Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biomarkers ; 21(5): 387-403, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26983778

RESUMEN

Context The silencing or activation of cancer-associated genes by epigenetic mechanisms can ultimately lead to the clonal expansion of cancer cells. Objective The aim of this review is to summarize all relevant epigenetic biomarkers that have been proposed to date for the diagnosis of some prevalent human cancers. Methods A Medline search for the terms epigenetic biomarkers, human cancers, DNA methylation, histone modifications and microRNAs was performed. Results One hundred fifty-seven relevant publications were found and reviewed. Conclusion To date, a significant number of potential epigenetic cancer biomarkers of human cancer have been investigated, and some have advanced to clinical implementation.


Asunto(s)
Biomarcadores de Tumor/genética , Epigénesis Genética , Genes Relacionados con las Neoplasias/genética , Neoplasias/diagnóstico , Metilación de ADN , Código de Histonas , Humanos , MEDLINE , MicroARNs , Neoplasias/genética
2.
Semin Liver Dis ; 34(2): 205-14, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24879984

RESUMEN

Several drugs have been associated with the potential for drug-induced hepatic steatosis (DIHS) and/or phospholipidosis (DIPL), a lysosomal storage disorder. Drug-induced hepatic steatosis is generally a chronic but reversible affliction and may involve drug accumulation in the liver. Fat accumulation may be either macrovesicular or microvesicular in nature. Commonly used medications associated with DIHS include amiodarone, valproate, tamoxifen, methotrexate, and some chemotherapeutic and antiretroviral agents. Two recently approved medications for the treatment of hereditary homozygous hypercholesterolemia have also been noted to cause hepatic steatosis. For some compounds such as methotrexate and tamoxifen, the underlying metabolic risk factors such as obesity and metabolic syndrome may exacerbate their potential to cause DIHS and its progression. In this article, the authors discuss the preclinical screening and mechanisms of DIHS and DIPL, and review specific examples of drugs commonly used in clinical practice that are known to cause DIHS.


Asunto(s)
Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Enfermedades por Almacenamiento Lisosomal/inducido químicamente , Mitocondrias Hepáticas/metabolismo , Fosfolípidos/metabolismo , Amiodarona/efectos adversos , Animales , Antiarrítmicos/efectos adversos , Antirretrovirales/efectos adversos , Anticolesterolemiantes/efectos adversos , Anticonvulsivantes/efectos adversos , Antineoplásicos Hormonales/efectos adversos , Evaluación Preclínica de Medicamentos , Hígado Graso/patología , Humanos , Inmunosupresores/efectos adversos , Metotrexato/efectos adversos , Mitocondrias Hepáticas/efectos de los fármacos , Tamoxifeno/efectos adversos , Ácido Valproico/efectos adversos
3.
Biomarkers ; 19(7): 541-52, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25189636

RESUMEN

CONTEXT: The definitive standard for the diagnosis of nonalcoholic fatty liver disease (NAFLD) is clinico-pathological correlation, but frequently the only laboratory abnormality is an elevation of serum aminotransferases. OBJECTIVE: This has resulted in the search for more specific laboratory biomarkers. METHODS: The literature was searched for novel plasma/serum markers of NAFLD. RESULTS: Studies reviewed here included histologically-confirmed patients presenting some stage of NAFLD and monitored one or more novel serum/plasma biomarkers. CONCLUSION: The most promising application of some of these novel biomarkers for the detection and quantification of NAFLD and particularly NASH appears to be in the combination of several into diagnostic panels.


Asunto(s)
Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/sangre , Biomarcadores/sangre , Progresión de la Enfermedad , Humanos , Hígado/patología , MicroARNs/sangre , Enfermedad del Hígado Graso no Alcohólico/diagnóstico , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/genética , Valor Predictivo de las Pruebas , Pronóstico , Factores de Riesgo , Índice de Severidad de la Enfermedad
4.
Regul Toxicol Pharmacol ; 68(1): 76-84, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24275050

RESUMEN

Diethanolamine (DEA) has been listed on the State of California's Proposition 65 List. This listing is based in part on tumors reported in a National Toxicology Program (NTP) 2-year dermal carcinogenicity study in mice which found clear evidence of carcinogenic activity in B6C3F1 mice based on increased incidences of liver neoplasms in both sexes, and increased incidences of renal tubule neoplasms in males. Although considerable controversy exists on the relevance of the NTP study to humans, industries are obligated to comply with the Proposition 65 labeling requirement and drinking water discharge prohibition, unless they are able to demonstrate that DEA levels in their products are below a specific No Significant Risk Level (NSRL). The State of California has not published an NSRL for DEA. In this article, a NSRL of 5.6 µg/day and a life-stage-adjusted NSRL(adj) of 1.4 µg/day are derived from the NTP carcinogenicity study using a benchmark dose modeling method based on the incidence of hepatocellular carcinomas in female mice, in accordance with the guidelines of California EPA.


Asunto(s)
Carcinógenos/normas , Etanolaminas/normas , Animales , California , Carcinógenos/farmacocinética , Carcinógenos/toxicidad , Etanolaminas/farmacocinética , Etanolaminas/toxicidad , Femenino , Regulación Gubernamental , Humanos , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/prevención & control , Masculino , Ratones , Pruebas de Mutagenicidad , Ratas , Ratas Endogámicas F344 , Medición de Riesgo/normas , Piel/metabolismo , Absorción Cutánea , Gobierno Estatal
5.
Toxicol Pathol ; 40(3): 435-47, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22301950

RESUMEN

Weak peroxisome proliferator-activated receptor (PPAR) α agonists (fibrates) are used to treat dyslipidemia. This study compared the effects of the potent and selective PPARα agonist CP-778875 on peroxisomal ß-oxidation and cardiac and/or skeletal muscle injury with those of the weak PPARα agonist fenofibrate. We hypothesized that these muscle effects are mediated through the PPARα receptor, leading to increased ß-oxidation and consequent oxidative stress. CP-778875 (5 or 500 mg/kg) and fenofibrate (600 or 2,000→1,200 mg/kg, dose lowered because of intolerance) were administered to rats for six weeks. Standard end points, serum troponin I, heart and skeletal muscle ß-oxidation of palmitoyl-CoA, and acyl co-oxidase (AOX) mRNA were assessed. Both compounds dose-dependently increased the incidence and/or severity of cardiomyocyte degeneration and necrosis, heart weight, troponin I, and skeletal muscle degeneration. Mean heart ß-oxidation (3.4- to 5.1-fold control) and AOX mRNA (2.4- to 3.2-fold control) were increased with CP-778875 500 mg/kg and both doses of fenofibrate. ß-Oxidation of skeletal muscle was not affected by either compound; however, a significant increase in AOX mRNA (1.6- to 2.1-fold control) was observed with CP-778875 500 mg/kg and both doses of fenofibrate. Taken together, these findings were consistent with PPARα agonism and support the link between increased cardiac and skeletal muscle ß-oxidation and resultant muscle injury in the rat.


Asunto(s)
Fenofibrato/toxicidad , Corazón/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , PPAR alfa/agonistas , Animales , Análisis Químico de la Sangre , Peso Corporal , Relación Dosis-Respuesta a Droga , Femenino , Fenofibrato/farmacocinética , Hígado/química , Hígado/efectos de los fármacos , Hígado/enzimología , Hígado/patología , Masculino , Proteínas Musculares/metabolismo , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/inducido químicamente , Enfermedades Musculares/metabolismo , Miocardio/química , Miocardio/metabolismo , Miocardio/patología , NAD/metabolismo , Peroxisomas/metabolismo , Ratas , Ratas Sprague-Dawley , Pruebas de Toxicidad , Troponina I/sangre , Troponina I/metabolismo
6.
Toxicol Pathol ; 40(5): 810-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22504321

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) represent therapeutic targets for the management of type 2 diabetes mellitus and dyslipidemia. Rodent carcinogenicity studies have revealed a link between γ and dual γ/α PPAR agonist treatment and the increased incidence of subcutaneous (SC) liposarcomas/fibrosarcomas or hemangiosarcomas, but very little has been reported for potent and selective PPARα agonists. We present a mode of action framework for the development of SC mesenchymal tumors in rodents given PPAR agonists. (1) Tumor promotion results from pharmacologically mediated recruitment (proliferation and differentiation), thermogenesis and adipogenesis of stromovascular cells, and subsequent generation of oxidative free radicals. (2) Tumor initiation consists of chemotype-driven mitochondrial dysfunction causing uncontrolled oxidative stress and permanent DNA damage. Promotion is characterized by enhanced adipogenesis in the SC adipose tissue, where the baseline PPARγ expression and responsiveness to PPARγ ligands is the highest, and by thermogenesis through expression of the uncoupling protein 1 (UCP-1) and the PPARγ co-activator 1 α (PGC-1α), two factors more highly expressed in brown versus white adipose tissue. Initiation is supported by the demonstration of mitochondrial uncoupling and OXPHOS Complexes dysfunction (Complexes III, IV and V) by compounds associated with increased incidences of sarcomas (muraglitazar and troglitazone), but not others lacking malignant tumor effects (pioglitazone, rosiglitazone).


Asunto(s)
Hipoglucemiantes/toxicidad , PPAR alfa/agonistas , PPAR gamma/agonistas , Sarcoma/inducido químicamente , Adipogénesis/efectos de los fármacos , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Animales , Diferenciación Celular , Cromanos/toxicidad , Daño del ADN/efectos de los fármacos , Diabetes Mellitus Tipo 2/fisiopatología , Diabetes Mellitus Tipo 2/terapia , Glicina/análogos & derivados , Glicina/toxicidad , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Oxazoles/toxicidad , Estrés Oxidativo/efectos de los fármacos , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Pioglitazona , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ratas , Roedores/metabolismo , Rosiglitazona , Sarcoma/patología , Termogénesis/efectos de los fármacos , Tiazolidinedionas/toxicidad , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Troglitazona , Proteína Desacopladora 1
7.
Toxicol Appl Pharmacol ; 245(1): 134-42, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20219512

RESUMEN

Biomarkers are biometric measurements that provide critical quantitative information about the biological condition of the animal or individual being tested. In drug safety studies, established toxicity biomarkers are used along with other conventional study data to determine dose-limiting organ toxicity, and to define species sensitivity for new chemical entities intended for possible use as human medicines. A continuing goal of drug safety scientists in the pharmaceutical industry is to discover and develop better trans-species biomarkers that can be used to determine target organ toxicities for preclinical species in short-term studies at dose levels that are some multiple of the intended human dose and again later in full development for monitoring clinical trials at lower therapeutic doses. Of particular value are early, predictive, noninvasive biomarkers that have in vitro, in vivo, and clinical transferability. Such translational biomarkers bridge animal testing used in preclinical science and human studies that are part of subsequent clinical testing. Although suitable for in vivo preclinical regulatory studies, conventional hepatic safety biomarkers are basically confirmatory markers because they signal organ toxicity after some pathological damage has occurred, and are therefore not well-suited for short-term, predictive screening assays early in the discovery-to-development progression of new chemical entities (NCEs) available in limited quantities. Efforts between regulatory agencies and the pharmaceutical industry are underway for the coordinated discovery, qualification, verification and validation of early predictive toxicity biomarkers. Early predictive safety biomarkers are those that are detectable and quantifiable prior to the onset of irreversible tissue injury and which are associated with a mechanism of action relevant to a specific type of potential hepatic injury. Potential drug toxicity biomarkers are typically endogenous macromolecules in biological fluids with varying immunoreactivity which can present bioanalytical challenges when first discovered. The potential success of these efforts is greatly enhanced by recent advances in two closely linked technologies, toxicoproteomics and targeted, quantitative mass spectrometry. This review focuses on the examination of the current status of these technologies as they relate to the discovery and development of novel preclinical biomarkers of hepatotoxicity. A critical assessment of the current literature reveals two distinct lines of safety biomarker investigation, (1) peripheral fluid biomarkers of organ toxicity and (2) tissue or cell-based toxicity signatures. Improved peripheral fluid biomarkers should allow the sensitive detection of potential organ toxicity prior to the onset of overt organ pathology. Advancements in tissue or cell-based toxicity biomarkers will provide sensitive in vitro or ex vivo screening systems based on toxicity pathway markers. An examination of the current practices in clinical pathology and the critical evaluation of some recently proposed biomarker candidates in comparison to the desired characteristics of an ideal toxicity biomarker lead this author to conclude that a combination of selected biomarkers will be more informative if not predictive of potential animal organ toxicity than any single biomarker, new or old. For the practical assessment of combinations of conventional and/or novel toxicity biomarkers in rodent and large animal preclinical species, mass spectrometry has emerged as the premier analytical tool compared to specific immunoassays or functional assays. Selected and multiple reaction monitoring mass spectrometry applications make it possible for this same basic technology to be used in the progressive stages of biomarker discovery, development, and more importantly, routine study applications without the use of specific antibody reagents. This technology combined with other "omics" technologies can provide added selectivity and sensitivity in preclinical drug safety testing.


Asunto(s)
Biomarcadores/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Proteoma/metabolismo , Pruebas de Toxicidad/métodos , Animales , Biomarcadores/química , Espectrometría de Masas , Pruebas de Toxicidad/instrumentación
8.
Toxicol Mech Methods ; 20(4): 159-66, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20218941

RESUMEN

The induction of hepatic cytochrome P450 (CYP) enzymes, conjugating enzymes, and drug transporters involved in the phase I-III metabolism of xenobiotics is frequently encountered in pre-clinical drug safety studies. As xenobiotics, new drug entities can serve as ligands to three major nuclear receptors; the aryl hydrocarbon receptor (AhR), the constitutive androstane receptor (CAR), and the pregnane X receptor (PXR). These act as xenosensors that often coordinate gene expression with several other nuclear receptors normally involved in endobiotic metabolism. A subsequent gene activation cascade can result in altered liver weights and histopathology and, in some cases, reduced therapeutic efficacy if the drug under test is also a substrate for the induced metabolic enzymes. In humans, CYP induction can result in therapeutic failure for autoinducers or drug-drug interactions if the pharmacokinetic and pharmacodynamic properties of co-administered drugs are altered because they are substrates for the induced enzymes. In addition to CYP gene expression, nuclear receptor proteins regulate the expression of complex gene networks, and therefore mediate the metabolism and modify the effects of steroid hormones, fat-soluble vitamins, and free fatty acids on the metabolic, reproductive, and developmental processes of mammals. CAR and PXR also regulate hepatic energy metabolism through cross-talk with insulin- or glucagon-responsive transcription factors. This review examines the perturbation of these endogenous regulatory systems by xenobiotic CYP inducers, which have potential pathophysiological consequences ranging from alterations in the biological clock to adverse effects on the cardiovascular system of pre-clinical species.


Asunto(s)
Sistema Enzimático del Citocromo P-450/biosíntesis , Xenobióticos/farmacología , Inducción Enzimática
9.
Drug Chem Toxicol ; 31(4): 427-45, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18850354

RESUMEN

Dexamethasone (DEXA) administration has been associated with serum alanine aminotransferase (ALT) elevations that may result from enhanced ALT expression. The aim of our current study was to compare liver vs. serum ALT activity and to examine the onset of any hepatocellular changes. Groups of 4 male Sprague-Dawley rats were administered a single dose of DEXA or corn oil at 12, 16, and 24 h prior to euthanasia or once-daily for 2, 3, or 4 days. All (nonfasted) rats were necropsied together on Day 5. While DEXA incrementally increased liver ALT activity in the 1-, 2-, 3-, and 4-day treatment groups (maximal, 3.7-fold), liver aspartate aminotransferase (AST) never exceeded 1.4-fold over control. Significant hepatic glycogen elevations were detected after DEXA treatment, which correlated with microscopic observations. Serum ALT, AST, sorbitol dehydrogenase, and glutamate dehydrogenase (GLDH) increased after 2, 3, and 4 days of DEXA dosing (1.3-10.3-fold). DEXA-related necropsy findings included pale livers consistent with glycogen deposition. The relative percent liver to body weight was elevated in all DEXA-treated rats. Hepatocellular necrosis was observed in 1/4 rats at 12 h, 2/4 rats at 2 days, 4/4 rats at 3 days, and 3/4 rats at 4 days. DEXA treatment <2 days failed to produce consistent evidence of hepatic injury, as detected by serum biomarkers and pathology assessment. However, early DEXA treatment did correlate with apparent ALT induction. Ultimately, this may explain some early asymptomatic serum ALT elevations seen clinically.


Asunto(s)
Alanina Transaminasa/metabolismo , Dexametasona/administración & dosificación , Glucocorticoides/administración & dosificación , Hígado/efectos de los fármacos , Alanina Transaminasa/sangre , Animales , Biomarcadores/metabolismo , Glucógeno/metabolismo , Hígado/enzimología , Hígado/patología , Masculino , Necrosis , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Regulación hacia Arriba
10.
Curr Drug Metab ; 7(3): 219-29, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16611018

RESUMEN

Severe adverse drug responses are infrequent but occasionally serious events that are not readily predictable at the preclinical development level using only non-human or in vitro models. A common characteristic of the more serious toxicities is generation of short-lived and highly reactive electrophilic species in some individuals. The objective here is to review the literature for toxicological mechanisms that underlie known adverse drug reactions and then categorize the biological consequences of reactive chemical intermediates and radicals in terms of human risk factors and known metabolic variables. Xenobiotics described as being associated with rare but potentially serious adverse events affecting liver, skin, or causing blood dyscrasias tend to have three of four essential characteristics, (1) they are capable of forming short-lived reactive intermediates (RI) or free radicals in target tissues under ideal conditions that are distinct from primary metabolic products, (2) these RI escape/overwhelm the detoxification mechanisms associated with the site of origin or form toxic conjugates, (3) the unconjugated RI must either selectively damage critical proteins or other key macromolecules or (4) the RI acts as a hapten and stimulates an immunological (hypersensitivity) response or overcomes tolerance. Some risk factors may increase the probability of susceptibility, but this remains an active area of research. Because of the complexity of the pathogenesis of some injuries and the role of individual factors, no highly predictive in vitro screening methods are available; however, several methods are evolving that may be used to reveal mechanisms of action when a serious adverse effect is encountered.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Animales , Antioxidantes/metabolismo , Biotransformación , Radicales Libres/efectos adversos , Radicales Libres/química , Humanos , Preparaciones Farmacéuticas/química , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo , Toxicogenética
11.
Toxicol In Vitro ; 20(8): 1488-99, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16782301

RESUMEN

Xenobiotic-inflammation interactions lead to hepatotoxicity in vivo. Selected xenobiotic agents (acetaminophen, APAP; chlorpromazine, CPZ; allyl alcohol, AlOH; monocrotaline, MCT) for which this occurs were evaluated for ability to elicit the release of Kupffer cell (KC)-derived inflammatory mediators and to modulate lipopolysaccharide (LPS)-stimulated release of these mediators. Using KCs and hepatocytes (HPCs) isolated from rat, KC/HPC cocultures were treated with either LPS, xenobiotic, vehicle or a combination. Six hours later, the release of inflammatory mediators was assessed. LPS alone caused a concentration-dependent increase in TNF-alpha release but had no significant effect on the release of PGE(2). APAP by itself did not alter release of TNF-alpha, PGE(2), IL-10, Gro/KC or IFN-gamma; however, in the presence of LPS, APAP enhanced LPS-induced TNF-alpha and Gro/KC release. APAP also attenuated LPS-induced increases in IL-10 and MCP-1. CPZ alone caused a concentration-dependent increase in TNF-alpha release, which was approximately additive in the presence of LPS. AlOH alone did not affect TNF-alpha release, but decreased TNF-alpha production in the presence of LPS. AlOH increased PGE(2) production, and this effect was potentiated in the presence of LPS. MCT by itself did not affect release of TNF-alpha but increased the response to LPS. Neither MCT, LPS, nor the combination affected production of PGE(2). These results demonstrate that KC/HPC cocultures can be used to evaluate interactions of xenobiotics with LPS. Furthermore, data from these studies qualitatively mirror reported data from whole animal studies, suggesting that this model could be useful for predicting aspects of xenobiotic-inflammation interactions in vivo.


Asunto(s)
Hepatocitos/efectos de los fármacos , Inflamación/patología , Macrófagos del Hígado/efectos de los fármacos , Acetaminofén/toxicidad , Analgésicos no Narcóticos/toxicidad , Animales , Antipsicóticos/toxicidad , Carcinógenos/toxicidad , Separación Celular , Clorpromazina/toxicidad , Técnicas de Cocultivo , Interacciones Farmacológicas , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoensayo , Inmunohistoquímica , Lipopolisacáridos/farmacología , Masculino , Monocrotalina/toxicidad , Cloruro de Potasio/farmacología , Propanoles/toxicidad , Ratas , Ratas Sprague-Dawley
12.
Food Chem Toxicol ; 44(4): 528-37, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16274908

RESUMEN

The purpose of this investigation was to examine the relationship among hepatic microsomal enzyme induction, liver weight, histological evidence of hepatic injury, and serum clinical chemistry markers of hepatic origin in the cynomolgus monkey. We report here the results from independent toxicology studies for 10 investigative drug candidates representing four therapeutic classes. Study conditions were selected to elicit target organ toxicity. We found that six of the 10 compounds altered cytochrome P450-associated activities in both male and female monkeys, two in females only, and one altered similar activities in males only. Frequently, significant treatment-related elevations in NADPH cytochrome c reductase and ethylmorphine N-demethylase were noted. When the results from all 10 studies were pooled, 14 cytochrome P450-associated activities were significantly increased and five were decreased in males compared to 15 significantly increased and three decreased in the females. Treatment-associated liver weight increases were noted in four studies. Except for hepatocellular hypertrophy in one study, no significant treatment-related microscopic changes in liver and no elevations of serum biomarkers commonly associated with liver toxicity were observed in any of the studies that demonstrated significant hepatic enzyme induction. Compared to parallel rat studies, one compound was an inducer only in monkeys and one was an inducer only in rats. Significant elevations of microsomal drug-metabolizing enzymes in the cynomolgus monkey liver are not accompanied by substantial hepatic changes except for hepatomegaly. These alterations in the hepatic drug-metabolizing enzyme system were benign based the absence of histopathological lesions and serum biomarkers of hepatobiliary toxicity.


Asunto(s)
Sistema Enzimático del Citocromo P-450/biosíntesis , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Animales , Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Inducción Enzimática , Femenino , Haplorrinos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Nivel sin Efectos Adversos Observados , Ratas , Especificidad de la Especie , Pruebas de Toxicidad
13.
Toxicol Mech Methods ; 16(7): 385-94, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-20021012

RESUMEN

The assessment of hepatic microsomal enzyme induction at the completion of preclinical toxicology studies in rodents and large mammals provides a wealth of information to the toxicologist and pharmacokineticist regarding how the drug-metabolizing system of the hepatocyte endoplasmic reticulum responded to high-dose levels of a xenobiotic designed for a specific pharmacological target in any of several target organs. The interpretation of these data can be greatly enhanced by a clear understanding of how this system functions and what the immediate and long-term ramifications are to organs and organ systems. This review focuses on how drugs modify the hepatic cytochrome P450 system, how those modifications are detected, the various consequences of these modifications, and some differences in the induction response among species.

14.
Expert Opin Drug Metab Toxicol ; 12(12): 1463-1477, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27548410

RESUMEN

INTRODUCTION: If a drug is found to be an inducer of hepatic drug metabolizing enzymes via activation of nuclear receptors such as pregnane X receptor (PXR) or constitutive androstane receptor (CAR), it is likely that drug transporters regulated through these same receptors will be induced as well. This review highlights what is currently known about the molecular mechanisms that regulate transporter expression and where the research is directed. Areas covered: This review is focused on publications that describe the role of activated hepatic nuclear receptors in the subsequent regulation of drug uptake and/or efflux transporters following exposure to xenobiotics. Expert opinion: Many of the published studies on the role of nuclear receptors in the regulation of drug transporters involve non-human test animals. But due to species response differences, these associations are not always applicable to humans. For this reason, some relevant human in vitro models have been developed, such as primary or cryopreserved human hepatocytes, human liver slices, or HepG2 or HuH7 cell lines transiently or stably transfected with PXR expression and reporter constructs as well as in vivo models such as PXR-humanized mice. These human-relevant test systems will continue to be developed and applied for the testing of investigational drugs.


Asunto(s)
Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Xenobióticos/metabolismo , Animales , Transporte Biológico/fisiología , Línea Celular , Receptor de Androstano Constitutivo , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ratones , Receptor X de Pregnano , Receptores de Esteroides/metabolismo , Especificidad de la Especie
15.
Curr Med Chem ; 12(16): 1829-39, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16101504

RESUMEN

Mitochondrial dysfunction is a fundamental mechanism in the pathogenesis of several significant toxicities in mammals, especially those associated with the liver, skeletal and cardiac muscle, and the central nervous system. These changes can also occur as part of the natural aging process and have been linked to cellular mechanisms in several human disease states including Parkinson's and Alzheimer's, as well as ischemic perfusion injury and the effects of hyperglycemia in diabetes mellitus. Our knowledge of the effects of xenobiotics on mitochondrial function has expanded to the point that chemical structure and properties can guide the pharmaceutical scientist in anticipating mitochondrial toxicity. Recognition that maintenance of the mitochondrial membrane potential is essential for normal mitochondrial function has resulted in the development of predictive cell-based or isolated mitochondrial assay systems for detecting these effects with new chemical entities. The homeostatic role of some uncoupling proteins, differences in mitochondrial sensitivity to toxicity, and the pivotal role of mitochondrial permeability transition (MPT) as the determinant of apoptotic cell death are factors that underlie the adverse effects of some drugs in mammalian systems. In order to preserve mitochondrial integrity in potential target organs during therapeutic regimens, a basic understanding of mitochondrial function and its monitoring in the drug development program are essential. Toward this end, this review focuses on two topics, (1) the specific effects of xenobiotics on mitochondrial structure and function and (2) a summarization of current methods for quantifying these changes in a preclinical toxicology laboratory.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Animales , Humanos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/fisiología
16.
Toxicology ; 172(3): 217-30, 2002 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-11893420

RESUMEN

The nonsteroidal anti-inflammatory drug (NSAID) diclofenac (DF) is associated with idiosyncratic hepatotoxicity and several other distinct hypersensitivity reactions. The mechanism(s) are unknown but evidence suggests both cell-mediated and antibody-mediated immune effector systems may be involved. In the present studies, the immunostimulating potential of DF was evaluated using the direct and TNP-Ficoll (trinitrophenyl (TNP)-Ficoll) popliteal lymph node assays (PLNA). These assays were conducted in naive mice, T-cell-deficient mice, or in mice that had been pretreated with a single oral dose of DF. In naive mice, DF induced a dose-, and time-dependent reaction in the direct PLNA. A significant increase in popliteal lymph node (PLN) weight and PLN cellularity was detected 7 days after the injection of 0.50 and 0.75 mg DF, whereas 0.25 mg DF produced no observable effect. With 0.75 mg, there was a rapid accumulation of cells in the PLN between days 5 and 6, with maximum PLN cellularity observed between days 7 and 10. The immunostimulating effects of DF were significantly attenuated in T-cell-deficient mice. In the TNP-Ficoll PLNA conducted in naive mice, DF caused a dose-dependent increase in PLN cellularity on day 7 with a time-dependent increase in anti-TNP antibody forming cells (AFCs) in the PLN; the reaction was dominated by IgM anti-TNP AFCs from day 4 through day 7, but IgG1 anti-TNP AFCs and IgG3 anti-TNP AFCs were detected beginning on day 5 and day 6, respectively. Relative to mice pretreated with vehicle (ddH2O), mice orally pretreated with DF had a significantly greater increase in PLN weight 5 days following the injection of 0.25 mg DF and a significantly greater increase in PLN weight and cellularity 4 days following the injection of 0.50 mg DF. Oral pretreatment with DF had no observable effect on the direct PLN reaction induced following the footpad injection of the irrelevant drugs, D-penicillamine (D-PEN) or streptozotocin. When 0.50 mg DF was co-injected with TNP-Ficoll, mice orally pretreated with DF, compared to vehicle-pretreated mice, and had a significantly greater increase in IgM anti-TNP AFCs on day 4, and a significant increase in both IgG1 and IgG3 anti-TNP AFCs on day 7. Additionally, IgG1 anti-TNP AFCs were detected in the PLN of DF-pretreated mice as early as day 4. No differences in anti-TNP AFCs were detected when orally pretreated mice were injected with 0.50 mg D-PEN. Collectively, these results demonstrated that DF (i) is an immunostimulating drug that induced a dose-, time- and T-cell-dependent PLN reaction in naive mice, (ii) provided non-cognate help that produced antibody against co-injected TNP-Ficoll, and (iii) mice orally pretreated with DF had DF-specific increased responsiveness in the direct PLNA, which (iv) resulted in accelerated and augmented AFC production against co-injected TNP-Ficoll. These novel findings suggest that oral administration of DF may result in primed T cells that respond with footpad injection. Thus, the oral pretreatment modification of the PLNA should be further explored as a possible alternative to hypersensitivity testing with drugs administered via the oral route. Additional studies with other compounds known to produce hypersensitivity reactions are needed.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Diclofenaco/farmacología , Ficoll/análogos & derivados , Inmunidad Celular/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Fosfatasa Alcalina/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Edema/inducido químicamente , Edema/prevención & control , Femenino , Ficoll/inmunología , Inmunoglobulina G/biosíntesis , Inmunoglobulina M/biosíntesis , Indicadores y Reactivos , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos BALB C , Tamaño de los Órganos/efectos de los fármacos , Factores de Tiempo
17.
Toxicol Lett ; 131(3): 167-80, 2002 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-11992736

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) are frequently associated with immune-mediated hypersensitivity reactions. The NSAID diclofenac is associated with several distinct allergic and autoimmune-like reactions including anaphylaxis, idiosyncratic hepatotoxicity and autoimmune hemolytic anemia. The aim of this study was to examine the immunostimulating potential of diclofenac in the direct popliteal lymph node assay (PLNA) and reporter antigen PLNA. In BALB/c mice, diclofenac caused dose-dependent increases in PLN weight and PLN cellularity in the direct PLNA; 0.25 mg was non-immunostimulating whereas 0.50-1.00 mg caused a significant PLN reaction. In the direct PLNA, diclofenac also increased the percent of T cells in the PLN with activated phenotypes (CD44(high)CD62L(low) and CD44(high)CD62L(high)). Finally, the magnitude of the diclofenac-induced direct PLN reaction was significantly reduced when the assay was conducted in T-cell-deficient mice. When co-injected with the reporter antigen TNP-Ficoll (trinitrophenyl Ficoll), 0.50 mg diclofenac caused significant increases in PLN weight, PLN cellularity, and induced IgM and IgG(1) anti-TNP antibody forming cells (AFCs) in the PLN. In a final set of studies, a TNP-OVA PLNA was conducted using diclofenac, phenobarbital (negative control) and streptozotocin (positive control). As expected, phenobarbital (1.00 mg) failed to cause an increase in PLN cellularity or induce AFCs in the PLN. Streptozotocin (1.00 mg) caused significant increases in PLN cellularity, IgM AFCs, and selectively induced IgG(2a) and IgG(2b) AFCs against TNP-OVA. Likewise, diclofenac caused dose-dependent increases (0.25-1.00 mg) in PLN cellularity and IgM AFCs. However, in contrast to streptozotocin, diclofenac caused a selective dose-dependent increase in both IgG(1) and IgE AFCs. Finally, an increase in the intracellular level of IL-4, but not INFgamma, was detected in CD4(+) PLN cells following the injection of diclofenac mixed with TNP-OVA. Collectively, these data suggest that diclofenac: (i) induces a T-cell-dependent direct PLN reaction that; (ii) provides non-cognate help for IgG AFC production when co-injected with TNP-Ficoll, possibly through the formation of neo-antigens; and (iii) possesses intrinsic adjuvant activity that selectively induces IL-4 mediated production of IgG(1) and IgE against co-injected TNP-OVA.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Diclofenaco/farmacología , Inmunoglobulina E/biosíntesis , Inmunoglobulina G/biosíntesis , Ganglios Linfáticos/citología , Activación de Linfocitos/efectos de los fármacos , Ovalbúmina/inmunología , Linfocitos T/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Animales , Diabetes Mellitus Experimental/metabolismo , Femenino , Citometría de Flujo , Interleucina-4/inmunología , Ganglios Linfáticos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Fenotipo
18.
Hum Exp Toxicol ; 33(9): 928-39, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24299907

RESUMEN

Adverse drug reactions (ADRs) can involve all tissues and organs, but liver injuries are considered among the most serious. A number of prospective, multicenter studies have confirmed a higher risk of ADRs in general among female subjects compared to a male cohort. Although drug-induced liver injury (DILI) is infrequently encountered, the preponderance of evidence suggests that women appear to be more susceptible than men to fulminate hepatic/acute liver failure especially in response to some anti-infective drugs and to autoimmune-like hepatitis following exposure to certain other therapeutic drugs. A number of hypotheses have been proposed to explain this sex difference in susceptibility to DILI. Collectively, these hypotheses suggest three basic sex-dependent mechanisms that include differences in various aspects of drug pharmacokinetics (PK) or pharmacodynamics following the administration of certain drugs; specific hormonal effects or interactions with immunomodulating agents or signaling molecules; and differences in the adverse response of the immune system to some drugs, reactive drug metabolites, or drug-protein adducts. At the preclinical drug safety stage, there is a need for more research on hormonal effects on drug PK and for additional research on gender differences in aberrant immune responses that may lead to idiosyncratic DILI in some female patients. Because the detection of rare but serious hepatic ADRs requires the exposure of very large patient populations, pharmacovigilance networks will continue to play a key role in the postmarketing surveillance for their detection and reporting.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Femenino , Humanos , Hígado/efectos de los fármacos , Masculino , Factores de Riesgo , Factores Sexuales
19.
Genome Med ; 5(9): 85, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24073687

RESUMEN

Biomarkers of drug-induced liver injury (DILI) are essential for the diagnosis of severe cases of DILI in clinical trials and clinical practice, but the currently used biomarker paradigm detects damage after it has occurred and has limited prognostic value. The development of new biomarker strategies that improve the diagnosis of DILI by providing increased specificity and/or by identifying individual patients who are at risk for DILI is therefore crucial. See related Research, http://genomemedicine.com/content/5/9/86.

20.
Mol Diagn Ther ; 17(6): 343-54, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23868512

RESUMEN

Drug-induced liver injury (DILI) remains a rare but serious complication in drug therapy that is a primary cause of drug failure during clinical trials. Conventional biomarkers, particularly the serum transaminases and bilirubin, serve as useful indicators of hepatocellular or cholestatic liver injury, respectively, but only after substantial and sometimes irreversible tissue damage. Ideally, more sensitive biomarkers that respond very early before irreversible injury has occurred would offer improved outcomes. Novel biomarkers are initially being developed in animal models exposed to intrinsically hepatotoxic stimuli. However, the eventual translation to human populations, even those with known risk factors that predispose the liver to drug toxicity, would be the fundamental goal. Ultimately, some might even be applicable for the early identification of individuals predisposed to idiosyncratic hepatotoxicity potential. This article reviews recent progress in the discovery and qualification of novel biomarkers for DILI and delineates the path to eventual utilization for risk assessment. Some major categories of plasma or serum biomarkers surveyed include proteins, cytokines, circulating mRNAs, and microRNAs.


Asunto(s)
Biomarcadores/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Hígado/metabolismo , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Citocinas/sangre , Bases de Datos Bibliográficas , Modelos Animales de Enfermedad , Humanos , Metabolómica , MicroARNs/sangre , MicroARNs/metabolismo , ARN Mensajero/sangre , ARN Mensajero/metabolismo , Medición de Riesgo , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA