Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Food Sci Nutr ; 8(3): 1433-1450, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32180953

RESUMO

Glutaminase (glutamine aminohydrolase EC 3.5.1.2) is used in the production of food ingredients rich in l-glutamic acid that are added to finished foods for the purpose of enhancing or improving the savory flavor profile of food. The glutaminase enzyme preparation evaluated in these studies, designated as Sumizyme GT hereafter, is obtained by fermentation of Aspergillus niger strain GT147. The safety of Sumizyme GT was evaluated in a series of standard toxicological studies, including a 90-day oral toxicity study in rats, an in vitro bacterial reverse mutation assay, an in vitro mammalian chromosome aberration test, and an in vivo alkaline Comet assay. Sumizyme GT was not mutagenic or genotoxic, and administration of the enzyme by gavage at doses up to 2,570 mg total organic solids (TOS)/kg body weight (bw) per day for 90 days was without any systemic toxicity. The no-observed-adverse-effect level was concluded to be 2,570 mg TOS/kg bw per day, the highest dose tested. Considering that A. niger has an established history of safe use in the food industry and its safety in the production of food ingredients and food enzymes is well documented, the results of these studies provide further support of the safety of glutaminase from A. niger when used in food production.

3.
Food Chem Toxicol ; 133: 110762, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31421212

RESUMO

Miraculin is a glycoprotein with the ability to make sour substances taste sweet. The safety of miraculin has been evaluated using an approach proposed by the Food and Agriculture Organization of the United Nations and the World Health Organization for assessing the safety of novel proteins. Miraculin was shown to be fully and rapidly digested by pepsin in an in vitro digestibility assay. The proteomic analysis of miraculin's pepsin digests further corroborated that it is highly unlikely that any of the protein will remain intact within the gastrointestinal tract for potential absorption. The potential allergenicity and toxigenicity of miraculin, investigated using in silico bioinformatic analyses, demonstrated that miraculin does not represent a risk of allergy or toxicity to humans with low potential for cross-reactivity with other allergens. The results of a sensory study, characterizing the taste receptor activity of miraculin, showed that the taste-modifying effect of miraculin at the concentration intended for product development has a rapid onset and disappearance with no desensitizing impact on the receptor. Overall, the results of this study demonstrate that the use of miraculin to impact the sensory qualities of orally administered products with a bitter/sour taste profile is not associated with any safety concerns.


Assuntos
Glicoproteínas/toxicidade , Edulcorantes/toxicidade , Alérgenos/química , Alérgenos/isolamento & purificação , Alérgenos/toxicidade , Simulação por Computador , Frutas/química , Glicoproteínas/química , Glicoproteínas/isolamento & purificação , Humanos , Pepsina A/química , Proteólise , Edulcorantes/química , Edulcorantes/isolamento & purificação , Synsepalum/química , Paladar/efeitos dos fármacos
4.
Regul Toxicol Pharmacol ; 87: 71-87, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28487066

RESUMO

The potential toxicity of two savory food ingredients produced by fermentation of enzymatically hydrolyzed corn starch (Savory Base 100 and Savory Base 200) was evaluated individually in a bacterial reverse mutation assay, an in vitro mammalian cell gene mutation assay, an acute oral study and as a mixture in a 90-day dietary study. In the bacterial reverse mutation and in vitro mammalian cell gene mutation assays, neither ingredient was mutagenic at concentrations up to 5000 µg/plate and 5000 µg/mL, respectively in the presence and absence of metabolic activation. In the acute study, the no-observed-adverse-effect level (NOAEL) for each Savory Base 100 and Savory Base 200 in male and female rats was 2000 mg/kg body weight. In the 90-day study, the hematology and clinical chemistry findings and histopathological changes noted in the liver, heart and kidneys were deemed to be of no toxicological significance, as the mean values were within the historical control range, were not dose-dependent, occurred at a similar frequency in control groups, or only occurred in the control group. Considering these findings, the NOAEL for Savory Base 100 and Savory Base 200 was 2333 and 1167 mg/kg body weight, respectively, the highest dose tested in each case.


Assuntos
Ingredientes de Alimentos/toxicidade , Satureja/toxicidade , Testes de Toxicidade Aguda , Testes de Toxicidade Subcrônica , Animais , Dano ao DNA , Feminino , Fermentação , Coração/efeitos dos fármacos , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Testes de Mutagenicidade , Mutagênicos/administração & dosagem , Mutagênicos/toxicidade , Nível de Efeito Adverso não Observado , Ratos
5.
Regul Toxicol Pharmacol ; 70(1): 107-21, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24995591

RESUMO

The potential use of pyrroloquinoline quinone disodium salt (BioPQQ™), as a supplemental food ingredient, was evaluated in a range of oral toxicity studies in rats including an acute study, a 14-day preliminary and a 28-day repeated-dose study, and a 13-week subchronic study. The median lethal dose of BioPQQ™ was shown to be 1000-2000mg/kg body weight (bw) in male and 500-1000mg/kgbw in female rats. In the 14-day study, high doses of BioPQQ™ resulted in increases in relative kidney weights with associated histopathology in female rats only, while a follow-up 28-day study in female animals resulted in increases in urinary protein and crystals. These findings were reversible, and resolved during the recovery period. In the 13-week study, a number of clinical chemistry findings and histopathological changes were noted, which were deemed to be of no toxicological significance, as the levels were within the historical control range, were not dose-dependent, occurred at a similar frequency in control groups, or only occurred in the control group. Based on these findings, a no-observed-adverse-effect level of 100mg/kgbw/day was determined for BioPQQ™ in rats, the highest dose tested in the 13-week study.


Assuntos
Rim/efeitos dos fármacos , Cofator PQQ/toxicidade , Testes de Toxicidade Aguda/métodos , Testes de Toxicidade Subcrônica/métodos , Animais , Relação Dose-Resposta a Droga , Feminino , Rim/metabolismo , Dose Letal Mediana , Masculino , Nível de Efeito Adverso não Observado , Tamanho do Órgão/efeitos dos fármacos , Cofator PQQ/administração & dosagem , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
Crit Rev Toxicol ; 41(1): 22-49, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21077788

RESUMO

Nanotechnology is poised to impact the food and food-related industries through improvements in areas as diverse as production, packaging, shelf life, and bioavailability of food and beverage components. An evaluation was undertaken to characterize the published literature pertaining to the safety of oral exposure to food-related nanomaterials and to identify research needs in this area. Thirty publications were identified in which a toxicological endpoint was assessed following in vivo (oral) or in vitro exposure to food-related nanomaterials. These publications were evaluated for overall quality using a two-step method that determined the reliability of the study design and the extent of nanomaterial characterization within each study. Of the 21 in vivo studies evaluated, 20 used mice or rats, 15 were lacking in some critical component of study design (e.g., oral gavage dose volume was not reported), none was longer than 90 days in duration, and only seven reported more than five physicochemical parameters for the nanomaterial(s) being evaluated. Of the nine in vitro studies evaluated, seven focused on cytotoxicity, two evaluated genotoxicity, only five reported more than five physicochemical parameters for the nanomaterial(s) being evaluated, and none discussed the potential interference by the nanomaterial(s) of the experimental assays that were employed. The results of this evaluation indicate that there is currently insufficient reliable data to allow clear assessment of the safety of oral exposure to food-related nanomaterials. Significant investment must be made to generate studies of sufficient quality and duration and that report comprehensive nanomaterial characterization such that results can be judged reliable and interpretable. Failure to do so will result in the perpetuation of the publication of studies that are inadequate for use in risk characterization.


Assuntos
Inocuidade dos Alimentos , Nanopartículas/toxicidade , Nanoestruturas/toxicidade , Testes de Toxicidade , Administração Oral , Animais , Fenômenos Químicos , Alimentos , Camundongos , Nanotecnologia , Ratos
7.
Regul Toxicol Pharmacol ; 57(2-3): 210-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20197075

RESUMO

An enzymatically-synthesized glycogen (ESG), intended for use as a food ingredient, was investigated for potential toxicity. ESG is synthesized in vitro from short-chain amylose by the co-operative action of branching enzyme and amylomaltase. In an acute toxicity study, oral administration of ESG to Sprague-Dawley rats at a dose of 2000 mg/kg body weight did not result in any signs of toxicity. ESG did not exhibit mutagenic activity in an in vitro bacterial reverse mutation assay. In a subchronic toxicity study, increased cecal weights noted in the mid- (10%) and high-dose (30%) animals are common findings in rodents fed excess amounts of carbohydrates that increase osmotic value of the cecal contents, and thus were considered a physiological rather than toxicological response. The hematological and histopathological effects observed in the high-dose groups were of no toxicological concern as they were secondary to the physiological responses resulting from the high carbohydrate levels in the test diets. The no-observed-adverse-effect level for ESG in rats was therefore established to be 30% in the diet (equivalent to approximately 18 and 21 g/kg body weight/day for male and female rats, respectively). These results support the safety of ESG as a food ingredient for human consumption.


Assuntos
Amilose/química , Qualidade de Produtos para o Consumidor , Dextrinas/química , Sistema da Enzima Desramificadora do Glicogênio/química , Glicogênio/toxicidade , Isoamilase/química , Animais , Carboidratos da Dieta/análise , Carboidratos da Dieta/toxicidade , Relação Dose-Resposta a Droga , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Feminino , Glicogênio/síntese química , Glicogênio/química , Humanos , Masculino , Testes de Mutagenicidade , Ratos , Ratos Sprague-Dawley , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética , Testes de Toxicidade Aguda , Testes de Toxicidade Crônica
8.
Regul Toxicol Pharmacol ; 57(1): 62-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20043969

RESUMO

A recombinant amylomaltase, MQ-01, obtained by cultivation of Bacillus subtilis expressing the amylomaltase gene from Thermus aquaticus is to be used in the production of enzymatically-synthesized glycogen; which is intended for use as a food ingredient. In order to establish the safety of MQ-01, the enzyme was subjected to standard toxicological testing. In a battery of standard Salmonella typhimurium strains (TA98, TA100, TA1535, and TA1537) and in Escherichia coli WP2 uvrA, both with and without metabolic activation, MQ-01 failed to exhibit mutagenic activity. Similarly, MQ-01 did not display clastogenic properties in Chinese hamster lung fibroblast cells (CHL/IU), in an in vitro chromosomal aberration assay. In a 13-week subchronic toxicity study in rats, oral administration of MQ-01 at doses of up to 15 mL/kg body weight/day (corresponding to approximately 1230 mg/kg body weight/day) did not produce compound-related clinical signs or toxicity, changes in body weight gain, food consumption, hematology, clinical chemistry, urinalysis, organ weights, or in any gross and microscopic findings. The results of this study support the safety of MQ-01 in food production.


Assuntos
Qualidade de Produtos para o Consumidor , Sistema da Enzima Desramificadora do Glicogênio/toxicidade , Proteínas Recombinantes/toxicidade , Thermus/enzimologia , Animais , Bacillus subtilis/enzimologia , Bacillus subtilis/genética , Aberrações Cromossômicas/induzido quimicamente , Cricetinae , Cricetulus , Feminino , Aditivos Alimentares , Sistema da Enzima Desramificadora do Glicogênio/genética , Masculino , Testes de Mutagenicidade , Ratos , Ratos Sprague-Dawley , Thermus/genética , Testes de Toxicidade Crônica
9.
Artigo em Inglês | MEDLINE | ID: mdl-19938328

RESUMO

The risk assessment of migration products resulting from packaging material has and continues to pose a difficult challenge. In most jurisdictions, there are regulatory requirements for the approval or notification of food contact substances that will be used in packaging. These processes generally require risk assessment to ensure safety concerns are addressed. The science of assessing food contact materials was instrumental in the development of the concept of Threshold of Regulation and the Threshold of Toxicological Concern procedures. While the risk assessment process is in place, the technology of food packaging continues to evolve to include new initiatives, such as the inclusion of antimicrobial substances or enzyme systems to prevent spoilage, use of plastic packaging intended to remain on foods as they are being cooked, to the introduction of more rigid, stable and reusable materials, and active packaging to extend the shelf-life of food. Each new technology brings with it the potential for exposure to new and possibly novel substances as a result of migration, interaction with other chemical packaging components, or, in the case of plastics now used in direct cooking of products, degradation products formed during heating. Furthermore, the presence of trace levels of certain chemicals from packaging that were once accepted as being of low risk based on traditional toxicology studies are being challenged on the basis of reports of adverse effects, particularly with respect to endocrine disruption, alleged to occur at very low doses. A recent example is the case of bisphenol A. The way forward to assess new packaging technologies and reports of very low dose effects in non-standard studies of food contact substances is likely to remain controversial. However, the risk assessment paradigm is sufficiently robust and flexible to be adapted to meet these challenges. The use of the Threshold of Regulation and the Threshold of Toxicological Concern concepts may play a critical role in the risk assessment of new food packaging technologies in the future.


Assuntos
Contaminação de Alimentos/prevenção & controle , Embalagem de Alimentos/normas , Qualidade de Produtos para o Consumidor/legislação & jurisprudência , Qualidade de Produtos para o Consumidor/normas , Contaminação de Alimentos/legislação & jurisprudência , Embalagem de Alimentos/legislação & jurisprudência , Humanos , Nanotecnologia/métodos , Medição de Risco/métodos
10.
Toxicology ; 256(1-2): 101-9, 2009 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-19059302

RESUMO

Amodiaquine is an antimalarial drug that causes life-threatening agranulocytosis and hepatotoxicity in about 1 in 2000 patients, which is usually associated with an inflammatory response. It was found that the LC(50) (2h) of amodiaquine towards isolated rat hepatocytes was 1mM. The cytotoxic mechanism involved protein carbonylation as well as P450 activation to a reactive metabolite. The cytotoxicity, however, was not reactive oxygen species (ROS)-mediated, as ROS scavengers did not prevent cytotoxicity or protein carbonylation, and it was not accompanied by glutathione (GSH) oxidation or intracellular H(2)O(2) formation. On the other hand, the cytotoxicity could be attributed to a quinoneimine metabolite formation which formed GSH conjugates and GSH-depleted hepatocytes were much more susceptible to amodiaquine. Furthermore, when a non-toxic H(2)O(2) generating system and peroxidase was used to mimic the products formed by inflammatory immune cells, only 15microM amodiaquine was required to cause 50% cell death. In the absence of amodiaquine, hepatocyte viability and glutathione levels were not affected by the H(2)O(2) generating system with or without peroxidase. The toxicity mechanism of amodiaquine in this hepatocyte H(2)O(2)/peroxidase model involved oxidative stress, as cytotoxicity was accompanied by GSH oxidation, decreased mitochondrial membrane potential and protein carbonyl formation which were inhibited by ROS scavengers, 4-hydroxy-2,2,6,6-tetramethylpiperidene-1-oxyl (TEMPOL) or mannitol suggesting a role for a semiquinoneimine radical and ROS in the amodiaquine-H(2)O(2)-mediated cytotoxic mechanism.


Assuntos
Amodiaquina/toxicidade , Antimaláricos/toxicidade , Hepatócitos/efeitos dos fármacos , Inflamação/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Glutationa/metabolismo , Hepatócitos/enzimologia , Peróxido de Hidrogênio/toxicidade , Inflamação/enzimologia , Inflamação/patologia , Masculino , Manitol/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Membranas Mitocondriais/efeitos dos fármacos , Oxirredução , Carbonilação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Marcadores de Spin
11.
Chem Res Toxicol ; 21(4): 904-10, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18393451

RESUMO

Long-term treatment of hypertensive disorders with hydralazine has resulted in some patients developing hepatitis and lupus erythematosus, an autoimmune syndrome. The concentration of hydralazine required to cause 50% cytotoxicity in 2 h (LC(50)) toward isolated rat hepatocytes was found to be 8 mM. Cytotoxicity was delayed by the P450 inhibitor, 1-aminobenzotriazole, suggesting that P450 catalyzed the formation of toxic metabolites from hydralazine. No hydralazine-induced oxidative stress was apparent as there was little effect on hepatocyte lipid peroxidation, protein carbonyl formation, intracellular H(2)O(2), or hepatocyte GSH levels and no effect of butylated hydroxyanisole (BHA) on cytotoxicity. Drug-induced hepatotoxicity in vivo has often been attributed to infiltrating inflammatory cells, for example, neutrophils or resident Kupffer cells whose NADPH oxidase generates H(2)O(2), when activated. The effect of a nontoxic continuous infusion of H(2)O(2) on hydralazine cytotoxicity was investigated. It was found that H(2)O(2) increased hepatocyte susceptibility to hydralazine 4-fold (LC(50), 2 mM). Cytotoxicity was still prevented by the P450 inhibitor but now involved some oxidative stress as shown by increased protein carbonyls, endogenous H(2)O(2), and GSH oxidation. Lipid peroxidation was not increased, and cytotoxicity was not inhibited by BHA. Cytotoxicity, however, was inhibited by 4-hydroxy-2,2,6,6-tetramethylpiperidene-1-oxyl (TEMPOL), a ROS scavenger. Because neutrophils or Kupffer cells release myeloperoxidase on activation, the effect of adding peroxidase to the hepatocytes exposed to H(2)O(2) on hydralazine cytotoxicity was investigated. It was found that peroxidase/H(2)O(2) increased hepatocyte susceptibility to hydralazine 80-fold (LC 50, 0.1 mM). Furthermore, cytotoxicity occurred following extensive oxidative stress that included lipid peroxidation, and cytotoxicity that was now prevented by the antioxidant BHA. These results indicate that three cytotoxic pathways exist for hydralazine: a P450-catalyzed pathway not involving oxidative stress, a P450/H(2)O(2)-catalyzed oxidative stress-mediated cytotoxic pathway not involving lipid peroxidation, and a peroxidase/H(2)O(2)-catalyzed lipid peroxidation-mediated cytotoxic pathway.


Assuntos
Anti-Hipertensivos/toxicidade , Hepatócitos/efeitos dos fármacos , Hidralazina/toxicidade , Estresse Oxidativo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Peróxido de Hidrogênio/metabolismo , Peroxidação de Lipídeos , Masculino , Peroxidase/metabolismo , Carbonilação Proteica , Ratos , Ratos Sprague-Dawley , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
12.
Toxicol Appl Pharmacol ; 229(1): 94-101, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18295292

RESUMO

Isoniazid is an anti-tuberculosis drug that can cause hepatotoxicity in 20% of patients that is usually associated with an inflammatory response. Hepatocytes when exposed to non-toxic levels of H2O2, to simulate H2O2 formation by inflammatory cells, became twice as sensitive to isoniazid toxicity. Isoniazid cytotoxicity was prevented by 1-aminobenzotriazole, a non-selective P450 inhibitor or by bis-p-nitrophenyl phosphate (BNPP), an esterase inhibitor. Moreover, the cytotoxicity of hydrazine, the metabolite formed by amidase-catalyzed hydrolysis of isoniazid, was increased 16-fold by a non-toxic H2O2-generating system. The acetylhydrazine metabolite was found to be much less cytotoxic than hydrazine in this hepatocyte inflammation model. Hydrazine, therefore, seems to be the isoniazid reactive metabolite in this inflammation model. The molecular mechanism of hydrazine-induced cytotoxicity was attributed to oxidative stress as reactive oxygen species (ROS) and protein carbonyl formation occurred before the onset of hepatocyte toxicity. Hydrazine toxicity also involved significant production of endogenous H2O2 which resulted in lysosomal membrane damage and leads to a collapse in mitochondrial membrane potential. These results implicated H2O2, a cellular mediator of inflammation, as a potential risk factor for the manifestation of adverse drug reactions, particularly those caused by hydrazine containing drugs.


Assuntos
Antituberculosos/toxicidade , Hepatócitos/efeitos dos fármacos , Hidrazinas/toxicidade , Isoniazida/toxicidade , Animais , Antituberculosos/metabolismo , Doença Hepática Induzida por Substâncias e Drogas , Hepatócitos/patologia , Hidrazinas/metabolismo , Peróxido de Hidrogênio/metabolismo , Técnicas In Vitro , Inflamação/induzido quimicamente , Isoniazida/metabolismo , Lisossomos/metabolismo , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Carbonilação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
13.
Drug Metabol Drug Interact ; 21(1): 19-29, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16086553

RESUMO

Vimang is an aqueous extract of Mangifera indica used in Cuba to improve the quality of life in patients suffering from inflammatory diseases. In the present study we evaluated the effects of Vimang at preventing reactive oxygen species (ROS) formation and lipid peroxidation in intact isolated rat hepatocytes. Vimang at 20, 50 and 100 microg/ml inhibited hepatocyte ROS formation induced by glucose-glucose oxidase. Hepatocyte cytotoxicity and lipid peroxidation induced by cumene hydroperoxide was also inhibited by Vimang in a dose and time dependent manner at the same concentration. Vimang also inhibited superoxide radical formation by xanthine oxidase and hypoxanthine. The superoxide radical scavenging and antioxidant activity of the Vimang extract was likely related to its gallates, catechins and mangiferin content. To our knowledge, this is the first report of cytoprotective antioxidant effects of Vimang in cellular oxidative stress models.


Assuntos
Hepatócitos/efeitos dos fármacos , Peroxidação de Lipídeos/fisiologia , Extratos Vegetais/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Antioxidantes/farmacologia , Derivados de Benzeno/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ácido Gálico/farmacologia , Glucose Oxidase/farmacologia , Hepatócitos/metabolismo , Hipoxantina/farmacologia , Masculino , Mangifera , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Galato de Propila/farmacologia , Ratos , Ratos Sprague-Dawley , Superóxidos/metabolismo , Xantina Oxidase/metabolismo
14.
Drug Metab Rev ; 37(2): 311-25, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15931767

RESUMO

The following describes a novel screening method for "new chemical entities" (NCEs), suitable for ADMET studies, that measures ability to form prooxidant radicals on metabolism and their ability to induce oxidative stress in intact cells. The accelerated molecular cytotoxic mechanism screening (ACMS) techniques used with isolated rat hepatocytes showed that cytotoxicity is usually initiated as a result of macromolecular covalent binding or macromolecular oxidative stress. While P450 is likely responsible for drug metabolic activation in the liver, intestine, lung, and in other nonhepatic tissues, where P450 levels are low, peroxidases including prostaglandin synthetase peroxidase can catalyze xenobiotic one-electron oxidation to form prooxidant free radicals that may cause toxicity or carcinogenesis. Inflammation markedly activates H2O2, generating NADPH oxidase and peroxidase of certain immune cells when they infiltrate tissues including the liver. Myeloperoxidase and NADPH oxidase in the Kupffer cells (resident macrophages of the liver) also become activated during inflammation. The addition of noncytotoxic concentrations of peroxidase/H2O2 to the hepatocyte incubate markedly increased drug cytotoxicity and prooxidant radical formation as shown by glutathione or lipid oxidation. Many drugs that have hepato- or gastrointestinal (GI) toxicity problems or were withdrawn from the market for safety problems, e.g., troglitazone, tolcapone, mefenamic acid, diclofenac, and phenylbutazone, were markedly more toxic and prooxidant in this inflammation model system, whereas other drugs, e.g., entacapone, were not toxic in this inflammation model. Some of the idiosyncratic hepatotoxicity responsible for recent drug withdrawals may therefore result from commonplace sporadic inflammatory episodes during drug therapy.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Estresse Oxidativo/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/patologia , Cromatografia Líquida de Alta Pressão , Glutationa/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Inflamação/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Modelos Biológicos , NAD/metabolismo , Oxirredução , Consumo de Oxigênio/efeitos dos fármacos , Ratos
15.
Drug Discov Today ; 10(9): 617-25, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15894226

RESUMO

Current safety screening of drug candidates or new chemical entities for reactive metabolite formation focuses on the role of cytochrome P450. However, peroxidases also have a major role in drug metabolism, and peroxidase-catalyzed drug oxidation could lead to reactive metabolite formation, resulting in oxidative stress and cytotoxicity. Here, the different classes of human peroxidases are summarized and the molecular mechanisms of peroxidase-catalyzed drug metabolism are discussed. In addition, evidence is presented that indicates a role of these enzymes in drug toxicity.


Assuntos
Peroxidases/metabolismo , Preparações Farmacêuticas/metabolismo , Biotransformação , Sistema Enzimático do Citocromo P-450 , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Oxirredução , Estresse Oxidativo
16.
Chem Res Toxicol ; 18(10): 1567-74, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16533021

RESUMO

The order of antioxidant effectiveness of low concentrations of vitamin E analogues, in preventing cumene hydroperoxide-induced hepatocyte lipid peroxidation and cytotoxicity, was 2,2,5,7,8-pentamethyl-6-hydroxychromane (PMC) > troglitazone > Trolox C > alpha-tocopherol > gamma-tocopherol > delta-tocopherol. However, vitamin E analogues, including troglitazone at higher concentrations, induced microsomal lipid peroxidation when oxidized to phenoxyl radicals by peroxidase/H2O2. Ascorbate or GSH was also cooxidized, and GSH cooxidation by vitamin E analogue phenoxyl radicals was also accompanied by extensive oxygen uptake and oxygen activation. When oxidized by nontoxic concentrations of peroxidase/H2O2, vitamin E analogues except PMC also caused hepatocyte cytotoxicity, lipid peroxidation, and GSH oxidation. The prooxidant order of vitamin E analogues in catalyzing hepatocyte cytotoxicity, lipid peroxidation, and GSH oxidation was troglitazone > Trolox C > delta-tocopherol > gamma-tocopherol > alpha-tocopherol > PMC. A similar order of effectiveness was found for GSH cooxidation or microsomal lipid peroxidation but not for ascorbate cooxidation. Except for troglitazone, the toxic prooxidant activity of vitamin E analogues was therefore inversely proportional to their antioxidant activity. The high troglitazone prooxidant activity could be a contributing factor to its hepatotoxicity. We have also derived equations for three-parameter quantitative structure-activity relationships (QSARs), which described the correlation between antioxidant and prooxidant activity of vitamin E ananlogues and their lipophilicity (log P), ionization potential (E(HOMO)), and dipole moment.


Assuntos
Antioxidantes/farmacologia , Hepatócitos/efeitos dos fármacos , Microssomos Hepáticos/efeitos dos fármacos , Oxidantes/farmacologia , Vitamina E/análogos & derivados , Animais , Antioxidantes/química , Sobrevivência Celular , Sistema Livre de Células , Cromanos/química , Glutationa/metabolismo , Hepatócitos/metabolismo , Peróxido de Hidrogênio , Peroxidação de Lipídeos , Masculino , Microssomos Hepáticos/metabolismo , Oxidantes/química , Oxirredução , Peroxidase , Fenóis/metabolismo , Relação Quantitativa Estrutura-Atividade , Ratos , Ratos Sprague-Dawley , Tiazolidinedionas/química , Tocoferóis/química , Troglitazona , Vitamina E/química
17.
Chem Biol Interact ; 151(1): 43-51, 2004 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-15607761

RESUMO

The antipsychotic phenothiazines may have other therapeutic applications because of their ability to kill bacteria, plasmids and tumor cells. They are also known to undergo a peroxidase-catalysed oxidation to form cation radicals that are stable at acid pH, but are not detected at a neutral pH. The objective of this project was to determine whether phenothiazine cation radical metabolites could cause oxidative stress at a neutral pH resulting in cytotoxicity. At a neutral pH, catalytic amounts of phenothiazines were found to be oxidised by a peroxidase/H2O2 system and also caused ascorbate, GSH and NADH cooxidation. NADH and GSH co-oxidation was accompanied by oxygen uptake and was increased by the addition of catalytic amounts of superoxide dismutase, indicating that the superoxide radical was formed. The phenothazines were different from other peroxidase substrates in that the NADH, ascorbate or GSH cooxidation was faster at pH 6.0 than pH 7.4, thereby partly reflecting the cation radical stability. The order of catalytic effectiveness found was promazine > chlorpromazine > trifluoperazine. Peroxidase/H2O2 also markedly increased phenothiazine cytotoxicity towards isolated rat hepatocytes at nontoxic phenothiazine concentrations. At both pH 6.0 and 7.4, the same order of phenothiazine catalytic effectiveness was observed as seen in the co-oxidation experiments. Cytotoxicity to hepatocytes could be attributed to oxidative stress as most hepatocyte glutathione oxidation and lipid peroxidation preceded phenothiazine induced cytotoxicity and that cytotoxicity was prevented by the antioxidant butylated hydroxyanisole. This hepatocyte/peroxidase/H2O2 system could be a useful model for studying drug induced idiosyncratic hepatic injury enhanced by inflammation.


Assuntos
Antipsicóticos/farmacologia , Hepatócitos/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Peroxidases/metabolismo , Fenotiazinas/farmacologia , Espécies Reativas de Oxigênio/farmacologia , Animais , Antipsicóticos/metabolismo , Catálise , Radicais Livres , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Masculino , NAD/metabolismo , Fenotiazinas/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
18.
Chem Res Toxicol ; 17(10): 1350-5, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15487895

RESUMO

Previously, it was shown that indole-3-acetic acid (IAA) is a nontoxic prodrug that forms a radical, toxic to tumor cells when activated by peroxidase. Because of this, IAA and peroxidase conjugated to an antibody specific to an extracelluar tumor antigen are currently in phase II clinical trials. In the following, the prooxidant activities of the radicals formed were compared when IAA or its derivatives were metabolically oxidized by peroxidase/H(2)O(2). In general, it was found that the effectiveness of IAA analogues for catalyzing the cooxidation of ascorbate, NADH, or GSH increased as the IAA derivatives were more readily oxidized by HRP/H(2)O(2). The order of effectiveness of IAA derivatives at cooxidizing NADH, ascorbate, GSH, and hepatocyte GSH was 5MeO-2Me-IAA > 2Me-IAA > 5MeO-IAA > IAA. The rates of NADH and ascorbate cooxidation were faster at pH 7.4 than at pH 6.0, whereas GSH cooxidation was faster at pH 6.0 than at pH 7.4. Furthermore, NADH, ascorbate, and GSH prevented the oxidation of IAA derivatives, which suggested that the indolyl cation radical was responsible for the prooxidant activity. The effectiveness of IAA derivatives in catalyzing lipid peroxidation at pH 7.4 was similar and also correlated with the rate of oxidation of IAA derivatives by HRP-I and the one-electron potential of these compounds. The IAA derivative-induced lipid peroxidation was faster at pH 6.0 than at pH 7.4. IAA derivative effectiveness at catalyzing microsomal and hepatocyte lipid peroxidation or hepatocyte reactive oxygen species formation at pH 6.0 was IAA > 5MeO-2Me-IAA > 2Me-IAA > 5MeO-IAA, but at pH 7.4, it was 5MeO-2Me-IAA > 2Me-IAA > 5MeO-IAA > IAA. Previously, the rate of radical cation decarboxylation to skatole radicals and (skatole) peroxyl radicals was reported to be faster at an acid pH with IAA being more effective than the derivatives. This suggests that IAA skatole and/or (skatole) peroxyl radicals catalyze lipid peroxidation at pH 6.0. Incubation of isolated rat hepatocytes with IAA analogues/H(2)O(2)/peroxidase also resulted in cytotoxicity with 5MeO-2Me-IAA being the most effective at pH 7.4 and IAA being the most effective at pH 6.0. Cytotoxicity was also prevented by antioxidants.


Assuntos
Radicais Livres/toxicidade , Ácidos Indolacéticos/toxicidade , Animais , Antioxidantes/química , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Ácido Ascórbico/metabolismo , Catálise , Glutationa/metabolismo , Peroxidase do Rábano Silvestre/metabolismo , Peroxidase do Rábano Silvestre/farmacologia , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Concentração de Íons de Hidrogênio , Ácidos Indolacéticos/química , Modelos Químicos , NAD/metabolismo , Oxirredução , Peroxidase/metabolismo , Ratos , Ratos Sprague-Dawley
19.
Chem Biol Interact ; 142(1-2): 25-41, 2002 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-12399153

RESUMO

Many idiosyncratic non-steroidal anti-inflammatory drugs (NSAIDs) cause GI, liver and bone marrow toxicity in some patients which results in GI bleeding/ulceration/fulminant hepatic failure/hepatitis or agranulocytosis/aplastic anemia. The toxic mechanisms proposed have been reviewed. Evidence is presented showing that idiosyncratic NSAID drugs form prooxidant radicals when metabolised by peroxidases known to be present in these tissues. Thus GSH, NADH and/or ascorbate were cooxidised by catalytic amounts of NSAIDs and hydrogen peroxide in the presence of peroxidase. During GSH and NADH cooxidation, oxygen uptake and activation occurred. Furthermore the formation of NSAID oxidation products was prevented during the cooxidation indicating that the cooxidation involved redox cycling of the first formed NSAID radical product. The order of prooxidant catalytic effectiveness of fenamate and arylacetic acid NSAIDs was mefenamic acid>tolfenamic acid>flufenamic acid, meclofenamic acid or diclofenac. Diphenylamine, a common moiety to all of these NSAIDs was a more active prooxidant for NADH and ascorbate cooxidation than these NSAIDs which suggests that oxidation of the NSAID diphenylamine moiety to a cation and/or nitroxide radical was responsible for the NSAID prooxidant activity. The order of catalytic effectiveness found for sulfonamide derivatives was sulfaphenazole>sulfisoxazolez.Gt;dapsone>sulfanilic acid>procainamide>sulfamethoxazole>sulfadiazine>sulfadimethoxine whereas sulfanilamide, sulfapyridine or nimesulide had no prooxidant activity. Although indomethacin had little prooxidant activity, its major in vivo metabolite, N-deschlorobenzoyl indomethacin had significant prooxidant activity. Aminoantipyrine the major in vivo metabolite of aminopyrine or dipyrone was also more prooxidant than the parent drugs. It is hypothesized that the NSAID radicals and/or the resulting oxidative stress initiates the cytotoxic processes leading to idiosyncratic toxicity.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Estresse Oxidativo , Animais , Anti-Inflamatórios não Esteroides/toxicidade , Ácido Ascórbico/metabolismo , Radicais Livres/metabolismo , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/metabolismo , Glutationa/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/efeitos dos fármacos , Masculino , NAD/metabolismo , Oxirredução , Peroxidase/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...