST was overexpressed in the livers of normal SW mice using adenovirus before challenging them with acetaminophen (APAP) overdose. These mice exhibited markedly attenuated progression of liver injury and 57% survival. Whereas APAP-bioactivating enzymes and covalent binding of the APAP-derived reactive metabolites remained unaffected, degradation of calpain specific target substrates such as fodrin was significantly reduced in these mice. In conclusion, CAST overexpression could be used as a therapeutic strategy to prevent progression of liver injury where liver regeneration is severely hampered.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Fallo Hepático Agudo/metabolismo , Regeneración Hepática , Hígado/metabolismo , Acetaminofén/toxicidad , Animales , Animales Recién Nacidos , Proteínas de Unión al Calcio/genética , Calpaína/antagonistas & inhibidores , Tetracloruro de Carbono/toxicidad , División Celular , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hepatocitos/metabolismo , Hepatocitos/patología , Inmunohistoquímica , Hígado/crecimiento & desarrollo , Fallo Hepático Agudo/inducido químicamente , Fallo Hepático Agudo/patología , Masculino , Ratones , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
10.
Toxicol Appl Pharmacol ; 216(1): 108-21, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16815507

RESUMEN

Protection offered by pre-exposure priming with a small dose of a toxicant against the toxic and lethal effects of a subsequently administered high dose of the same toxicant is autoprotection. Although autoprotection has been extensively studied with diverse toxicants in acute exposure regimen, not much is known about autoprotection after priming with repeated exposure. The objective of this study was to investigate this concept following repeated exposure to a common water contaminant, chloroform. Swiss Webster (SW) mice, exposed continuously to either vehicle (5% Emulphor, unprimed) or chloroform (150 mg/kg/day po, primed) for 30 days, were challenged with a normally lethal dose of chloroform (750 mg chloroform/kg po) 24 h after the last exposure. As expected, 90% of the unprimed mice died between 48 and 96 h after administration of the lethal dose in contrast to 100% survival of mice primed with chloroform. Time course studies indicated lower hepato- and nephrotoxicity in primed mice as compared to unprimed mice. Hepatic CYP2E1, glutathione levels (GSH), and covalent binding of (14)C-chloroform-derived radiolabel did not differ between livers of unprimed and primed mice after lethal dose exposure, indicating that protection in liver is neither due to decreased bioactivation nor increased detoxification. Kidney GSH and glutathione reductase activity were upregulated, with a concomitant reduction in oxidized glutathione in the primed mice following lethal dose challenge, leading to decreased renal covalent binding of (14)C-chloroform-derived radiolabel, in the absence of any change in CYP2E1 levels. Buthionine sulfoximine (BSO) intervention led to 70% mortality in primed mice challenged with lethal dose. These data suggest that higher detoxification may play a role in the lower initiation of kidney injury observed in primed mice. Exposure of primed mice to a lethal dose of chloroform led to 40% lower chloroform levels (AUC(15-360 min)) in the systemic circulation. Exhalation of (14)C-chloroform was unchanged in primed as compared to unprimed mice (AUC(1-6 h)). Urinary excretion of (14)C-chloroform was higher in primed mice after administration of the lethal dose. However, neither slightly higher urinary elimination nor unchanged expiration can account for the difference in systemic levels of chloroform. Liver and kidney regeneration was inhibited by the lethal dose in unprimed mice leading to progressive injury, organ failure, and 90% mortality. In contrast, sustained and highly stimulated compensatory hepato- and nephrogenic repair prevented the progression of injury resulting in 100% survival of primed mice challenged with the lethal dose. These findings affirm the critical role of tissue regeneration and favorable detoxification (only in kidney) of the lethal dose of chloroform in subchronic chloroform priming-induced autoprotection.


Asunto(s)
Cloroformo/administración & dosificación , Enfermedades Renales/prevención & control , Hepatopatías/prevención & control , Alanina Transaminasa/metabolismo , Animales , Área Bajo la Curva , Butionina Sulfoximina/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas , Cloroformo/farmacocinética , Cloroformo/toxicidad , Citocromo P-450 CYP2E1/metabolismo , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Glutatión/metabolismo , Glutatión Reductasa/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Intubación Gastrointestinal , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/inducido químicamente , Enfermedades Renales/mortalidad , Hepatopatías/mortalidad , Ratones , Tasa de Supervivencia
11.
Toxicol Appl Pharmacol ; 213(3): 267-81, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16630638

RESUMEN

The aims of the present study were to characterize the subchronic toxicity of chloroform by measuring tissue injury, repair, and distribution of chloroform and to assess the reasons for the development of tolerance to subchronic chloroform toxicity. Male Swiss Webster (SW) mice were given three dose levels of chloroform (150, 225, and 300 mg/kg/day) by gavage in aqueous vehicle for 30 days. Liver and kidney injury were measured by plasma ALT and BUN, respectively, and by histopathology. Tissue regeneration was assessed by (3)H-thymidine incorporation into hepato- and nephro-nuclear DNA and by proliferating cell nuclear antigen staining. In addition, GSH and CYP2E1 in liver and kidney were assessed at selected time points. The levels of chloroform were measured in blood, liver, and kidney during the dosing regimen (1, 7, 14, and 30 days). Kidney injury was evident after 1 day with all three doses and sustained until 7 days followed by complete recovery. Mild to moderate liver injury was observed from 1 to 14 days with all three dose levels followed by gradual decrease. Significantly higher regenerative response was evident in liver and kidney at 7 days, but the response was robust in kidney, preventing progression of injury beyond first week of exposure. While the kidney regeneration reached basal levels by 21 days, moderate liver regeneration with two higher doses sustained through the end of the dosing regimen and 3 days after that. Following repeated exposure for 7, 14, and 30 days, the blood and tissue levels of chloroform were substantially lower with all three dose levels compared to the levels observed with single exposure. Increased exhalation of (14)C-chloroform after repeated exposures explains the decreased chloroform levels in circulation and tissues. These results suggest that toxicokinetics and toxicodynamics (tissue regeneration) contribute to the tolerance observed in SW mice to subchronic chloroform toxicity. Neither bioactivation nor detoxification appears to play a decisive role in the development of this tolerance.


Asunto(s)
Cloroformo/toxicidad , Riñón/efectos de los fármacos , Regeneración Hepática , Hígado/efectos de los fármacos , Regeneración , Animales , Dióxido de Carbono/metabolismo , Cloroformo/sangre , Cloroformo/farmacocinética , Citocromo P-450 CYP2E1/metabolismo , Relación Dosis-Respuesta a Droga , Tolerancia a Medicamentos , Espiración , Glutatión/metabolismo , Riñón/patología , Riñón/fisiología , Hígado/patología , Hígado/fisiología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...