Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 126(7): 926-941, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32213135

RESUMEN

Anthracycline-based chemotherapy can result in the development of a cumulative and progressively developing cardiomyopathy. Doxorubicin is one of the most highly prescribed anthracyclines in the United States due to its broad spectrum of therapeutic efficacy. Interference with different mitochondrial processes is chief among the molecular and cellular determinants of doxorubicin cardiotoxicity, contributing to the development of cardiomyopathy. The present review provides the basis for the involvement of mitochondrial toxicity in the different functional hallmarks of anthracycline toxicity. Our objective is to understand the molecular determinants of a progressive deterioration of functional integrity of mitochondria that establishes a historic record of past drug treatments (mitochondrial memory) and renders the cancer patient susceptible to subsequent regimens of drug therapy. We focus on the involvement of doxorubicin-induced mitochondrial oxidative stress, disruption of mitochondrial oxidative phosphorylation, and permeability transition, contributing to altered metabolic and redox circuits in cardiac cells, ultimately culminating in disturbances of autophagy/mitophagy fluxes and increased apoptosis. We also suggest some possible pharmacological and nonpharmacological interventions that can reduce mitochondrial damage. Understanding the key role of mitochondria in doxorubicin-induced cardiomyopathy is essential to reduce the barriers that so dramatically limit the clinical success of this essential anticancer chemotherapy.


Asunto(s)
Cardiomiopatías/metabolismo , Doxorrubicina/farmacología , Mitocondrias Cardíacas/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/envenenamiento , Apoptosis/efectos de los fármacos , Cardiomiopatías/inducido químicamente , Doxorrubicina/envenenamiento , Humanos , Mitocondrias Cardíacas/metabolismo , Mitofagia/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/metabolismo
2.
Regul Toxicol Pharmacol ; 97: A1-A3, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30017904

RESUMEN

Several recent and prominent articles in Science and Nature deliberately mischaracterized the nature of genuine scientific evidence. Those articles take issue with the United States Environmental Protection Agency's recent proposal to structure its policies and rules only from studies with transparently published raw data. The articles claim it is an effort to obfuscate with transparency, by eliminating a host of studies not offering raw data. A remarkable declaration by a Science editorial is that properly trained experts can verify the scientific evidence of studies without access to raw data, We assert the Agency's proposal must be sustained. Transparency in reporting is a fundamental ethical imperative of objective scientific research justifying massive official regulations and policies. Putative hazards bereft of independent scientific evidence will continue to stoke public anxieties, calling for precautionary regulations and policies. These should rely not on spurious science but on transparent tradeoffs between the smallest exposures compatible with utility and with social perceptions of affordable precaution.


Asunto(s)
Agencias Gubernamentales/organización & administración , Formulación de Políticas , Animales , Humanos , Estados Unidos , United States Environmental Protection Agency
3.
Toxicol Appl Pharmacol ; 274(1): 107-16, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24211725

RESUMEN

Doxorubicin (DOX) is a widely prescribed treatment for a broad scope of cancers, but clinical utility is limited by the cumulative, dose-dependent cardiomyopathy that occurs with repeated administration. DOX-induced cardiotoxicity is associated with the production of reactive oxygen species (ROS) and oxidation of lipids, DNA and proteins. A major cellular defense mechanism against such oxidative stress is activation of the Keap1/Nrf2-antioxidant response element (ARE) signaling pathway, which transcriptionally regulates expression of antioxidant genes such as Nqo1 and Gstp1. In the present study, we address the hypothesis that an initial event associated with DOX-induced oxidative stress is activation of the Keap1/Nrf2-dependent expression of antioxidant genes and that this is regulated through drug-induced changes in redox status of the Keap1 protein. Incubation of H9c2 rat cardiac myoblasts with DOX resulted in a time- and dose-dependent decrease in non-protein sulfhydryl groups. Associated with this was a near 2-fold increase in Nrf2 protein content and enhanced transcription of several of the Nrf2-regulated down-stream genes, including Gstp1, Ugt1a1, and Nqo1; the expression of Nfe2l2 (Nrf2) itself was unaltered. Furthermore, both the redox status and the total amount of Keap1 protein were significantly decreased by DOX, with the loss of Keap1 being due to both inhibited gene expression and increased autophagic, but not proteasomal, degradation. These findings identify the Keap1/Nrf2 pathway as a potentially important initial response to acute DOX-induced oxidative injury, with the primary regulatory events being the oxidation and autophagic degradation of the redox sensor Keap1 protein.


Asunto(s)
Doxorrubicina/toxicidad , Péptidos y Proteínas de Señalización Intracelular/fisiología , Mioblastos Cardíacos/efectos de los fármacos , Mioblastos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Animales , Antibióticos Antineoplásicos/toxicidad , Línea Celular , Relación Dosis-Respuesta a Droga , Proteína 1 Asociada A ECH Tipo Kelch , Oxidación-Reducción/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo
5.
Cell Cycle ; 20(11): 1067-1079, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33978554

RESUMEN

Doxorubicin induces both DNA damage and metabolic interference. How these effects interact to modulate cellular toxicity is not completely understood but important given the widespread use of doxorubicin in cancer treatment. This study tests the hypothesis that cell cycle arrest and survival are affected by distinct mitochondrial activities during doxorubicin exposure.Parental and mutant S. cerevisiae strains deficient in selected genes with mitochondrial function were treated with doxorubicin and assayed for changes in proliferation rates, cell survival and cell cycle arrest kinetics. Mitochondrial DNA content was estimated using quantitative PCR. Mitochondrial function was assessed by measuring oxygen consumption with and without an uncoupler.Parental cells growing in a non-fermentable carbon source medium and mutants lacking mitochondria and grown in glucose medium both show abrupt cell cycle and proliferation arrest during doxorubicin exposure compared to parental cells grown in glucose. Mitochondrial DNA increases during doxorubicin exposure in S. cerevisiae and in human breast cancer cells. Yeast strains deficient in TCA cycle activity or electron transport both show more abrupt cell cycle arrest than parental cells when exposed to doxorubicin. Concurrent treatment with the mitochondrial uncoupler dinitrophenol facilitates cell cycle progression and proliferation during doxorubicin exposure.Doxorubicin exposure induces mitochondrial DNA synthesis with TCA cycle and oxidative phosphorylation activity having opposing effects on cell proliferation, survival and cell cycle kinetics. TCA cycle activity provides biosynthetic substrates to support cell cycle progression and cell proliferation while electron transport and oxidative phosphorylation facilitate cell cycle arrest and possibly increased cytotoxicity.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Doxorrubicina/toxicidad , Mitocondrias/efectos de los fármacos , Saccharomyces cerevisiae/efectos de los fármacos , Ciclo Celular/fisiología , Proliferación Celular/fisiología , ADN Mitocondrial/metabolismo , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/fisiología , Humanos , Células MCF-7 , Mitocondrias/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Saccharomyces cerevisiae/metabolismo
6.
Toxicology ; 464: 152997, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34695511

RESUMEN

Per- and polyfluoroalkyl substances (PFAS) represent a large class of structurally diverse chemicals of increasing public concern, mostly due to their chemical stability and undetermined toxicity profiles. In laboratory animals, adverse effects implicated for certain PFAS, perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS) in particular, include liver toxicity and the associated metabolic dysregulation, immune and thyroid alterations, reproductive toxicity, and selected tumors. The broad commercialization and environmental distribution of PFAS has drawn attention to the need for understanding risks associated with combined exposure to multiple PFAS in complex mixtures. The purpose of this investigation is to determine whether binary combinations of PFAS elicit a molecular response that is either greater than or less than the sum of the individual responses. Exposure of FaO rat hepatoma cells for 24 h to 25 µM-200 µM of the 4- and 8-carbon perfluorocarboxylic acids (PFBA and PFOA) or the 4, 6, and 8-carbon perfluorosulfonic acids (PFBS, PFHxS, and PFOS, respectively) individually caused a dose-dependent increase in PPARα-regulated expression of peroxisomal bifunctional enzyme (Ehhadh). Potency increased with carbon number, with the carboxylates eliciting a greater transcriptional response than the corresponding sulfonates. Combined exposure to PFOA and PFBA produced an effect that was significantly less than the sum of the individual responses. The response to the combination of PFOA and PFOS produced a summative effect at concentrations that were not cytotoxic. Combined exposures to PFOS and either PFBS or PFHxS at low noncytotoxic concentrations produced a transcriptional effect that was significantly less than the sum of the individual effects. The results demonstrate that among the five structurally related perfluoroalkyl acids included in this investigation, PPARα transcriptional activation in response to combined binary exposures is consistently at or below that predicted by the sum of the individual effects.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Caprilatos/toxicidad , Fluorocarburos/toxicidad , PPAR alfa/metabolismo , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/química , Animales , Caprilatos/administración & dosificación , Caprilatos/química , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/química , Contaminantes Ambientales/toxicidad , Fluorocarburos/administración & dosificación , Fluorocarburos/química , Humanos , Neoplasias Hepáticas/metabolismo , Ratas
7.
Cardiovasc Toxicol ; 20(6): 557-570, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32500386

RESUMEN

Doxorubicin (DOX) is a widely prescribed anthracycline antineoplastic drug for treating human solid tumors and leukemias. However, DOX therapy is limited by a cumulative, dose-dependent, and irreversible cardiomyopathy that occurs with repeated administration. Presumably, a pivotal initiating event of DOX-induced cardiotoxicity is the production of reactive oxygen species (ROS) and oxidation of lipids, DNA, and proteins. We recently identified activation of the Keap1/Nrf2-antioxidant response system-a major cellular defense mechanism against such oxidative stress-as an important response to acute DOX exposure in vitro. In the present study, we address the hypothesis that dysregulation of this pathway in cardiac tissue is also manifested in vivo following chronic DOX administration. Male, Sprague-Dawley rats received 6 weekly injections of 2 mg/kg (s.c.) DOX or saline followed by a 5-week drug-free period prior to analysis of cardiac tissue transcripts and proteins. In contrast to in vitro findings, the Keap1/Nrf2-antioxidant response system was suppressed in hearts of DOX-treated animals and consistent with the observed decrease in protein abundance for Nrf2 and PGAM5, both of which are substrates for Keap1. Although this shift in Keap1/Nrf2 suppresses the antioxidant pathway, the concurrent loss of PGAM5 could function as a signal for disposal of damaged mitochondria from the cell, thus removing the source of ROS. These findings identify the Keap1/Nrf2 and Keap1/PGAM5 pathways as important responses to DOX-induced cardiac injury in vivo; disruption of this system for mitochondrial hormesis may be an important contributing factor to cardiotoxicity after chronic drug administration.


Asunto(s)
Cardiopatías/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Mitocondrias Cardíacas/metabolismo , Mitofagia , Miocitos Cardíacos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Cardiotoxicidad , Modelos Animales de Enfermedad , Doxorrubicina , Cardiopatías/inducido químicamente , Cardiopatías/genética , Cardiopatías/patología , Proteína 1 Asociada A ECH Tipo Kelch/genética , Masculino , Mitocondrias Cardíacas/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/patología , Factor 2 Relacionado con NF-E2/genética , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
8.
Trends Pharmacol Sci ; 29(7): 361-6, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18501972

RESUMEN

The bioenergetic features of mitochondria have long been exploited in the design of pharmacological agents suited to accomplish a desired physiological effect; uncoupling of oxidative phosphorylation to induce weight loss, for example. However, more recent experience demonstrates mitochondria to be unintended off targets of other drug therapies and responsible, at least in part, for the dose-limiting adverse events associated with a large array of pharmaceuticals. Review of the fundamentals of mitochondrial molecular biology and bioenergetics reveals a multiplicity of off targets that can be invoked to explain drug-induced mitochondrial failure. It is this redundancy of mitochondrial off targets that complicates identification of discrete mechanisms of toxicity and confounds QSAR-based design of new small molecules devoid of this potential for mitochondrial toxicity. The present review article briefly reviews the molecular biology and biophysics of mitochondrial bioenergetics, which then serves as a platform for identifying the various potential off targets for drug-induced mitochondrial toxicity.


Asunto(s)
Mitocondrias/efectos de los fármacos , Animales , Humanos , Mitocondrias/metabolismo , Biogénesis de Organelos , Oxidación-Reducción , Fosforilación Oxidativa/efectos de los fármacos
9.
Cell Biol Toxicol ; 25(3): 227-43, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18386138

RESUMEN

Doxorubicin (Dox) is a very potent antineoplastic agent used against several types of cancer, despite a cumulative cardiomyopathy that reduces the therapeutic index for treatment. H9c2 myoblast cells have been used as an in vitro model to study biochemical alterations induced by Dox treatment on cardiomyocyte cells. Despite the extensive work already published, few data are available regarding morphological alterations of H9c2 cells during Dox treatment. The purpose of the present work was to evaluate Dox-induced morphological alterations in H9c2 myoblasts, focusing especially on the nuclei, mitochondria, and structural fibrous proteins. Treatment of H9c2 cell with low concentrations of Dox causes alterations in fibrous structural proteins including the nuclear lamina and sarcomeric cardiac myosin, as well as mitochondrial depolarization and fragmentation, membrane blebbing with cell shape changes, and phosphatidylserine externalization. For higher Dox concentrations, more profound alterations are evident, including nuclear swelling with disruption of nuclear membrane structure, mitochondrial swelling, and extensive cytoplasm vacuolization. The results obtained indicate that Dox causes morphological alterations in mitochondrial, nuclear, and fibrous protein structures in H9c2 cells, which are dependent on the drug concentration. Data obtained with the present study allow for a better characterization of the effects of Dox on H9c2 myoblasts, used as a model to study Dox-induced cardiotoxicity. The results obtained also provide new and previously unknown targets that can contribute to understand the mechanisms involved in the cardiotoxicity of Dox.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Núcleo Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Doxorrubicina/toxicidad , Mitocondrias/efectos de los fármacos , Mioblastos/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Miosinas Cardíacas/metabolismo , Caspasas/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Núcleo Celular/patología , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Relación Dosis-Respuesta a Droga , Microscopía Confocal , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Dilatación Mitocondrial/efectos de los fármacos , Mioblastos/metabolismo , Mioblastos/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Proteínas/metabolismo , Ratas , Sarcómeros/efectos de los fármacos , Sarcómeros/metabolismo , Sarcómeros/ultraestructura , Vacuolas/efectos de los fármacos , Vacuolas/patología
10.
Biology (Basel) ; 8(2)2019 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-31083551

RESUMEN

Mitochondrial function declines with age, leading to a variety of age-related diseases (metabolic, central nervous system-related, cancer, etc.) and medication usage increases with age due to the increase in diseases. Drug-induced mitochondrial toxicity has been described for many different drug classes and can lead to liver, muscle, kidney and central nervous system injury and, in rare cases, to death. Many of the most prescribed medications in the geriatric population carry mitochondrial liabilities. We have demonstrated that, over the past decade, each class of drugs that demonstrated mitochondrial toxicity contained drugs with both more and less adverse effects on mitochondria. As patient treatment is often essential, we suggest using medication(s) with the best safety profile and the avoidance of concurrent usage of multiple medications that carry mitochondrial liabilities. In addition, we also recommend lifestyle changes to further improve one's mitochondrial function, such as weight loss, exercise and nutrition.

11.
Food Chem Toxicol ; 124: 450-461, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30557669

RESUMEN

Dose-dependent and cumulative cardiotoxicity associated with doxorubicin (DOX) is the main limitation of anticancer therapy. Pediatric cancer survivors are particularly vulnerable, and no effective prevention measures are available. The aim of the present study was to investigate the persistent effects of nanomolar DOX concentrations and determine whether a pretreatment would induce mitochondrial adaptations in H9c2 cardiomyoblasts. H9c2 cells were incubated with DOX (10 and 25 nM) for 24 h, followed by 9 days of recovery in drug-free medium. We found that the sub-therapeutic DOX treatment induced persistent hypertrophy and dose-dependent cell cycle arrest in G2/M. Glycolytic activity, indirectly based on extracellular acidification rate, and basal respiration were significantly decreased in DOX-treated cells compared to controls, although both groups showed similar maximal respiration. Additionally, nanomolar DOX pretreatment resulted in upregulation of mitochondrial DNA transcripts accompanied by a decrease in DNA methyltransferase 1 (DNMT1) and global methylation levels. Finally, the pretreatment with DOX ameliorated H9c2 cells resistance against a subsequent exposure to DOX. These results suggest that nanomolar DOX pretreatment induced a beneficial and possibly epigenetic-based mitochondrial adaptation, raising the possibility that an early sub-therapeutic DOX treatment can be used as a preconditioning and protective approach during anticancer therapies.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Mitocondrias Cardíacas/efectos de los fármacos , Mioblastos Cardíacos/efectos de los fármacos , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN Mitocondrial/genética , Regulación hacia Abajo/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Expresión Génica/genética , Humanos , Mioblastos Cardíacos/patología , Ratas
12.
Toxicol Sci ; 169(1): 137-150, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30698778

RESUMEN

Doxorubicin (DOX) is an anticancer drug widely used to treat human and nonhuman tumors but the late and persistent cardio-toxicity reduces the therapeutic utility of the drug. The full mechanism(s) of DOX-induced acute, subchronic and delayed toxicity, which has a preponderant mitochondrial component, remains unclear; therefore, it is clinically relevant to identify early markers to identify patients who are predisposed to DOX-related cardiovascular toxicity. To address this, Wistar rats (16 weeks old) were treated with a single DOX dose (20 mg/kg, i.p.); then, mRNA, protein levels and functional analysis of mitochondrial endpoints were assessed 24 h later in the heart, liver, and kidney. Using an exploratory data analysis, we observed cardiac-specific alterations after DOX treatment for mitochondrial complexes III, IV, and preferentially for complex I. Conversely, the same analysis revealed complex II alterations are associated with DOX response in the liver and kidney. Interestingly, H2O2 production by the mitochondrial respiratory chain as well as loss of calcium-loading capacity, markers of subchronic toxicity, were not reliable indicators of acute DOX cardiotoxicity in this animal model. By using sequential principal component analysis and feature correlation analysis, we demonstrated for the first time alterations in sets of transcripts and proteins, but not functional measurements, that might serve as potential early acute markers of cardiac-specific mitochondrial toxicity, contributing to explain the trajectory of DOX cardiac toxicity and to develop novel interventions to minimize DOX cardiac liabilities.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Doxorrubicina/toxicidad , Cardiopatías/inducido químicamente , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Animales , Calcio/metabolismo , Cardiotoxicidad , Respiración de la Célula/efectos de los fármacos , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Cardiopatías/genética , Cardiopatías/metabolismo , Cardiopatías/patología , Peróxido de Hidrógeno/metabolismo , Masculino , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas Wistar , Factores de Tiempo
13.
Toxicol Sci ; 102(1): 3-14, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18003598

RESUMEN

The perfluoroalkyl acid salts (both carboxylates and sulfonates, hereafter designated as PFAAs) and their derivatives are important chemicals that have numerous consumer and industrial applications. However, recent discoveries that some of these compounds have global distribution, environmental persistence, presence in humans and wildlife, as well as toxicity in laboratory animal models, have generated considerable scientific, regulatory, and public interest on an international scale. The Society of Toxicology Contemporary Concepts in Toxicology Symposium, entitled "Perfluoroalkyl Acids and Related Chemistries: Toxicokinetics and Modes-of-Action Workshop" was held February 14-16, 2007 at the Westin Arlington Gateway, Arlington, VA. In addition to the Society of Toxicology, this symposium was sponsored by 3M Company, DuPont, Plastics Europe, and the U.S. Environmental Protection Agency. The objectives of this 3-day meeting were to (1) provide an overview of PFAA toxicity and description of recent findings with the sulfonates, carboxylates, and telomer alcohols; (2) address the toxicokinetic profiles of various PFAAs among animal models and humans, and the biological processes that are responsible for these observations; (3) examine the possible modes of action that determine the PFAA toxicities observed in animal models, and their relevance to human health risks; and (4) identify the critical research needs and strategies to fill the existing informational gaps that hamper risk assessment of these chemicals. This report summarizes the discourse that occurred during the symposium.


Asunto(s)
Ácidos Carboxílicos/toxicidad , Núcleo Celular/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Ácidos Sulfónicos/toxicidad , Pruebas de Toxicidad , Animales , Ácidos Carboxílicos/química , Ácidos Carboxílicos/farmacocinética , Núcleo Celular/metabolismo , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/química , Contaminantes Ambientales/farmacocinética , Fluorocarburos/química , Fluorocarburos/farmacocinética , Humanos , Modelos Moleculares , Estructura Molecular , Receptores Activados del Proliferador del Peroxisoma/efectos de los fármacos , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Reproducibilidad de los Resultados , Medición de Riesgo , Especificidad de la Especie , Relación Estructura-Actividad , Ácidos Sulfónicos/química , Ácidos Sulfónicos/farmacocinética , Pruebas de Toxicidad/métodos
14.
Toxicology ; 251(1-3): 8-20, 2008 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-18692542

RESUMEN

In utero exposure of laboratory rats to perfluorooctane sulfonate (PFOS, C(8)F(17)SO(3)(-)), a chemically stable surfactant that is widely disseminated in the environment and present in serum samples from wildlife and humans, is associated with decreased neonatal survival, and growth deficits as well as hepatomegaly. This hepatomegaly in newborn rats exposed to PFOS in utero resembles that observed in adults and is characterized by peroxisome proliferation and decreased liver triglycerides, both of which are suspected to be manifested through PPARalpha-mediated transcriptional regulation. The purpose of the present investigation was to determine whether these changes in metabolic status are a reflection of transcriptional changes in fetal rat liver using global gene expression array analyses. Gravid Sprague-Dawley rats were administered 3mg/kg PFOS by gavage daily from gestational day 2-20 and terminated on day 21. Although there was no treatment-related frank terata, there was a substantial effect of PFOS on the perinatal hepatic transcriptome-225 unique transcripts were identified as statistically increased and 220 decreased by PFOS exposure; few transcripts were changed by more than two-fold. Although the PPARalpha transcript (Ppara) itself was not affected, there was a significant increase in expression of gene transcripts associated with hepatic peroxisomal proliferation as well as those responsible for fatty acid activation, transport and oxidation pathways (both mitochondrial and peroxisomal). Additional metabolic pathways altered by in utero PFOS exposure were a stimulation of fetal hepatic fatty acid biosynthesis and a net reduction of Cyp7a1 transcript, which is required for bile acid synthesis. There were minimal effects on the expression of thyroid-related gene transcripts. In conclusion, gene expression analysis provides strong evidence indicating transcriptional control of the altered metabolic status of neonates following PFOS exposure in utero, much of which appears to be under the influence of a functional perinatal PPARalpha regulatory pathway.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Contaminantes Ambientales/toxicidad , Fluorocarburos/toxicidad , Expresión Génica/efectos de los fármacos , Hígado , Exposición Materna/efectos adversos , Animales , Ácidos y Sales Biliares/biosíntesis , Ácidos y Sales Biliares/metabolismo , Transporte Biológico , Colesterol/biosíntesis , Ácidos Grasos/biosíntesis , Femenino , Perfilación de la Expresión Génica , Edad Gestacional , Cuerpos Cetónicos/biosíntesis , Hígado/efectos de los fármacos , Hígado/embriología , Hígado/metabolismo , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , PPAR alfa/biosíntesis , Embarazo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Hormonas Tiroideas/biosíntesis , Hormonas Tiroideas/metabolismo
15.
Toxicology ; 243(3): 330-9, 2008 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-18063289

RESUMEN

INTRODUCTION: Perfluorooctanesulfonate (PFOS) is widely distributed and persistent in humans and wildlife. Prior toxicological studies have reported decreased total and free thyroid hormones in serum without a major compensatory rise in thyrotropin (TSH) or altered thyroid gland histology. Although these animals (rats, mice and monkeys) might have maintained an euthyroid state, the basis for hypothyroxinemia remained unclear. We undertook this study to investigate the causes for the PFOS-induced reduction of serum total thyroxine (TT4) in rats. HYPOTHESES: We hypothesized that exposure to PFOS may increase free thyroxine (FT4) in the rat serum due to the ability of PFOS to compete with thyroxine for binding proteins. The increase in FT4 would increase the availability of the thyroid hormone to peripheral tissues for utilization, metabolic conversation, and excretion. We also hypothesized that PFOS does not directly interfere with the regulatory functions of the hypothalamic-pituitary-thyroid (HPT) axis in rats. EXPERIMENTS: Three experimental designs were employed to test these hypotheses. (1) Female Sprague-Dawley (SD) rats were given a single oral dose of 15 mg potassium PFOS/kg body weight. At intervals of 2, 6, and 24h thereafter, measurements were made for serum FT4, TT4, triiodothyronine (TT3), reverse triiodothyronine (rT3), thryrotropin (TSH), and PFOS concentrations, as well as liver PFOS concentrations, UDP-glucuronosyltransferase 1A (UGT1A) family mRNA transcripts, and malic enzyme (ME) mRNA transcripts and activity. (2) To provide evidence for increased uptake and metabolism of thyroxine (T4), 125 I-T4 was given to male and female SD rats by intravenous injection, followed in 2h by a single oral dose of 15 mg potassium PFOS/kg body weight. 125 I radioactivity was determined in urine and feces collected over a 24-h period and in serum and liver collected at 24h. (3) To assess the potentials effect of PFOS on the hypothalamic-pituitary-thyroid axis, over an 8-day period, groups of male SD rats were given PFOS (3mg/kg-d), propyl thiouracil (PTU, 10 microg/mL in water), or PTU and PFOS in combination, with controls receiving 0.5% Tween 20 vehicle. On days 1, 3, 7, and 8, TT4, TT3, and TSH were monitored. On day 8, pituitaries were removed and placed in static culture for assessment of thyrotropin releasing hormone (TRH)-mediated release of TSH. RESULTS: (1) PFOS transiently increased FT4 and decreased TSH within 6h, with values returning to control levels by 24h. TT4 was decreased by 55% over a 24-h period. TT3 and rT3 were decreased at 24h to a lesser extent than TT4. ME mRNA transcripts were increased at 2h and activity was increased at 24h. UGT1A mRNA transcripts were increased at 2 and 6h. (2) 125 I decreased in serum and liver relative to controls and consistent with a reduction in serum TT4. Concomitantly, 125 I activity was increased in urine and feces collected from PFOS-treated rats. (3) During the 8 days of dosing with PFOS, TSH was not elevated in male rats, while TT4 and TT3 were decreased. Pituitary response to TRH-mediated TSH release was not diminished after 8-daily oral doses of PFOS. CONCLUSIONS: These findings suggest that oral dosing in rats with PFOS results in transiently increased tissue availability of the thyroid hormones and turnover of T4 with a resulting reduction in serum TT4. PFOS does not induce a classical hypothyroid state under dosing conditions employed nor does it alter HPT activities.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Fluorocarburos/toxicidad , Hipófisis/efectos de los fármacos , Hormonas Tiroideas/sangre , Administración Oral , Ácidos Alcanesulfónicos/administración & dosificación , Ácidos Alcanesulfónicos/sangre , Animales , Cromatografía Liquida , Relación Dosis-Respuesta a Droga , Heces/química , Femenino , Fluorocarburos/administración & dosificación , Fluorocarburos/sangre , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Radioisótopos de Yodo , Hígado/efectos de los fármacos , Hígado/metabolismo , Mediciones Luminiscentes , Masculino , Espectrometría de Masas , Hipófisis/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Hormona Liberadora de Tirotropina/análisis , Tiroxina/sangre , Factores de Tiempo , Triyodotironina/sangre
16.
Toxicol Pathol ; 36(7): 999-1005, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19126794

RESUMEN

Based on epidemiological data, chronic exposure to high levels of inorganic arsenic in the drinking water is carcinogenic to the urinary bladder of humans. Recently, models have been developed involving transplacental administration of inorganic arsenic and subsequent administration of another substance that produces a low incidence of urogenital neoplasms. Administration of arsenite or arsenate in the diet or drinking water to five-to eight-week-old mice or rats rapidly induces urothelial cytotoxicity and regenerative hyperplasia. In mice administered arsenite, we observed eosinophilic intracytoplasmic granules present in the urothelial cells. These granules were not present in urothelial cells of untreated mice or in treated or untreated rats. By transmission electron microscopy, the granules were located within the mitochondrial matrix, that is, mitochondrial inclusions. Arsenic, primarily as arsenite, was present in partially purified mitochondria containing these granules. Cells containing the granules were not usually associated with degenerative changes. Lack of these granules in rats suggests that they are not necessary for inorganic arsenic-induced urothelial cytotoxicity or hyperplasia. These granules have also been observed with exposures to other metals in other tissues and other species, suggesting that they represent a protective mechanism against metal-induced toxicity.


Asunto(s)
Arsénico/toxicidad , Carcinógenos/toxicidad , Gránulos Citoplasmáticos/ultraestructura , Mitocondrias/ultraestructura , Urotelio/efectos de los fármacos , Animales , Arsénico/administración & dosificación , Arsenicales/análisis , Pruebas de Carcinogenicidad , Carcinógenos/administración & dosificación , Gránulos Citoplasmáticos/efectos de los fármacos , Gránulos Citoplasmáticos/metabolismo , Femenino , Hiperplasia/patología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Animales , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Urotelio/patología , Urotelio/ultraestructura
17.
Toxicol Lett ; 180(2): 85-92, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18588961

RESUMEN

This paper will provide some perspective on the role that a consideration of the dose-response has played (past), is playing (present) and will play (future) in human risk assessment with special emphasis on a number of recent activities undertaken by various components of the International Life Sciences Institute (ILSI). The dose-response is a critically important concept in every aspect of biomedical science, including toxicology. A characterization of the dose response has been recognized as one of the four essential components of risk assessment since the release of the NRC/NAS report in 1983, and understanding the dose-response curve is the basis for regulatory toxicology. The introduction of concepts such as hormesis, thresholds of toxicological concern (TTC), and dose-dependent transitions in mechanisms of toxicity have emphasized the complexities associated with a characterization of the dose-response. The transitions to emphasizing predictive toxicology, systems biology, the new 'omics technologies, and high-throughput screening (HTS) have provided a new vision for toxicity testing. One impact of fully integrating these new concepts and technologies is that we will have unprecedented capabilities to explore the dose-response relationship, especially at low doses. How these new insights into the dose-response will affect our definition of threshold, and our understanding of the distinction between adverse and adaptive effects remain to be determined.


Asunto(s)
Relación Dosis-Respuesta a Droga , Medición de Riesgo , Academias e Institutos , Animales , Humanos , Nivel sin Efectos Adversos Observados , Toxicología/tendencias
18.
Reprod Toxicol ; 78: 150-168, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29694846

RESUMEN

Potassium perfluorohexanesulfonate (K+PFHxS) was evaluated for reproductive/developmental toxicity in CD-1 mice. Up to 3 mg/kg-d K+PFHxS was administered (n = 30/sex/group) before mating, for at least 42 days in F0 males, and for F0 females, through gestation and lactation. F1 pups were directly dosed with K+PFHxS for 14 days after weaning. There was an equivocal decrease in live litter size at 1 and 3 mg/kg-d, but the pup-born-to-implant ratio was unaffected. Adaptive hepatocellular hypertrophy was observed, and in 3 mg/kg-d F0 males, it was accompanied by concomitant decreased serum cholesterol and increased alkaline phosphatase. There were no other toxicologically significant findings on reproductive parameters, hematology/clinical pathology/TSH, neurobehavioral effects, or histopathology. There were no treatment-related effects on postnatal survival, development, or onset of preputial separation or vaginal opening in F1 mice. Consistent with previous studies, our data suggest that the potency of PFHxS is much lower than PFOS in rodents.


Asunto(s)
Efectos Tardíos de la Exposición Prenatal , Ácidos Sulfónicos/toxicidad , Fosfatasa Alcalina/sangre , Animales , Colesterol/sangre , Femenino , Fluorocarburos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Masculino , Intercambio Materno-Fetal , Ratones Endogámicos ICR , Embarazo
19.
BMC Cell Biol ; 8: 11, 2007 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-17362523

RESUMEN

BACKGROUND: When exposed to oxidative conditions, cells suffer not only biochemical alterations, but also morphologic changes. Oxidative stress is a condition induced by some pro-oxidant compounds, such as by tert-butylhydroperoxide (tBHP) and can also be induced in vivo by ischemia/reperfusion conditions, which is very common in cardiac tissue. The cell line H9c2 has been used as an in vitro cellular model for both skeletal and cardiac muscle. Understanding how these cells respond to oxidative agents may furnish novel insights into how cardiac and skeletal tissues respond to oxidative stress conditions. The objective of this work was to characterize, through vital imaging, morphological alterations and the appearance of apoptotic hallmarks, with a special focus on mitochondrial changes, upon exposure of H9c2 cells to tBHP. RESULTS: When exposed to tBHP, an increase in intracellular oxidative stress was detected in H9c2 cells by epifluorescence microscopy, which was accompanied by an increase in cell death that was prevented by the antioxidants Trolox and N-acetylcysteine. Several morphological alterations characteristic of apoptosis were noted, including changes in nuclear morphology, translocation of phosphatidylserine to the outer leaflet of the cell membrane, and cell blebbing. An increase in the exposure period or in tBHP concentration resulted in a clear loss of membrane integrity, which is characteristic of necrosis. Changes in mitochondrial morphology, consisting of a transition from long filaments to small and round fragments, were also detected in H9c2 cells after treatment with tBHP. Bax aggregates near mitochondrial networks were formed after short periods of incubation. CONCLUSION: Vital imaging of alterations in cell morphology is a useful method to characterize cellular responses to oxidative stress. In the present work, we report two distinct patterns of morphological alterations in H9c2 cells exposed to tBHP, a pro-oxidant agent frequently used as model to induce oxidative stress. In particular, dynamic changes in mitochondrial networks could be visualized, which appear to be centrally involved in how these cells respond to oxidative stress. The data also indicate that the cause of H9c2 cell death following tBHP exposure is increased intracellular oxidative stress.


Asunto(s)
Muerte Celular , Mioblastos/efectos de los fármacos , Mioblastos/ultraestructura , terc-Butilhidroperóxido/toxicidad , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Fluoresceínas/metabolismo , Microscopía Fluorescente , Necrosis , Oxidantes/toxicidad , Estrés Oxidativo/efectos de los fármacos , Fosfatidilserinas/metabolismo , Ratas
20.
Cardiovasc Toxicol ; 7(2): 101-7, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17652813

RESUMEN

Adriamycin (doxorubicin) is a potent and broad-spectrum antineoplastic agent, the clinical utility of which is limited by the development of a cumulative and irreversible cardiomyopathy. Although the drug affects numerous structures in different cell types, the mitochondrion appears to a principal subcellular target for the development of cardiomyopathy. This review describes evidence demonstrating that adriamycin redox cycles on complex I of the mitochondrial electron transport chain to liberate highly reactive free radical species of molecular oxygen. The primary effect of adriamycin on mitochondrial performance is the interference with oxidative phosphorylation and inhibition of ATP synthesis. Free radicals liberated from adriamycin redox cycling are thought to be responsible for many of the secondary effects of adriamycin, including lipid peroxidation, the oxidation of both proteins and DNA, and the depletion of glutathione and pyridine nucleotide reducing equivalents in the cell. It is this altered redox status that is believed to cause assorted changes in intracellular regulation, including the induction of the mitochondrial permeability transition and complete loss of mitochondrial integrity and function. Associated with this is the interference with mitochondrial-mediated cell calcium signaling, which is implicated as essential to the capacity of mitochondria to participate in bioenergetic regulation in response to external signals reflecting changes in metabolic demand. If taken to an extreme, this loss of mitochondrial plasticity may manifest in the liberation of signals mediating either oncotic or necrotic cell death, further perpetuating the cardiac failure associated with adriamycin-induced mitochondrial cardiomyopathy.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Calcio/metabolismo , Doxorrubicina/toxicidad , Homeostasis/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Animales , Humanos , Permeabilidad/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA