Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol In Vitro ; 70: 105010, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33022361

RESUMO

Primary mouse hepatocytes isolated from genetically defined and/or diverse lines and disease models are a valuable resource for studying the impact of genetic and environmental factors on drug response and disease. However, standard monolayer cultures result in a rapid decline in mouse hepatocyte viability and functionality. Therefore, we evaluated 3D spheroid methodology for long-term culture of primary mouse hepatocytes, initially to support investigations of drug-induced liver injury (DILI). Primary hepatocytes isolated from male and female C57BL/6J mice were used to generate spheroids by spontaneous self-aggregation in ultra-low attachment plates. Spheroids with well-defined perimeters were observed within 5 days after seeding and retained morphology, ATP, and albumin levels for an additional 2 weeks in culture. Global microarray profiling and quantitative targeted proteomics assessing 10 important drug metabolizing enzymes and transporters demonstrated maintenance of mRNA and protein levels in spheroids over time. Activities for 5 major P450 enzymes were also stable and comparable to activities previously reported for human hepatocyte spheroids. Time- and concentration-dependent decreases in ATP and albumin were observed in response to the DILI-causing drugs acetaminophen, fialuridine, AMG-009, and tolvaptan. Collectively, our results demonstrate successful long-term culture of mouse hepatocytes as spheroids and their utility to support investigations of DILI.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Modelos Biológicos , Acetaminofen/toxicidade , Trifosfato de Adenosina/metabolismo , Albuminas/metabolismo , Animais , Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Feminino , Hepatócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fenilacetatos/toxicidade , Proteômica , Esferoides Celulares/metabolismo , Sulfonamidas/toxicidade , Tolvaptan/toxicidade , Transcriptoma
2.
Arch Toxicol ; 93(4): 1021-1037, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30915487

RESUMO

Drug-induced liver injury remains a frequent reason for drug withdrawal. Accordingly, more predictive and translational models are required to assess human hepatotoxicity risk. This study presents a comprehensive evaluation of two promising models to assess mechanistic hepatotoxicity, microengineered Organ-Chips and 3D hepatic spheroids, which have enhanced liver phenotype, metabolic activity and stability in culture not attainable with conventional 2D models. Sensitivity of the models to two hepatotoxins, acetaminophen (APAP) and fialuridine (FIAU), was assessed across a range of cytotoxicity biomarkers (ATP, albumin, miR-122, α-GST) as well as their metabolic functionality by quantifying APAP, FIAU and CYP probe substrate metabolites. APAP and FIAU produced dose- and time-dependent increases in miR-122 and α-GST release as well as decreases in albumin secretion in both Liver-Chips and hepatic spheroids. Metabolic turnover of CYP probe substrates, APAP and FIAU, was maintained over the 10-day exposure period at concentrations where no cytotoxicity was detected and APAP turnover decreased at concentrations where cytotoxicity was detected. With APAP, the most sensitive biomarkers were albumin in the Liver-Chips (EC50 5.6 mM, day 1) and miR-122 and ATP in the liver spheroids (14-fold and EC50 2.9 mM, respectively, day 3). With FIAU, the most sensitive biomarkers were albumin in the Liver-Chip (EC50 126 µM) and miR-122 (15-fold) in the liver spheroids, both on day 7. In conclusion, both models exhibited integrated toxicity and metabolism, and broadly similar sensitivity to the hepatotoxicants at relevant clinical concentrations, demonstrating the utility of these models for improved hepatotoxicity risk assessment.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Modelos Biológicos , Esferoides Celulares/efeitos dos fármacos , Acetaminofen/toxicidade , Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/toxicidade , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Glutationa Transferase/metabolismo , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Esferoides Celulares/metabolismo
3.
Sci Rep ; 6: 25187, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27143246

RESUMO

Liver biology and function, drug-induced liver injury (DILI) and liver diseases are difficult to study using current in vitro models such as primary human hepatocyte (PHH) monolayer cultures, as their rapid de-differentiation restricts their usefulness substantially. Thus, we have developed and extensively characterized an easily scalable 3D PHH spheroid system in chemically-defined, serum-free conditions. Using whole proteome analyses, we found that PHH spheroids cultured this way were similar to the liver in vivo and even retained their inter-individual variability. Furthermore, PHH spheroids remained phenotypically stable and retained morphology, viability, and hepatocyte-specific functions for culture periods of at least 5 weeks. We show that under chronic exposure, the sensitivity of the hepatocytes drastically increased and toxicity of a set of hepatotoxins was detected at clinically relevant concentrations. An interesting example was the chronic toxicity of fialuridine for which hepatotoxicity was mimicked after repeated-dosing in the PHH spheroid model, not possible to detect using previous in vitro systems. Additionally, we provide proof-of-principle that PHH spheroids can reflect liver pathologies such as cholestasis, steatosis and viral hepatitis. Combined, our results demonstrate that the PHH spheroid system presented here constitutes a versatile and promising in vitro system to study liver function, liver diseases, drug targets and long-term DILI.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/fisiologia , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/fisiologia , Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/toxicidade , Células Cultivadas , Humanos , Modelos Biológicos , Estudo de Prova de Conceito , Proteoma/análise
5.
PLoS Med ; 11(4): e1001628, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24736310

RESUMO

BACKGROUND: Seven of 15 clinical trial participants treated with a nucleoside analogue (fialuridine [FIAU]) developed acute liver failure. Five treated participants died, and two required a liver transplant. Preclinical toxicology studies in mice, rats, dogs, and primates did not provide any indication that FIAU would be hepatotoxic in humans. Therefore, we investigated whether FIAU-induced liver toxicity could be detected in chimeric TK-NOG mice with humanized livers. METHODS AND FINDINGS: Control and chimeric TK-NOG mice with humanized livers were treated orally with FIAU 400, 100, 25, or 2.5 mg/kg/d. The response to drug treatment was evaluated by measuring plasma lactate and liver enzymes, by assessing liver histology, and by electron microscopy. After treatment with FIAU 400 mg/kg/d for 4 d, chimeric mice developed clinical and serologic evidence of liver failure and lactic acidosis. Analysis of liver tissue revealed steatosis in regions with human, but not mouse, hepatocytes. Electron micrographs revealed lipid and mitochondrial abnormalities in the human hepatocytes in FIAU-treated chimeric mice. Dose-dependent liver toxicity was detected in chimeric mice treated with FIAU 100, 25, or 2.5 mg/kg/d for 14 d. Liver toxicity did not develop in control mice that were treated with the same FIAU doses for 14 d. In contrast, treatment with another nucleotide analogue (sofosbuvir 440 or 44 mg/kg/d po) for 14 d, which did not cause liver toxicity in human trial participants, did not cause liver toxicity in mice with humanized livers. CONCLUSIONS: FIAU-induced liver toxicity could be readily detected using chimeric TK-NOG mice with humanized livers, even when the mice were treated with a FIAU dose that was only 10-fold above the dose used in human participants. The clinical features, laboratory abnormalities, liver histology, and ultra-structural changes observed in FIAU-treated chimeric mice mirrored those of FIAU-treated human participants. The use of chimeric mice in preclinical toxicology studies could improve the safety of candidate medications selected for testing in human participants. Please see later in the article for the Editors' Summary.


Assuntos
Antivirais/toxicidade , Arabinofuranosiluracila/análogos & derivados , Falência Hepática Aguda/induzido quimicamente , Fígado/efeitos dos fármacos , Animais , Arabinofuranosiluracila/toxicidade , Quimera , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Falência Hepática Aguda/fisiopatologia , Masculino , Camundongos , Modelos Animais , Testes de Toxicidade
6.
Chemosphere ; 81(2): 253-60, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20624627

RESUMO

In spite of growing scientific concern about pharmaceuticals in the environment, there is still a lack of information especially with regard to their metabolites. The present study investigated ecotoxicity and genotoxicity of three widely used cytostatic agents 5-fluorouracil (5-FU), cytarabine (CYT) and gemcitabine (GemC) and their major human metabolites, i.e. alpha-fluoro-beta-alanine (FBAL), uracil-1-beta-D-arabinofuranoside (AraU) and 2',2'-difluorodeoxyuridine (dFdU), respectively. Effects were studied in acute immobilization and reproduction assays with crustacean Daphnia magna and growth inhibition tests with alga Desmodesmus subspicatus and bacteria Pseudomonas putida. Genotoxicity was tested with umu-test employing Salmonella choleraesius subsp. chol. Toxicity was relatively high at parent compounds with EC(50) values ranging from 44 microg L(-1) (5-fluorouracil in the P. putida test) to 200 mg L(-1) (cytarabine in D. magna acute test). In general, the most toxic compound was 5-FU. Studied metabolites showed low or no toxicity; only FBAL (metabolite of 5-FU) showed low toxicity to D. subspicatus and P. putida with EC(50) values 80 and 140 mg L(-1), respectively. All parent cytostatics showed genotoxicity with minimum genotoxic concentrations (MGC) ranging from 40 to 330 mg L(-1). From metabolites, only FBAL was genotoxic in high concentrations. To our knowledge, the present study provides some of the first ecotoxicity data for both cytostatics and their metabolites, which might further serve for serious evaluation of ecological risks. The observed EC(50) values within the microg L(-1) range were fairly close to concentrations reported in hospital sewage water, which indicates further research needs, especially studies of chronic toxicity.


Assuntos
Antineoplásicos/toxicidade , Citarabina/toxicidade , Desoxicitidina/análogos & derivados , Fluoruracila/toxicidade , Mutagênicos/toxicidade , Animais , Antineoplásicos/química , Arabinofuranosiluracila/química , Arabinofuranosiluracila/toxicidade , Clorófitas/efeitos dos fármacos , Clorófitas/crescimento & desenvolvimento , Citarabina/química , Daphnia/efeitos dos fármacos , Desoxicitidina/química , Desoxicitidina/toxicidade , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Fluoruracila/química , Inibidores do Crescimento/toxicidade , Testes de Mutagenicidade , Pseudomonas/efeitos dos fármacos , Pseudomonas/crescimento & desenvolvimento , Salmonella/efeitos dos fármacos , Salmonella/crescimento & desenvolvimento , Testes de Toxicidade , beta-Alanina/análogos & derivados , beta-Alanina/química , beta-Alanina/toxicidade , Gencitabina
7.
J Biol Chem ; 279(6): 4490-7, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14607828

RESUMO

Many antiviral drugs (e.g. fialuridine; FIAU) produce clinically significant mitochondrial toxicity that limits their dose or prevents their use in the clinic. Because the majority of nucleoside drugs is too hydrophilic to cross the highly impermeable mitochondrial membrane, we have hypothesized that they must be transported into the mitochondria to produce their toxicity. To test this hypothesis, we have sought to determine whether the nucleoside transporters, human equilibrative nucleoside transporter 1 (hENT1) or human concentrative nucleoside transporter 1 (hCNT1), when stably expressed in Madin-Darby canine kidney cells as yellow fluorescent fusion protein (YFP), are localized to the mitochondria. By using organelle-selective dyes and confocal microscopy, we have found that hENT1-YFP is localized to the mitochondria as well as the plasma membrane, whereas hCNT1-YFP was found predominantly on the plasma membrane. hENT1-YFP was not localized to the nuclear envelope, endosomes, lysosomes, or Golgi complex. Western blotting confirmed the presence of hENT1-YFP or endogenous hENT1 in mitochondria isolated from hENT1-YFP-expressing cells and human livers, respectively. In agreement with these localization data, [14C]FIAU was efficiently transported into the mitochondria of cells expressing hENT1-YFP but not of cells expressing hCNT1-YFP. The mitochondrial toxicity of FIAU to Madin-Darby canine kidney cells was enhanced by hENT1-YFP, even when hENT1 activity on the plasma membrane was selectively blocked by 10 nm nitrobenzylthioinosine. Moreover, FIAU (50 microm) produced significant mitochondrial toxicity ( approximately 70% decrease in mitochondrial DNA synthesis) when it was directly incubated with mitochondria isolated from hENT1-expressing cells. In conclusion, we have identified for the first time that hENT1 is expressed on the mitochondrial membrane and that this expression enhances the mitochondrial toxicity of nucleoside drugs such as FIAU. Mitochondrial expression of hENTs may explain the clinically significant mitochondrial toxicity caused by the anti-HIV nucleoside drugs such as zidovudine, stavudine, and didanosine.


Assuntos
Antivirais/toxicidade , Arabinofuranosiluracila/análogos & derivados , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias Hepáticas/metabolismo , Animais , Antivirais/farmacocinética , Arabinofuranosiluracila/farmacocinética , Arabinofuranosiluracila/toxicidade , Transporte Biológico Ativo , Linhagem Celular , Cães , Transportador Equilibrativo 1 de Nucleosídeo/genética , Humanos , Técnicas In Vitro , Membranas Intracelulares/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Frações Subcelulares/metabolismo
8.
Cancer Gene Ther ; 7(2): 215-23, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10770629

RESUMO

The inhibitory effects of (E)-5-(2-bromovinyl)-2'-deoxyuridine (BVDU) and its arabinosyl derivative (E)-5-(2-bromovinyl)-1-beta-D-arabinofuranosyluracil (BVaraU) on the growth of both MDA-MB-435 human breast carcinoma and 9L rat gliosarcoma cells expressing the thymidine kinase (tk)-encoding gene of the Varicella zoster virus (VZV) or the Herpes simplex virus (HSV) were evaluated. In vitro, BVDU and BVaraU effectively killed both cell types expressing VZVtk, with 50% inhibitory concentration values ranging from 0.06 to 0.4 microM, whereas ganciclovir (GCV) lacked activity. On HSVtk+ cells, BVDU had high cytotoxic activity, with 50% inhibitory concentration values that were similar to those of GCV, whereas BVaraU was inactive. In vivo, BVDU applied intraperitoneally caused a 50% tumor growth inhibition in nude mice inoculated subcutaneously with VZVtk+ as well as HSVtk+ mammary tumor cells. In mice and at variance with the in vitro results, BVaraU had very little activity against the VZVtk+ mammary cells; GCV had the highest activity on the HSVtk+ cells, resulting in a 50% eradication of the tumors. With the 9L rat gliosarcoma model, the VZVtk/BVDU system completely failed to inhibit the development of VZVtk+ glioma tumors induced subcutaneously in syngeneic rats, although BVDU had a similar 45-minute half-life in both rats and mice. Factors other than degradation of the prodrug and related to the mode of action of these analogs are possibly involved in the observed discrepancies between the in vitro and in vivo results.


Assuntos
Antineoplásicos/toxicidade , Antivirais/toxicidade , Arabinofuranosiluracila/análogos & derivados , Bromodesoxiuridina/análogos & derivados , Herpesvirus Humano 3/efeitos dos fármacos , Simplexvirus/efeitos dos fármacos , Timidina Quinase/biossíntese , Animais , Antineoplásicos/metabolismo , Antivirais/metabolismo , Arabinofuranosiluracila/toxicidade , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Bromodesoxiuridina/metabolismo , Bromodesoxiuridina/toxicidade , Feminino , Vetores Genéticos , Herpesvirus Humano 3/enzimologia , Herpesvirus Humano 3/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Ratos , Ratos Endogâmicos F344 , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/genética , Células Tumorais Cultivadas
9.
Curr Opin Investig Drugs ; 1(3): 308-13, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11249713

RESUMO

The pyrimidine analog, clevudine (L-FMAU: 2'-fluoro-5-methyl-beta-L-arabinofuranosyluridine) is a potent antihepatitis B virus (HBV) and anti-Epstein-Barr virus (EBV) agent, discovered by researchers at the University of Georgia, in collaboration with Yale University and Bukwang. Bukwang transferred its technology to Triangle Pharmaceuticals in 1998 together with a license to develop clevudine worldwide except Korea [279649], [281942]. In June 1999, Triangle and Abbott Laboratories entered into a strategic alliance to copromote antiviral products including L-FMAU [326798]. In September 2000, Triangle Pharmaceuticals Inc initiated a 30-day phase I/II evaluation of clevudine in HBV-infected patients [381755]. Clevudine is a much less toxic derivative of the toxic agent P-D-FMAU. The mechanism of action of clevudine is not yet clear, but the agent induces a rapid decrease in HBV nucleic acid as doses increase from 0.3 to 10 mg/kg [319145]. It is believed that the target for clevudine lies in the viral replication mechanism. Clevudine is phosphorylated to the triphosphate form intracellularly. This is removed slowly from the cells, thus exerting a sustained inhibitory antiviral activity [178173], [320720], [320721].


Assuntos
Antivirais/farmacologia , Arabinofuranosiluracila/farmacologia , Drogas em Investigação/farmacologia , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Hepatite B/tratamento farmacológico , Animais , Antivirais/efeitos adversos , Antivirais/farmacocinética , Antivirais/uso terapêutico , Antivirais/toxicidade , Arabinofuranosiluracila/efeitos adversos , Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/farmacocinética , Arabinofuranosiluracila/uso terapêutico , Arabinofuranosiluracila/toxicidade , Ensaios Clínicos como Assunto , Contraindicações , Drogas em Investigação/efeitos adversos , Drogas em Investigação/farmacocinética , Drogas em Investigação/uso terapêutico , Drogas em Investigação/toxicidade , Humanos , Relação Estrutura-Atividade
10.
Toxicol Lett ; 102-103: 545-50, 1998 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-10022310

RESUMO

The development of efficacious and safe new human pharmaceuticals continues to be highly dependent on well-designed and carefully executed animal toxicology studies. Although the protocols of most toxicology studies now conform to a standard format, careful selection of the most appropriate and relevant laboratory species is essential. This selection is usually based on pilot toxicity and general pharmacology studies in rodent and non-rodent animals, together with supporting comparative drug disposition studies to determine bioavailability and metabolic profile. A knowledge of the human metabolic profile from early in vitro studies with liver preparations can be particularly valuable.


Assuntos
Toxicologia , Animais , Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/toxicidade , Humanos , Farmacocinética , Especificidade da Espécie
11.
Antivir Ther ; 3(Suppl 3): 113-21, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-10726061

RESUMO

Preclinical aspects of a potent anti-hepatitis B virus (HBV) L-nucleoside, 1-(2-fluoro-5-methyl-beta-L-arabino-furanosyl)uracil (L-FMAU) are described. L-FMAU was prepared from L-ribose derivatives via either L-xylose or L-arabinose. L-FMAU shows potent antiviral activity against hepatitis B virus (EC50 5.0 microM in H1 cells) with high selectivity in vitro. L-FMAU is not incorporated into mitochondrial DNA and no significant lactic acid production was observed in vitro. L-FMAU is phosphorylated by thymidine kinase as well as deoxycytidine kinase, ultimately to the triphosphate, which inhibits HBV DNA polymerase as the mechanism of antiviral action. Preliminary in vivo toxological studies suggest no apparent toxicity for 30 days at 50 mg/kg/day in mice and for 3 months in woodchucks (10 mg/kg/day). L-FMAU also has respectable bioavailability in rats. L-FMAU shows potent anti-HBV activity in vivo against woodchuck hepatitis virus in chronically infected woodchucks and there is no significant virus rebound after cessation of the drug treatment.


Assuntos
Antivirais/farmacologia , Arabinofuranosiluracila/análogos & derivados , Vírus da Hepatite B/efeitos dos fármacos , Animais , Antivirais/toxicidade , Arabinofuranosiluracila/química , Arabinofuranosiluracila/farmacologia , Arabinofuranosiluracila/toxicidade , Disponibilidade Biológica , Linhagem Celular , Desoxicitidina Quinase/metabolismo , Avaliação Pré-Clínica de Medicamentos , Patos , Infecções por Hepadnaviridae/tratamento farmacológico , Infecções por Hepadnaviridae/virologia , Vírus da Hepatite B do Pato/efeitos dos fármacos , Vírus da Hepatite B da Marmota/efeitos dos fármacos , Hepatite B Crônica/tratamento farmacológico , Hepatite B Crônica/virologia , Ácido Láctico/sangue , Ácido Láctico/metabolismo , Dose Letal Mediana , Marmota , Camundongos , Fosforilação , Ratos , Timidina Quinase/metabolismo
12.
Lab Invest ; 76(4): 487-95, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9111511

RESUMO

Fialuridine (FIAU), when used experimentally in the treatment of patients with chronic hepatitis B, caused irreversible acute hepatic failure, myopathy, myoglobinuria, severe lactic acidosis, neuropathy, and death. To investigate the primary cellular elements involved in the neuromuscular toxicity of FIAU, we examined its effects on human myotubes in cultures and searched for signs of recovery. From a total of 75 flasks of normal myotubes prepared from human muscle biopsies, 63 were exposed to various concentrations of FIAU (0.01 microM, 0.1 microM, 1 microM, 10 microM, 50 microM, and 100 microM) for 1 to 3 weeks, whereas 12 served as controls. After 3 weeks of FIAU treatment, 27 flasks were observed for 3 additional weeks to assess spontaneous recovery. All cultures were evaluated with: (a) light microscopy; (b) quantitative immunocytochemistry examining the number of myotubes immunostained for neural-cell adhesion molecule (N-CAM); (c) Oil-Red-O stain, to assess the lipid droplet accumulation; and d) electron microscopy with morphometric measurements of the volumetric density of each organelle per unit volume of tissue (magnification, x24,000). After 3 weeks of FIAU treatment, we found a severe reduction in the number of N-CAM-positive myotubes that varied according to the concentration of FIAU and the duration of treatment. Electron microscopy demonstrated a varying degree of destruction of the myotubes with significant increase in lipid droplets, lysosomes, and the rough endoplasmic reticulum. Major changes in mitochondria were noted even early in the treatment and consisted of concentric lamellar structures, paracrystalline inclusions, and vacuolization. These abnormalities remained unchanged without signs of recovery for up to 3 weeks after withdrawal of FIAU. We conclude that FIAU induces mitochondrial changes and intracellular lipid accumulations similar, but more severe, to those described with the other nucleoside analogues, such as zidovudine. In contrast to zidovudine, however, the FIAU-induced abnormalities do not improve or reverse after withdrawal of the drug. The observations are consistent with the irreversible mitochondrial changes noted in the FIAU-treated patients due to defective mitochondrial DNA replication.


Assuntos
Antivirais/toxicidade , Arabinofuranosiluracila/análogos & derivados , Mitocôndrias Musculares/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Anticorpos Monoclonais/análise , Arabinofuranosiluracila/toxicidade , Células Cultivadas , Humanos , Microscopia Eletrônica , Mitocôndrias Musculares/patologia , Mitocôndrias Musculares/ultraestrutura , Fibras Musculares Esqueléticas/patologia , Fibras Musculares Esqueléticas/ultraestrutura , Músculo Esquelético/citologia , Músculo Esquelético/patologia , Moléculas de Adesão de Célula Nervosa/análise , Organelas/patologia , Organelas/ultraestrutura
13.
Drug Metab Dispos ; 25(2): 270-3, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9029059

RESUMO

Rats were orally co-administered sorivudine (SRV: 1-beta-D-arabinofuranosyl-(E)-5-(2-bromovinyl)uracil), a new oral antiviral drug for herpes zoster, with the oral anticancer drug tegafur (FT: 1-(2-tetrahydrofuryl)-5-fluorouracil) as a prodrug of 5-flourouracil (5-FU) once daily to investigate a toxicokinetic mechanism of 15 Japanese patients' deaths recently caused within a brief period by the drug interaction of these drugs. All the rats showed extremely elevated levels of 5-FU in plasma and tissues, including bone marrow and small intestine, and died within 10 days, whereas the animals given the same dose of SRV or FT alone were still alive over 20 days without any appreciable toxic symptom. Before their death, there was marked damage of bone marrow, marked atrophy of intestinal membrane mucosa, marked decreases in white blood cells and platelets, diarrhea with bloody flux, and severe anorexia as reported with the Japanese patients. Data obtained by in vivo and in vitro studies strongly suggested that (E)-5-(2-bromovinyl)uracil generated from SRV by gut flora was reduced in the presense of NADPH to a reactive form by hepatic dihydropyrimidine dehydrogenase (DPD), a key enzyme determining the tissue 5-FU levels, bound covalently to DPD as a suicide inhibitor, and markedly retarded the catabolism of 5-FU.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Antivirais/toxicidade , Arabinofuranosiluracila/análogos & derivados , Bromouracila/análogos & derivados , Oxirredutases/antagonistas & inibidores , Tegafur/toxicidade , Administração Oral , Animais , Antimetabólitos Antineoplásicos/farmacocinética , Antivirais/farmacocinética , Arabinofuranosiluracila/farmacocinética , Arabinofuranosiluracila/toxicidade , Área Sob a Curva , Doenças da Medula Óssea/induzido quimicamente , Doenças da Medula Óssea/metabolismo , Doenças da Medula Óssea/patologia , Bromouracila/farmacologia , Di-Hidrouracila Desidrogenase (NADP) , Sinergismo Farmacológico , Feminino , Intestino Delgado/metabolismo , Fígado/efeitos dos fármacos , Fígado/enzimologia , Ratos , Ratos Wistar , Tegafur/farmacocinética , Distribuição Tecidual
14.
Lab Invest ; 76(1): 77-87, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9010451

RESUMO

Fialuridine (FIAU, 1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)-5-iodouracil) is toxic to liver, heart, muscle, and nerve in clinical trials for chronic viral hepatitis (CH). Mitochondrial toxicity was hypothesized. To address pathophysiologic mechanisms, we examined mitochondrial changes in FIAU-treated woodchucks (WC) with CH from woodchuck hepatitis virus infection. WC (with and without CH from woodchuck hepatitis virus infection) were treated with FIAU (1.5 mg/kg/day) for 12 weeks. WC were killed. Liver, heart, skeletal muscle, and kidney samples underwent DNA extraction and were analyzed ultrastructurally (transmission electron microscopy). Myocardium, skeletal muscles, and liver samples were analyzed histologically. Abundance of hepatic, myocardial, muscle, and kidney mtDNA decreased in FIAU-treated WC, but the magnitude varied. mtDNA decreased 55% in heart, 65% in kidney, 74% in liver, and 87% in muscle (p < 0.02 for each tissue: FIAU-treated versus FIAU-untreated). Cellular damage was characterized ultrastructurally by mitochondrial enlargement, cristae dissolution, and lipid droplets. Lipid droplets found in the heart, diaphragm, biceps, and liver were sufficient to identify FIAU-treated WC (p < 0.05 each). Widespread mitochondrial damage to many tissues resulted from chronic FIAU treatment and occurred irrespective of CH. It manifested with mtDNA depletion, intracytoplasmic lipid droplets, and destroyed mitochondrial cristae. Defective mtDNA replication with mtDNA depletion seems central to the subcellular pathophysiology of altered energy metabolism and multiorgan failure in FIAU toxicity.


Assuntos
Antivirais/toxicidade , Arabinofuranosiluracila/análogos & derivados , DNA Mitocondrial/efeitos dos fármacos , Vírus da Hepatite B da Marmota , Hepatite B/patologia , Lipídeos/análise , Mitocôndrias/patologia , Animais , Arabinofuranosiluracila/toxicidade , Dano ao DNA , DNA Mitocondrial/metabolismo , Coração/efeitos dos fármacos , Hepatite B/veterinária , Rim/efeitos dos fármacos , Rim/patologia , Rim/ultraestrutura , Fígado/efeitos dos fármacos , Fígado/patologia , Fígado/ultraestrutura , Marmota , Mitocôndrias/efeitos dos fármacos , Mitocôndrias Musculares/efeitos dos fármacos , Mitocôndrias Musculares/patologia , Mitocôndrias Musculares/ultraestrutura , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Miocárdio/patologia , Miocárdio/ultraestrutura
15.
Yakugaku Zasshi ; 117(10-11): 910-21, 1997 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-9414600

RESUMO

In 1993 eighteen Japanese patients with cancer and herpes zoster, a viral disease, died from interactions of the new oral antiviral drug, sorivudine (SRV: 1-beta-D-arabinofuranosyl-(E)-5-(2-bromovinyl)uracil), with oral anticancer prodrugs of 5-fluorouracil (5-FU) within 40 d after SRV was approved by the Japanese government and began to be used clinically. Before the death, most of these patients had severe symptoms of toxicity, including diarrhea with bloody flux and marked decreases in white blood cell and platelet counts. All of these patients received SRV daily for several days while being administered long-term anticancer chemotherapy with one of the oral 5-FU prodrugs. There was no acute toxic symptom in patients who received SRV alone or SRV and the other types of anticancer drugs. A toxicokinetic study was carried out using rats to investigate the mechanism of the acute death in the patients caused by drug interactions between SRV and 5-FU prodrugs. Rats were orally coadministered SRV with tegafur (FT: 1-(2-tetrahydrofuryl)-5-fluorouracil), a 5-FU prodrug that most of the patients were considered to receive before the death. All the rats receiving SRV and FT once daily showed extremely elevated levels of 5-FU in the plasma and tissues, including bone marrow and small intestines, and died within 10 d, while the animals given the same repeated dose of SRV or FT alone were still alive over 20 d without any appreciable toxic symptom. Before their death, there were a marked damage of bone marrow, a marked atrophy of intestinal membrane mucosa, marked decreases in white blood cells and platelets, diarrhea with bloody flux, and severe anorexia as reported for the patients. Data obtained by in vivo and in vitro studies indicated that (E)-5-(2-bromovinyl)uracil (BVU), generated from SRV by the gut flora and absorbed through the intestinal membrane, was reduced in the presence of NADPH to a reactive form by hepatic dihydropyrimidine dehydrogenase (DPD), a key enzyme regulating the tissue 5-FU levels from FT, bound covalently to DPD as a suicide inhibitor, and markedly retarded the catabolism of 5-FU. An irreversible inactivation by BVU of rat and human DPDs, expressed in E. coli for the latter, was observed in the presence of NADPH with their purified preparations in a manner reciprocal to radiolabelling of the enzyme proteins with [14C]BVU. SRV showed no inhibitory effect on the rat and human DPDs in the presence of NADPH.


Assuntos
Antineoplásicos/toxicidade , Antivirais/toxicidade , Arabinofuranosiluracila/análogos & derivados , Fluoruracila/toxicidade , Pró-Fármacos/toxicidade , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Antivirais/administração & dosagem , Antivirais/farmacocinética , Arabinofuranosiluracila/administração & dosagem , Arabinofuranosiluracila/farmacocinética , Arabinofuranosiluracila/toxicidade , Di-Hidrouracila Desidrogenase (NADP) , Interações Medicamentosas , Fluoruracila/administração & dosagem , Fluoruracila/farmacocinética , Humanos , NADP/metabolismo , Oxirredutases/deficiência , Oxirredutases/metabolismo , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Ratos
16.
Antimicrob Agents Chemother ; 40(6): 1555-7, 1996 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8726039

RESUMO

The capacity of recombinant human cytosolic thymidine kinase (TK1) and bovine mitochondrial thymidine kinase (TK2) to phosphorylate the antiviral analogs 1-(2'-deoxy-2'-fluoro-1-beta-D-arabinofuranosyl)-5-iodouracil (FIAU) and 1-(2'-deoxy-2'-fluoro-1-beta-D-arabinofuranosyl)-5-methyluracil (FMAU) has been analyzed. The Vmax/Km ratios for FIAU and FMAU with TK2 are about 30% of that for deoxythymidine, while the corresponding values for TK1 are 2 and 5%, respectively. Thus, these two analogs are more efficient substrates for TK2 than for TK1, which may be part of the explanation for the mitochondrial toxicity associated with FIAU during treatment of hepatitis B infection.


Assuntos
Antivirais/metabolismo , Arabinofuranosiluracila/análogos & derivados , Timidina Quinase/metabolismo , Animais , Antivirais/toxicidade , Arabinofuranosiluracila/metabolismo , Arabinofuranosiluracila/toxicidade , Bovinos , Células Cultivadas , Citosol/enzimologia , Humanos , Mitocôndrias/enzimologia , Fosforilação , Especificidade por Substrato , Timidina Quinase/isolamento & purificação
17.
Proc Natl Acad Sci U S A ; 93(8): 3592-7, 1996 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-8622980

RESUMO

The thymidine analog fialuridine deoxy-2-fluoro-beta-D-arabinofuranosyl)-5-iodouracil (FIAU) was toxic in trials for chronic hepatitis B infection. One mechanism postulated that defective mtDNA replication was mediated through inhibition of DNA polymerase-gamma (DNA pol-gamma), by FIAU triphosphate (FIALTP) or by triphosphates of FIAU metabolites. Inhibition kinetics and primer-extension analyses determined biochemical mechanisms of FIAU, 1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl) -5-methyluracil (FAU), 1-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl)uracil triphosphate (TP) inhibition of DNA pol-gamma. dTMP incorporation by DNA pol-gamma was inhibited competitively by FIAUTP, FMAUTP, and FAUTP (K1=0.015, 0.03, and 1.0 microM, respectively). By using oliginucleotide template-primers. DNA pol-gamma incorporated each analog into DNA opposite a single adenosine efficiently without effects on DNA chain elongation. Incorporation of multiple adjacent analogs at positions of consecutive adenosines dramatically impaired chain elongation by DNA pol-gamma. Effects of FIAU, FMAU, and FAU on HepG2 cell mmtDNA abundance and ultrastructure were determined. After 14 days, mtDNA decreased by 30% with 20 microM FIAU or 20 microM FMAU and decreased less than 10% with 100 microM FAU. FIAU and FMAU disrupted mitochondria and caused accumulation of intracytoplasmic lipid droplets. Biochemical and cell biological findings suggest that FIAU and its metabolites inhibit mtDNA replication, most likely at positions of adenosine tracts, leading to decreased mtDNA and mitochondrial ultrastructural defects.


Assuntos
Antivirais/metabolismo , Antivirais/toxicidade , Arabinofuranosiluracila/análogos & derivados , DNA Polimerase III/antagonistas & inibidores , DNA Mitocondrial/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Arabinofuranosiluracila/metabolismo , Arabinofuranosiluracila/toxicidade , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Primers do DNA/genética , Humanos , Cinética , Microscopia Eletrônica , Mitocôndrias Hepáticas/ultraestrutura , Dados de Sequência Molecular
18.
Nat Med ; 1(5): 417-22, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7585087

RESUMO

Long-term treatment with antiviral nucleoside analogue drugs, such as AZT, can give rise to delayed and at times severe mitochondrial toxicity. Although these toxic effects are manifest in many tissues, a common disease mechanism can explain the diverse clinical events. A better understanding of these disorders will shed light on genetic mitochondrial diseases and lead to the design of safer and more effective antiviral drugs.


Assuntos
Antivirais/toxicidade , Mitocôndrias/efeitos dos fármacos , Síndrome da Imunodeficiência Adquirida/complicações , Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Animais , Antivirais/efeitos adversos , Antivirais/química , Arabinofuranosiluracila/análogos & derivados , Arabinofuranosiluracila/toxicidade , DNA Mitocondrial/efeitos dos fármacos , DNA Polimerase Dirigida por DNA/efeitos dos fármacos , Didanosina/toxicidade , Humanos , Zalcitabina/toxicidade , Zidovudina/toxicidade
19.
J Clin Invest ; 95(2): 555-63, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7860738

RESUMO

We have explored the mechanism(s) related to FIAU-induced liver toxicity, particularly focusing on its effect on mitochondrial function in a human hepatoma cell line-HepG2. The potential role of FMAU and FAU, metabolites detected in FIAU-treated patients were also ascertained. FIAU and FMAU inhibited cell growth and were effectively phosphorylated. A substantial increase in lactic acid production in medium of cells incubated with 1-10 microM FIAU or FMAU was consistent with mitochondrial dysfunction. Slot blot analysis demonstrated that a two week exposure to 10 microM FIAU or FMAU was not associated with a decrease in total mitochondrial (mt) DNA content. However, FIAU and FMAU were incorporated into nuclear and mtDNA and relative values suggest that both compounds incorporate at a much higher rate into mtDNA. Electron micrographs of cells incubated with 10 microM FIAU or FMAU revealed the presence of enlarged mitochondria with higher cristae density and lipid vesicles. In conclusion, these data suggest that despite the lack of inhibition of mtDNA content, incorporation of FIAU and FMAU into mtDNA of HepG2 cells leads to marked mitochondrial dysfunction as evidenced by disturbance in cellular energy metabolism and detection of micro- and macrovesicular steatosis.


Assuntos
Antivirais/toxicidade , Arabinofuranosiluracila/análogos & derivados , Mitocôndrias Hepáticas/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Arabinofuranosiluracila/metabolismo , Arabinofuranosiluracila/toxicidade , Carcinoma Hepatocelular , Divisão Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cromatografia Líquida de Alta Pressão , DNA Mitocondrial/biossíntese , DNA Mitocondrial/efeitos dos fármacos , DNA Mitocondrial/isolamento & purificação , DNA de Neoplasias/biossíntese , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/isolamento & purificação , Humanos , Cinética , Neoplasias Hepáticas , Microscopia Eletrônica , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , Mitocôndrias Hepáticas/patologia , Mitocôndrias Hepáticas/ultraestrutura , Fatores de Tempo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...