Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Annu Rev Pharmacol Toxicol ; 54: 559-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24160697

RESUMO

The fact that the bacteria in the human gastrointestinal (GI) tract play a symbiotic role was noted as early as 1885, well before we began to manage microbial infections using antibiotics. However, even with the first antimicrobial compounds used in humans, the sulfa drugs, microbes were recognized to be critically involved in the biotransformation of these therapeutics. Thus, the roles played by the microbiota in physiology and in the management of human health have long been appreciated. Detailed examinations of GI symbiotic bacteria that started in the early 2000s and the first phases of the Human Microbiome Project that were completed in 2012 have ushered in an exciting period of granularity with respect to the ecology, genetics, and chemistry of the mammalian-microbial axes of communication. Here we review aspects of the biochemical pathways at play between commensal GI bacteria and several mammalian systems, including both local-epithelia and nonlocal responses impacting inflammation, immunology, metabolism, and neurobiology. Finally, we discuss how the microbial biotransformation of therapeutic compounds, such as anticancer or nonsteroidal anti-inflammatory drugs, can be modulated to reduce toxicity and potentially improve therapeutic efficacy.


Assuntos
Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Mamíferos , Microbiota/efeitos dos fármacos , Animais , Anti-Inflamatórios/farmacologia , Camptotecina/administração & dosagem , Camptotecina/efeitos adversos , Camptotecina/análogos & derivados , Modelos Animais de Doenças , Enterite/induzido quimicamente , Enterite/tratamento farmacológico , Eosinofilia/induzido quimicamente , Eosinofilia/tratamento farmacológico , Gastrite/induzido quimicamente , Gastrite/tratamento farmacológico , Glucuronidase/farmacologia , Humanos , Irinotecano , Lipopolissacarídeos/farmacologia
2.
Hepatology ; 61(1): 326-36, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25142022

RESUMO

UNLABELLED: Acetaminophen (APAP) overdose is a frequent cause of drug-induced liver injury and the most frequent cause of acute liver failure in the Western world. Previous studies with mouse models have revealed that impairment of mitochondrial respiration is an early event in the pathogenesis, but the exact mechanisms have remained unclear, and therapeutic approaches to specifically target mitochondria have been insufficiently explored. Here, we found that the reactive oxidative metabolite of APAP, N-acetyl-p-benzoquinoneimine (NAPQI), caused the selective inhibition of mitochondrial complex II activity by >90% in both mouse hepatic mitochondria and yeast-derived complexes reconstituted into nanoscale model membranes, as well as the decrease of succinate-driven adenosine triphosphate (ATP) biosynthesis rates. Based on these findings, we hypothesized that methylene blue (MB), a mitochondria-permeant redox-active compound that can act as an alternative electron carrier, protects against APAP-induced hepatocyte injury. We found that MB (<3 µM) readily accepted electrons from NAPQI-altered, succinate-energized complex II and transferred them to cytochrome c, restoring ATP biosynthesis rates. In cultured mouse hepatocytes, MB prevented the mitochondrial permeability transition and loss of intracellular ATP without interfering with APAP bioactivation. In male C57BL/6J mice treated with APAP (450 mg/kg, intraperitoneally [IP]), MB (10 mg/kg, IP, administered 90 minutes post-APAP) protected against hepatotoxicity, whereas mice treated with APAP alone developed massive centrilobular necrosis and increased serum alanine aminotransferase activity. APAP treatment inhibited complex II activity ex vivo, but did not alter the protein expression levels of subunits SdhA or SdhC after 4 hours. CONCLUSION: MB can effectively protect mice against APAP-induced liver injury by bypassing the NAPQI-altered mitochondrial complex II, thus alleviating the cellular energy crisis. Because MB is a clinically used drug, its potential application after APAP overdose in patients should be further explored.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Inibidores Enzimáticos/uso terapêutico , Hepatócitos/efeitos dos fármacos , Azul de Metileno/uso terapêutico , Mitocôndrias Hepáticas/efeitos dos fármacos , Acetaminofen/efeitos adversos , Analgésicos não Narcóticos/efeitos adversos , Animais , Benzoquinonas/efeitos adversos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Avaliação Pré-Clínica de Medicamentos , Complexo II de Transporte de Elétrons/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Iminas/efeitos adversos , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Azul de Metileno/farmacologia , Camundongos Endogâmicos C57BL , Mitocôndrias Hepáticas/metabolismo , Necrose/prevenção & controle , Ácido Peroxinitroso/metabolismo , Succinato Desidrogenase/metabolismo
3.
J Gastroenterol Hepatol ; 29(4): 678-87, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24783247

RESUMO

Idiosyncratic drug-induced liver injury (DILI) is a significant adverse effect of antitubercular therapy with isoniazid (INH). Although the drug has been used for many decades, the underlying mode of action (both patient-specific and drug-specific mechanisms) leading to DILI are poorly understood. Among the patient-specific determinants of susceptibility to INH-associated DILI, the importance of HLA genetic variants has been increasingly recognized, whereas the role of polymorphisms of drug-metabolizing enzymes (NAT2 and CYP2E1) has become less important and remains controversial. However, these polymorphisms are merely correlative, and other molecular determinants of susceptibility have remained largely unknown. Regarding the drug-specific mechanisms underlying INH-induced liver injury, novel concepts have been emerging. Among these are covalent protein adduct formation via novel reactive intermediates, leading to hapten formation and a potential immune response, and interference with endogenous metabolism. Furthermore, INH and/or INH metabolites (e.g. hydrazine) can cause mitochondrial injury, which can lead to mitochondrial oxidant stress and impairment of energy homeostasis. Recent studies have revealed that underlying impairment of complex I function can trigger massive hepatocellular injury induced by otherwise nontoxic concentrations of INH superimposed on these mitochondrial deficiencies. This review discusses these emerging new paradigms of INH-induced DILI and highlights recent insights into the mechanisms, as well as points to the existing large gaps in our understanding of the pathogenesis.


Assuntos
Antituberculosos/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/genética , Predisposição Genética para Doença/genética , Isoniazida/efeitos adversos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Arilamina N-Acetiltransferase/genética , Citocromo P-450 CYP2E1/genética , Metabolismo Energético/efeitos dos fármacos , Antígenos HLA/genética , Homeostase/efeitos dos fármacos , Humanos , Hidrazinas/efeitos adversos , Polimorfismo Genético
4.
Xenobiotica ; 44(1): 28-35, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23829165

RESUMO

1. We have previously demonstrated that a small molecule inhibitor of bacterial ß-glucuronidase (Inh-1; [1-((6,8-dimethyl-2-oxo-1,2-dihydroquinolin-3-yl)-3-(4-ethoxyphenyl)-1-(2-hydroxyethyl)thiourea]) protected mice against diclofenac (DCF)-induced enteropathy. Here we report that Inh-1 was equally protective against small intestinal injury induced by other carboxylic acid-containing non-steroidal anti-inflammatory drugs (NSAIDs), indomethacin (10 mg/kg, ip) and ketoprofen (100 mg/kg, ip). 2. Inh-1 provided complete protection if given prior to DCF (60 mg/kg, ip), and partial protection if administered 3-h post-DCF, suggesting that the temporal window of mucosal protection can be extended for drugs undergoing extensive enterohepatic circulation. 3. Pharmacokinetic analysis of Inh-1 revealed an absolute bioavailability (F) of 21% and a short t1/2 of <1 h. This low F was shown to be due to hepatic first-pass metabolism, as confirmed with the pan-CYP inhibitor, 1-aminobenzotriazole. 4. Using the fluorescent probe 5 (and 6)-carboxy-2',7'-dichlorofluorescein, we demonstrated that Inh-1 did not interfere with hepatobiliary export of glucuronides in gall bladder-cannulated mice. 5. These data are compatible with the hypothesis that pharmacological inhibition of bacterial ß-glucuronidase-mediated cleavage of NSAID glucuronides in the small intestinal lumen can protect against NSAID-induced enteropathy caused by locally high concentrations of NSAID aglycones.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Inibidores Enzimáticos/farmacologia , Glucuronidase/antagonistas & inibidores , Enteropatias/induzido quimicamente , Enteropatias/prevenção & controle , Quinolonas/farmacologia , Tioureia/análogos & derivados , Animais , Cromatografia Líquida , Diclofenaco/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Hepatócitos/efeitos dos fármacos , Indometacina/efeitos adversos , Intestino Delgado/patologia , Cetoprofeno/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Quinolonas/farmacocinética , Estatísticas não Paramétricas , Espectrometria de Massas em Tandem , Tioureia/farmacocinética , Tioureia/farmacologia
5.
J Proteome Res ; 12(6): 2933-45, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23659346

RESUMO

Troglitazone, a first-generation thiazolidinedione of antihyperglycaemic properties, was withdrawn from the market due to unacceptable idiosyncratic hepatotoxicity. Despite intensive research, the underlying mechanism of troglitazone-induced liver toxicity remains unknown. Here we report the use of the Sod2(+/-) mouse model of silent mitochondrial oxidative-stress-based and quantitative mass spectrometry-based proteomics to track the mitochondrial proteome changes induced by physiologically relevant troglitazone doses. By quantitative untargeted proteomics, we first globally profiled the Sod2(+/-) hepatic mitochondria proteome and found perturbations including GSH metabolism that enhanced the toxicity of the normally nontoxic troglitazone. Short- and long-term troglitazone administration in Sod2(+/-) mouse led to a mitochondrial proteome shift from an early compensatory response to an eventual phase of intolerable oxidative stress, due to decreased mitochondrial glutathione (mGSH) import protein, decreased dicarboxylate ion carrier (DIC), and the specific activation of ASK1-JNK and FOXO3a with prolonged troglitazone exposure. Furthermore, mapping of the detected proteins onto mouse specific protein-centered networks revealed lipid-associated proteins as contributors to overt mitochondrial and liver injury when under prolonged exposure to the lipid-normalizing troglitazone. By integrative toxicoproteomics, we demonstrated a powerful systems approach in identifying the collapse of specific fragile nodes and activation of crucial proteome reconfiguration regulators when targeted by an exogenous toxicant.


Assuntos
Cromanos/toxicidade , Glutationa/antagonistas & inibidores , Hipoglicemiantes/toxicidade , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/genética , Proteômica , Tiazolidinedionas/toxicidade , Animais , Transportadores de Ácidos Dicarboxílicos/antagonistas & inibidores , Transportadores de Ácidos Dicarboxílicos/genética , Transportadores de Ácidos Dicarboxílicos/metabolismo , Feminino , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/agonistas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Transporte de Íons/efeitos dos fármacos , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase Quinase 5/genética , MAP Quinase Quinase Quinase 5/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais , Superóxido Dismutase/deficiência , Superóxido Dismutase/genética , Troglitazona
6.
J Pharmacol Exp Ther ; 341(2): 447-54, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22328575

RESUMO

Small intestinal mucosal injury is a frequent adverse effect caused by nonsteroidal anti-inflammatory drugs (NSAIDs). The underlying mechanisms are not completely understood, but topical (luminal) effects have been implicated. Many carboxylic acid-containing NSAIDs, including diclofenac (DCF), are metabolized to acyl glucuronides (AGs), and/or ether glucuronides after ring hydroxylation, and exported into the biliary tree. In the gut, these conjugates are cleaved by bacterial ß-glucuronidase, releasing the potentially harmful aglycone. We first confirmed that DCF-AG was an excellent substrate for purified Escherichia coli ß-D-glucuronidase. Using a previously characterized novel bacteria-specific ß-glucuronidase inhibitor (Inhibitor-1), we then found that the enzymatic hydrolysis of DCF-AG in vitro was inhibited concentration dependently (IC50 ∼164 nM). We next hypothesized that pharmacologic inhibition of bacterial ß-glucuronidase would reduce exposure of enterocytes to the aglycone and, as a result, alleviate enteropathy. C57BL/6J mice were administered an ulcerogenic dose of DCF (60 mg/kg i.p.) with or without oral pretreatment with Inhibitor-1 (10 µg per mouse, b.i.d.). Whereas DCF alone caused the formation of numerous large ulcers in the distal parts of the small intestine and increased (2-fold) the intestinal permeability to fluorescein isothiocyanate-dextran, Inhibitor-1 cotreatment significantly alleviated mucosal injury and reduced all parameters of enteropathy. Pharmacokinetic profiling of DCF plasma levels in mice revealed that Inhibitor-1 coadministration did not significantly alter the C(max), half-life, or area under the plasma concentration versus time curve of DCF. Thus, highly selective pharmacologic targeting of luminal bacterial ß-D-glucuronidase by a novel class of small-molecule inhibitors protects against DCF-induced enteropathy without altering systemic drug exposure.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Diclofenaco/toxicidade , Glucuronidase/antagonistas & inibidores , Enteropatias/induzido quimicamente , Enteropatias/enzimologia , Animais , Anti-Inflamatórios não Esteroides/farmacocinética , Diclofenaco/farmacocinética , Enterócitos/efeitos dos fármacos , Enterócitos/enzimologia , Enterócitos/metabolismo , Escherichia coli/enzimologia , Escherichia coli/metabolismo , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/metabolismo , Glucuronidase/metabolismo , Glicoproteínas/farmacologia , Enteropatias/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/enzimologia , Mucosa Intestinal/metabolismo , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/enzimologia , Intestino Delgado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Úlcera/induzido quimicamente , Úlcera/enzimologia , Úlcera/metabolismo
7.
Hepatology ; 54(3): 969-78, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21626531

RESUMO

UNLABELLED: Acetaminophen (APAP) is safe at therapeutic dosage but can cause severe hepatotoxicity if used at overdose. The mechanisms of injury are not yet fully understood, but previous reports had suggested that the mitochondrial permeability transition (mPT) may be involved in triggering hepatocellular necrosis. We aimed at inhibiting mitochondrial cyclophilin D (CypD), a key regulator of the mPT, as a potential therapeutic target in APAP hepatotoxicity. Wildtype mice treated with a high dose of APAP (600 mg/kg, intraperitoneal) developed typical centrilobular necrosis, which could not, however, be prevented by cotreatment with the selective CypD inhibitor, Debio 025 (alisporivir, DEB025, a nonimmunosuppressive cyclosporin A analog). Similarly, genetic ablation of mitochondrial CypD in Ppif-null mice did not afford protection from APAP hepatotoxicity. To determine whether APAP-induced peroxynitrite stress might directly activate mitochondrial permeabilization, independently of the CypD-regulated mPT, we coadministered the peroxynitrite decomposition catalyst Fe-TMPyP (10 mg/kg, intraperitoneal, 90 minutes prior to APAP) to CypD-deficient mice. Liver injury was greatly attenuated by Fe-TMPyP pretreatment, and mitochondrial 3-nitrotyrosine adduct levels (peroxynitrite marker) were decreased. Acetaminophen treatment increased both the cytosolic and mitochondria-associated P-JNK levels, but the c-jun-N-terminal kinase (JNK) signaling inhibitor SP600125 was hepatoprotective in wildtype mice only, indicating that the JNK pathway may not be critically involved in the absence of CypD. CONCLUSION: These data support the concept that an overdose of APAP results in liver injury that is refractory to pharmacological inhibition or genetic depletion of CypD and that peroxynitrite-mediated cell injury predominates in the absence of CypD.


Assuntos
Acetaminofen/intoxicação , Analgésicos não Narcóticos/intoxicação , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Ciclofilinas/fisiologia , Proteínas de Transporte da Membrana Mitocondrial , Ácido Peroxinitroso/fisiologia , Animais , Peptidil-Prolil Isomerase F , Overdose de Drogas , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Poro de Transição de Permeabilidade Mitocondrial , Porfirinas/farmacologia
8.
Mass Spectrom Rev ; 29(2): 179-96, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19294730

RESUMO

Oxidative stress has been implicated in the pathogenesis of numerous human diseases and disorders, but the mechanistic basis often remains enigmatic. The Sod2 mutant mouse, which is sensitized to mitochondrial stress, is an ideal mutant model for studying the role of oxidative stress in a diverse range of complications arising from mitochondrial dysfunction and diminished antioxidant defense. To fully appreciate the widespread molecular consequences under increased oxidative stress, a systems approach utilizing proteomics is able to provide a global overview of the complex biological changes, which a targeted single biomolecular approach cannot address fully. This review focuses on the applications of mass spectrometry and functional proteomics in the Sod2 mouse. The combinatorial approach provides novel insights into the interplay of chemistry and biology, free radicals and proteins, thereby augmenting our understanding of how redox perturbations influence protein dynamics. Ultimately, this knowledge can lead to the development of free radical-targeted therapies.


Assuntos
Mitocôndrias/metabolismo , Modelos Animais , Estresse Oxidativo/fisiologia , Proteoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Animais , Perfilação da Expressão Gênica/métodos , Humanos , Espectrometria de Massas/métodos , Camundongos , Camundongos Knockout , Modelos Biológicos , Mutação , Mapeamento de Peptídeos/métodos , Superóxido Dismutase/genética
9.
Chem Res Toxicol ; 23(5): 967-76, 2010 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-20405857

RESUMO

Nimesulide is a widely prescribed nitroaromatic sulfoanilide-type nonsteroidal anti-inflammatory drug that, despite its favorable safety profile, has been associated with rare cases of idiosyncratic drug-induced liver injury (DILI). Because reactive metabolites have been implicated in DILI, we aimed at investigating whether hepatic bioactivation of nimesulide produces a protein-reactive intermediate in hepatocytes. Also, we explored whether nimesulide can activate the transcription factor Nrf2 that would protect from drug-induced hepatocyte injury. We found that [(14)C]-nimesulide covalently bound to human liver microsomes (<50 pmol/mg under standard conditions) or immortalized human hepatocytes in a sulfaphenazole-sensitive, rifampicin-inducible manner; yet the overall extent of binding was modest. Although exposure of hepatocytes to nimesulide was not associated with increased net levels of superoxide anion, nimesulide (100 microM, 24 h) caused nuclear translocation of Nrf2 in a sulfaphenazole-sensitive manner, indicating a role of electrophilic metabolites. However, knockdown of Nrf2 with siRNA did not make the cells more sensitive to nimesulide-induced cell injury. Similarly, exposure of wild-type C57BL/6x129 Sv mice to nimesulide (100 mg/kg/day, po, for 5 days) was associated with nuclear translocation of immunoreactive Nrf2 in a small number of hepatocytes and induced >2-fold the expression levels of the Nrf2-target gene Nqo1 in wild-type but not Nrf2-null mice. Nimesulide administered to Nrf2(-/-) knockout mice did not cause increases in serum ALT activity or any apparent histopathological signs of liver injury. In conclusion, these data indicate that nimesulide is bioactivated by CYP2C to a protein-reactive electrophilic intermediate that activates the Nrf2 pathway even at nontoxic exposure levels.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Fígado/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Sulfonamidas/toxicidade , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Sistema Enzimático do Citocromo P-450/metabolismo , Hepatócitos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/genética , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo
10.
Am J Physiol Gastrointest Liver Physiol ; 297(5): G990-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20501447

RESUMO

Small intestinal ulceration, bleeding, and inflammation are major adverse effects associated with the use of diclofenac (DCF) or other nonsteroidal anti-inflammatory drugs (NSAIDs). The underlying mechanisms of DCF enteropathy are poorly understood, but there is increasing evidence that topical effects are involved. The aim of this study was to explore the role of c-Jun-N-terminal kinase (JNK) in DCF-induced enterocyte death because JNK not only regulates mitochondria-mediated apoptosis but also is a key node where many of the proximal stress signals converge. Male C57BL/6J mice were injected intraperitoneally with DCF or vehicle (Solutol HS-15), and the extent of small intestinal ulceration was determined. A single dose of DCF (60 mg/kg) produced numerous ulcers in the third and fourth quartiles of the jejunum and ileum, with maximal effects after 18 h and extensive recovery after 48 h. To study the molecular pathways leading to enterocyte injury, we isolated villi-enriched mucosal fractions from DCF-treated mice. Immunoblot studies with a phosphospecific JNK antibody revealed that JNK1/2 (p46) was activated at 6 h, leading to phosphorylation of the downstream target c-Jun. The levels of the JNK-regulated proapoptotic transcription factor C/EBP homologous protein (CHOP) were also increased after DCF. The selective JNK inhibitor SP600125 (30 mg/kg ip), given both 1 h before and 1 h after DCF, blocked JNK kinase activity and afforded significant protection against DCF enteropathy. In conclusion, these data demonstrate that the JNK pathway is critically involved in the pathogenesis of DCF-induced enteropathy and suggest a potential application of JNK inhibitors in the prevention of NSAID-induced enteropathy.


Assuntos
Antracenos/farmacologia , Diclofenaco/efeitos adversos , Enteropatias/induzido quimicamente , Enteropatias/prevenção & controle , Intestino Delgado/efeitos dos fármacos , Úlcera/induzido quimicamente , Úlcera/prevenção & controle , Fosfatase Alcalina/sangue , Fosfatase Alcalina/metabolismo , Animais , Antracenos/uso terapêutico , Apoptose/efeitos dos fármacos , Proteínas Sanguíneas/metabolismo , Diclofenaco/farmacologia , Enteropatias/patologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microvilosidades/efeitos dos fármacos , Microvilosidades/enzimologia , Microvilosidades/metabolismo , Mitocôndrias/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição CHOP/metabolismo , Úlcera/patologia
11.
Toxicol Appl Pharmacol ; 238(2): 150-9, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19442681

RESUMO

Flutamide, a widely used nonsteroidal anti-androgen, but not its bioisostere bicalutamide, has been associated with idiosyncratic drug-induced liver injury. Although the susceptibility factors are unknown, mitochondrial injury has emerged as a putative hazard of flutamide. To explore the role of mitochondrial sensitization in flutamide hepatotoxicity, we determined the effects of superimposed drug stress in a murine model of underlying mitochondrial abnormalities. Male wild-type or heterozygous Sod2(+/-) mice were injected intraperitoneously with flutamide (0, 30 or 100 mg/kg/day) for 28 days. A kinetic pilot study revealed that flutamide (100 mg/kg/day) caused approximately 10-fold greater exposure than the reported therapeutic mean plasma levels. Mutant (5/10), but not wild-type, mice in the high-dose group exhibited small foci of hepatocellular necrosis and an increased number of apoptotic hepatocytes. Hepatic GSSG/GSH, protein carbonyl levels, and serum lactate levels were significantly increased, suggesting oxidant stress and mitochondrial dysfunction. Measurement of mitochondrial superoxide in cultured hepatocytes demonstrated that mitochondria were a significant source of flutamide-enhanced oxidant stress. Indeed, mitochondria isolated from flutamide-treated Sod2(+/-) mice exhibited decreased aconitase activity as compared to vehicle controls. A transcriptomics analysis using MitoChips revealed that flutamide-treated Sod2(+/-) mice exhibited a selective decrease in the expression of all complexes I and III subunits encoded by mitochondrial DNA. In contrast, Sod2(+/-) mice receiving bicalutamide (50 mg/kg/day) did not reveal any hepatic changes. These results are compatible with our concept that flutamide targets hepatic mitochondria and exerts oxidant stress that can lead to overt hepatic injury in the presence of an underlying mitochondrial abnormality.


Assuntos
Antagonistas de Androgênios , Flutamida/toxicidade , Fígado/efeitos dos fármacos , Mitocôndrias/enzimologia , Superóxido Dismutase/metabolismo , Alanina Transaminase/efeitos dos fármacos , Alanina Transaminase/metabolismo , Antagonistas de Androgênios/toxicidade , Anilidas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Hepatócitos/patologia , Heterozigoto , Fígado/enzimologia , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Necrose/induzido quimicamente , Nitrilas/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/genética , Biologia de Sistemas , Compostos de Tosil/farmacologia
12.
Drug Discov Today ; 13(21-22): 982-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18762273

RESUMO

Mitochondria have been increasingly implicated in being a crucial subcellular target and amplifying oxidative injury induced by many drugs. Among the major cytoprotective antioxidants is the mitochondrial matrix protein, superoxide dismutase-2 (SOD2). Genetic modification of the expression of SOD2 by transgenic techniques or gene silencing has generated a number of distinct animal models with SOD2 deficiency including the heterozygous Sod2(+/-) knockout mouse model. These mice display a discreet underlying mitochondrial stress but are otherwise phenotypically normal and thus model a variety of clinically silent mitochondrial abnormalities. The model has found application in oxidative stress and age-related research, but it is only recently that it has been successfully used to study mechanisms of idiosyncratic drug-induced liver injury.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Mitocôndrias/fisiologia , Superóxido Dismutase/genética , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos
13.
Toxicol Sci ; 101(2): 341-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17975114

RESUMO

Although the mechanisms and susceptibility factors of troglitazone-associated idiosyncratic liver injury have not been elucidated, experimental evidence has identified oxidant stress and mitochondrial injury as a potential hazard in vitro. In search of upstream mediators of toxicity, we hypothesized that troglitazone-induced increased mitochondrial generation of superoxide might activate the thioredoxin-2 (Trx2)/apoptosis signal-regulating kinase 1 (Ask1) signaling pathway, leading to cell death, and that, hence, the mitochondrially targeted radical scavenger, mito-carboxy proxyl (CP), would prevent the increase in superoxide net levels and inhibit mitochondrial signaling and cell injury. Immortalized human hepatocytes (HC-04) were exposed to troglitazone (0-100 microM), which caused concentration and time-dependent apoptosis after 12-24 h (ketoconazole-insensitive). We found that troglitazone rapidly dissipated the mitochondrial inner transmembrane potential (DeltaPsi(m)) and independently increased the net levels of mitochondrial superoxide by 5-fold. This was followed by a shift of the redox ratio of mitochondrial Trx2 toward the oxidized state and subsequent activation of Ask1. Cell injury, but not the decrease in DeltaPsi(m), was prevented by cyclosporin A (3 microM), indicating that mitochondrial permeabilization, but not membrane depolarization, was causally involved in cell death. Mito-CP not only decreased troglitazone-induced superoxide levels but also prevented Trx2 oxidation and activation of Ask1 and protected cells from toxic injury. These data indicate that troglitazone, but not its oxidative metabolite(s), produce intramitochondrial oxidant stress that activates the Trx2/Ask1 pathway, leading to mitochondrial permeabilization. Furthermore, the data support our concept that targeted delivery of an antioxidant to mitochondria can inhibit upstream signaling and protect from troglitazone-induced lethal cell injury.


Assuntos
Cromanos/toxicidade , Hepatócitos/efeitos dos fármacos , Hipoglicemiantes/toxicidade , MAP Quinase Quinase Quinase 5/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Proteínas Mitocondriais/metabolismo , Superóxidos/metabolismo , Tiazolidinedionas/toxicidade , Tiorredoxinas/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Hepatócitos/enzimologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Hepáticas/enzimologia , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/patologia , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Troglitazona
15.
Drug Discov Today ; 12(7-8): 336-42, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17395094

RESUMO

Although the predictability of untoward drug effects in humans has improved in recent years, certain new drugs, with new pharmacological mechanisms, still pose a considerable challenge. This holds particularly true for biotherapeutics and their drug-related immune reactions, idiosyncratic drug hepatotoxicity and systemic toxicity. The selection of the 'right' animal models remains crucial; the species selected must be relevant (to humans) and sensitive with regard to three basic variables: pharmacodynamics, pharmacokinetics (including metabolism) and the mechanisms underlying the toxicity in the target human diseases. Furthermore, normal healthy animals might be a poor model in certain cases because the underlying disease in patients can be an important determinant of susceptibility to adverse effects. Therefore, we suggest that, where appropriate, new animal models of human disease (s) are introduced into drug safety assessment.


Assuntos
Anticorpos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Drogas em Investigação/farmacologia , Modelos Animais , Animais , Anticorpos/uso terapêutico , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/tendências , Drogas em Investigação/uso terapêutico , Humanos
16.
Toxicol Sci ; 97(1): 205-13, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17150972

RESUMO

Troglitazone, a first-generation thiazolidinedione antidiabetic drug, was withdrawn from the market due to an unacceptable risk of idiosyncratic hepatotoxicity. Troglitazone does not cause hepatotoxicity in normal healthy rodents, but it produces mitochondrial injury in vitro at high concentrations. The aim of this study was to explore whether genetic mitochondrial abnormalities might sensitize mice to hepatic adverse effects of troglitazone. We used heterozygous superoxide dismutase 2 (Sod2(+/-)) mice as a model of clinically silent mitochondrial stress. Troglitazone was daily administered for 4 weeks (0, 10 or 30 mg/kg/day, ip). We found that troglitazone caused overt liver injury in the high-dose group, manifested by increased serum alanine aminotransferase activity (> twofold) and midzonal areas of hepatic necrosis, in Sod2(+/-) but not in wild-type mice. No signs of hepatotoxicity were apparent at 2 weeks of treatment. Hepatic mitochondria isolated from troglitazone-treated mice exhibited decreased activities of aconitase (by 45%) and complex I (by 46%) and increased (by 58%) protein carbonyls, indicative of enhanced mitochondrial oxidant stress. This was paralleled by compensatory increases in mitochondrial glutathione levels. Finally, in hepatocytes isolated from untreated Sod2(+/-), but not wild-type mice, troglitazone caused a concentration-dependent increase in superoxide anion levels as demonstrated with a selective mitochondria-targeting fluorescent probe. In conclusion, prolonged administration of troglitazone can superimpose oxidant stress, potentiate mitochondrial damage, and induce delayed hepatic necrosis in mice with genetically compromised mitochondrial function. These data are consistent with our hypothesis that inherited or acquired mitochondrial abnormalities may be one of the contributing determinants of susceptibility to troglitazone-induced idiosyncratic liver injury.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Cromanos/toxicidade , Hipoglicemiantes/toxicidade , Fígado/efeitos dos fármacos , Mitocôndrias Hepáticas/efeitos dos fármacos , Doenças Mitocondriais/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Tiazolidinedionas/toxicidade , Aconitato Hidratase/metabolismo , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Complexo I de Transporte de Elétrons/metabolismo , Glutationa/metabolismo , Fígado/metabolismo , Fígado/patologia , Hepatopatias/genética , Hepatopatias/metabolismo , Hepatopatias/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/patologia , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia , Necrose , Carbonilação Proteica/efeitos dos fármacos , Superóxido Dismutase/deficiência , Superóxido Dismutase/genética , Superóxidos/metabolismo , Fatores de Tempo , Troglitazona
17.
Life Sci ; 81(25-26): 1659-67, 2007 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-17963788

RESUMO

Sulfotransferases (SULTs) play an important role in the detoxification and bioactivation of endogenous compounds and xenobiotics. Studies on rat sulfotransferases had shown that SULT genes, like cytochrome P450 genes, can be regulated by ligands that bind nuclear receptors. For human SULT genes, the regulation of human SULT2A1 expression is currently the best characterized. In this study, we systematically examined the regulation of human SULT1A genes by glucocorticoids. Treatment of the human hepatocellular carcinoma derived HepG2 cells with 10(-7) M dexamethasone did not affect the SULT1A1 activity toward p-nitrophenol. In contrast, SULT1A3 activity toward dopamine was significantly induced. Transient transfection of the SULT1A3 5'-flanking region/luciferase reporter construct showed that SULT1A3 was responsive to dexamethasone and prednisolone in a concentration-dependent manner with maximal induction at 10(-7) M dexamethasone or 1 microM prednisolone. In addition, induction by dexamethasone was dependent on the level of expression of the glucocorticoid receptor. Analysis of the 5'-flanking region led to the identification of a putative glucocorticoid response element at position (-1211 to -1193) upstream of the transcription start site and deletion or mutation of this element resulted in a loss of response. In summary, the data from this study shows that the human SULT1A3 gene is inducible by glucocorticoids through a glucocorticoid receptor-mediated mechanism and the glucocorticoid response element at position (-1211 to -1193) is necessary for this induction.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Glucocorticoides/metabolismo , Glucocorticoides/farmacologia , Sulfotransferases/genética , Animais , Dexametasona/metabolismo , Dexametasona/farmacologia , Hepatócitos/metabolismo , Humanos , Luciferases , Prednisolona/metabolismo , Prednisolona/farmacologia , Ratos , Receptores de Glucocorticoides/genética , Elementos de Resposta/genética , Sulfotransferases/efeitos dos fármacos , Transfecção , Células Tumorais Cultivadas
18.
Toxicol In Vitro ; 21(8): 1390-401, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17590308

RESUMO

In toxicological research, immortalized human hepatocytes provide a useful alternative to primary hepatocytes because interindividual variability in the expression of drug-metabolizing enzymes and drug transporters can largely be eliminated. However, it is essential that the cell line retain the original phenotype. The purpose of this study was to characterize a novel spontaneously immortalized human hepatocyte cell line, HC-04, with respect to the transcript and functional protein expression profile for the major drug-metabolizing enzymes and transmembrane transporters. HC-04 cells retained hepatocyte-specific function including albumin production and ornithine transcarbamoylase and glucose-6-phosphatase activity. Most of the major CYP forms were expressed at basal levels and responsive to inducing agents. In particular, CYP3A4 was expressed abundantly, and HC-04 cells were able to metabolize the CYP3A4 probe, midazolam, at a rate similar to primary human hepatocytes. Furthermore, the major human sulfotransferase and UDP-glucuronosyltransferase forms, as well as members of the ABC and SLC transporter superfamilies, nuclear receptors, and hepatic transcription factors were also expressed. HC-04 cells readily responded to standard hepatotoxicants that are dependent on CYP-mediated bioactivation, while another, tumor-derived cell line remained refractory to the drug challenge. Collectively, HC-04 cells provide a reliable, stable, and reproducible model for biomechanistic studies in drug toxicology.


Assuntos
Proteínas de Transporte/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Acetaminofen/metabolismo , Analgésicos não Narcóticos/metabolismo , Anti-Inflamatórios não Esteroides/metabolismo , Antifibrinolíticos/metabolismo , Biomarcadores , Proteínas de Transporte/genética , Linhagem Celular , Sistema Enzimático do Citocromo P-450/genética , Diclofenaco/metabolismo , Humanos , Vitamina K 3/metabolismo
19.
Curr Drug Metab ; 7(7): 715-27, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17073576

RESUMO

Certain drugs containing a nitroaromatic moiety (e.g., tolcapone, nimesulide, nilutamide, flutamide, nitrofurantoin) have been associated with organ-selective toxicity including rare cases of idiosyncratic liver injury. What they have in common is the potential for multistep nitroreductive bioactivation (6-electron transfer) that produces the potentially hazardous nitroanion radical, nitroso intermediate, and N-hydroxy derivative. These intermediates have been associated with increased oxidant stress and targeting of nucleophilic residues on proteins and nucleic acids. However, other mechanisms including the formation of oxidative metabolites and mitochondrial liability, as well as inherent toxicokinetic properties, also determine the drugs' overall potency. Therefore, structural modification not only of the nitro moiety but also of ring substituents can greatly reduce toxicity. Novel concepts have revealed that, besides the classical microsomal nitroreductases, cytosolic and mitochondrial enzymes including nitric oxide synthase can also bioactivate certain nitroarenes (nilutamide). Furthermore, animal models of silent mitochondrial dysfunction have demonstrated that a mitochondrial oxidant stress posed by certain nitroaromatic drugs (nimesulide) can produce significant mitochondrial injury if superimposed on a genetic mitochondrial abnormality. Finally, there may be mechanisms for all nitroaromatic drugs that do not involve bioactivation of the nitro group, e.g., AHR interactions with flutamide. Taken together, the focus of research on the hepatic toxicity of nitroarene-containing drugs has shifted over the past years from the identification of the reactive intermediates generated during the bioreductive pathway to the underlying biomechanisms of liver injury. Most likely one of the next paradigm shifts will include the identification of determinants of susceptibility to nitroaromatic drug-induced hepatotoxicity.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/etiologia , Nitrocompostos/efeitos adversos , Nitrocompostos/farmacocinética , Benzofenonas/efeitos adversos , Benzofenonas/farmacocinética , Biotransformação , Flutamida/efeitos adversos , Flutamida/farmacocinética , Imidazolidinas/efeitos adversos , Imidazolidinas/farmacocinética , Nifedipino/efeitos adversos , Nifedipino/farmacocinética , Nitrofurantoína/efeitos adversos , Nitrofurantoína/farmacocinética , Nitrofenóis/efeitos adversos , Nitrofenóis/farmacocinética , Sulfonamidas/efeitos adversos , Sulfonamidas/farmacocinética , Tolcapona
20.
Free Radic Biol Med ; 40(3): 420-9, 2006 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16443156

RESUMO

Nimesulide, a preferential COX-2 inhibitor, has been associated with rare idiosyncratic hepatotoxicity. The underlying mechanisms of liver injury are unknown, but experimental evidence has identified oxidative stress as a potential hazard and mitochondria as a target. The aim of this study was to explore whether genetic mitochondrial abnormalities, resulting in impaired mitochondrial function and mildly increased oxidative stress, might sensitize mice to the hepatic adverse effects of nimesulide. We used heterozygous superoxide dismutase 2 (Sod2(+/-)) mice as a model, as these mice develop clinically silent mitochondrial stress but otherwise appear normal. Nimesulide was administered for 4 weeks (10 mg/kg, ip, bid), at a dose equivalent to human therapeutic dosage. We found that the drug potentiated hepatic mitochondrial oxidative injury (decreased aconitase activity, increased protein carbonyls) in Sod2(+/-), but not wild-type, mice. Furthermore, the nimesulide-treated mutant mice exhibited increased hepatic cytosolic levels of cytochrome c and caspase-3 activity, as well as increased numbers of apoptotic hepatocytes. Finally, nimesulide in vitro caused a concentration-dependent net increase in superoxide anion in mitochondria from Sod2(+/-), but not Sod2(+/+) mice. In conclusion, repeated administration of nimesulide can superimpose an oxidant stress, potentiate mitochondrial damage, and activate proapoptotic factors in mice with genetically compromised mitochondrial function.


Assuntos
Apoptose/efeitos dos fármacos , Heterozigoto , Mitocôndrias Hepáticas/efeitos dos fármacos , Estresse Oxidativo , Sulfonamidas/farmacologia , Superóxido Dismutase/fisiologia , Aconitato Hidratase/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Caspases/metabolismo , Citocromos c/metabolismo , Citosol/metabolismo , Feminino , Glutationa/metabolismo , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/metabolismo , Superóxido Dismutase/genética , Superóxidos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA