Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Toxicol Sci ; 42(4): 427-436, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28717101

RESUMO

Cholestasis is one of the major causes of drug-induced liver injury (DILI), which can result in withdrawal of approved drugs from the market. Early identification of cholestatic drugs is difficult due to the complex mechanisms involved. In order to develop a strategy for mechanism-based risk assessment of cholestatic drugs, we analyzed gene expression data obtained from the livers of rats that had been orally administered with 12 known cholestatic compounds repeatedly for 28 days at three dose levels. Qualitative analyses were performed using two statistical approaches (hierarchical clustering and principle component analysis), in addition to pathway analysis. The transcriptional benchmark dose (tBMD) and tBMD 95% lower limit (tBMDL) were used for quantitative analyses, which revealed three compound sub-groups that produced different types of differential gene expression; these groups of genes were mainly involved in inflammation, cholesterol biosynthesis, and oxidative stress. Furthermore, the tBMDL values for each test compound were in good agreement with the relevant no observed adverse effect level. These results indicate that our novel strategy for drug safety evaluation using mechanism-based classification and tBMDL would facilitate the application of toxicogenomics for risk assessment of cholestatic DILI.


Assuntos
Clorpromazina/administração & dosagem , Clorpromazina/toxicidade , Colestase/induzido quimicamente , Ciclosporina/administração & dosagem , Ciclosporina/toxicidade , Diclofenaco/administração & dosagem , Diclofenaco/toxicidade , Medição de Risco/métodos , Toxicogenética/métodos , Administração Oral , Animais , Colesterol/biossíntese , Relação Dose-Resposta a Droga , Flutamida/administração & dosagem , Flutamida/toxicidade , Expressão Gênica , Humanos , Imipramina/administração & dosagem , Imipramina/toxicidade , Inflamação/genética , Cetoconazol/administração & dosagem , Cetoconazol/toxicidade , Fígado , Metiltestosterona/administração & dosagem , Metiltestosterona/toxicidade , Estresse Oxidativo/genética , Ratos , Sulindaco/administração & dosagem , Sulindaco/toxicidade , Tamoxifeno/administração & dosagem , Tamoxifeno/toxicidade
2.
J Neurosci Res ; 91(3): 393-406, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23280445

RESUMO

Malignant gliomas have low survival expectations regardless of current treatments. Nonsteroidal anti-inflammatory drugs (NSAIDs) prevent cell transformation and slow cancer cell growth by mechanisms independent of cyclooxygenase (COX) inhibition. Certain NSAIDs trigger the endoplasmic reticulum stress response (ERSR), as revealed by upregulation of molecular chaperones such as GRP78 and C/EBP homologous protein (CHOP). Although celecoxib (CELE) inhibits the sarcoendoplasmic reticulum Ca(2+) ATPase (SERCA), an effect known to induce ERSR, sulindac sulfide (SS) has not been reported to affect SERCA. Here, we investigated these two drugs for their effects on Ca(2+) homeostasis, ERSR, and glioma cell survival. Our findings indicate that SS is a reversible inhibitor of SERCA and that both SS and CELE bind SERCA at its cyclopiazonic acid binding site. Furthermore, CELE releases additional Ca(2+) from the mitochondria. In glioma cells, both NSAIDS upregulate GRP78 and activate ER-associated caspase-4 and caspase-3. Although only CELE upregulates the expression of CHOP, it appears that CHOP induction could be associated with mitochondrial poisoning. In addition, CHOP induction appears to be uncorrelated with the gliotoxicity of these NSAIDS in our experiments. Our data suggest that activation of ERSR is primarily responsible for the gliotoxic effect of these NSAIDS. Because SS has good brain bioavailability, has lower COX-2 inhibition, and has no mitochondrial effects, it represents a more appealing molecular candidate than CELE to achieve gliotoxicity via activation of ERSR.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Glioma/metabolismo , Pirazóis/toxicidade , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , Sulfonamidas/toxicidade , Sulindaco/análogos & derivados , Celecoxib , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/fisiologia , Inibidores Enzimáticos/toxicidade , Glioma/enzimologia , Humanos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/química , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Sulindaco/toxicidade
3.
Toxicol In Vitro ; 26(2): 197-205, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22138569

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used in the treatment of pain and inflammation. However, this group of drugs is associated with serious adverse drug reactions. Previously, we studied the mechanisms underlying toxicity of the NSAID diclofenac using Saccharomycescerevisiae as model system. We identified the involvement of several mitochondrial proteins, a transporter and cytochrome P450 activity in diclofenac toxicity. In this study, we investigated if these processes are also involved in the toxicity of other NSAIDs. We divided the NSAIDs into three classes based on their toxicity mechanisms. Class I consists of diclofenac, indomethacin and ketoprofen. Mitochondrial respiration and reactive oxygen species (ROS) play a major role in the toxicity of this class. Metabolism by cytochrome P450s further increases their toxicity, while ABC-transporters decrease the toxicity. Mitochondria and oxidative metabolism also contribute to toxicity of class II drugs ibuprofen and naproxen, but another cellular target dominates their toxicity. Interestingly, ibuprofen was the only NSAID that was unable to induce upregulation of the multidrug resistance response. The class III NSAIDs sulindac, ketorolac and zomepirac were relatively non-toxic in yeast. In conclusion, we demonstrate the use of yeast to investigate the mechanisms underlying the toxicity of structurally related drugs.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Leveduras/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/metabolismo , Transporte Biológico Ativo/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Diclofenaco/toxicidade , Transporte de Elétrons , Genes MDR/efeitos dos fármacos , Ibuprofeno/toxicidade , Indometacina/toxicidade , Cetoprofeno/toxicidade , Cetorolaco/toxicidade , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Naproxeno/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Sulindaco/toxicidade , Tolmetino/análogos & derivados , Tolmetino/toxicidade , Leveduras/crescimento & desenvolvimento , Leveduras/metabolismo
4.
Toxicology ; 290(2-3): 278-85, 2011 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-22019926

RESUMO

Previous studies indicated that lipopolysaccharide (LPS) interacts with the nonsteroidal anti-inflammatory drug sulindac (SLD) to produce liver injury in rats. In the present study, the mechanism of SLD/LPS-induced liver injury was further investigated. Accumulation of polymorphonuclear neutrophils (PMNs) in the liver was greater in SLD/LPS-cotreated rats compared to those treated with SLD or LPS alone. In addition, PMN activation occurred specifically in livers of rats cotreated with SLD/LPS. The hypothesis that PMNs and proteases released from them play critical roles in the hepatotoxicity was tested. SLD/LPS-induced liver injury was attenuated by prior depletion of PMNs or by treatment with the PMN protease inhibitor, eglin C. Previous studies suggested that tumor necrosis factor-α (TNF) and the hemostatic system play critical roles in the pathogenesis of liver injury induced by SLD/LPS. TNF and plasminogen activator inhibitor-1 (PAI-1) can contribute to hepatotoxicity by affecting PMN activation and fibrin deposition. Therefore, the role of TNF and PAI-1 in PMN activation and fibrin deposition in the SLD/LPS-induced liver injury model was tested. Neutralization of TNF or inhibition of PAI-1 attenuated PMN activation. TNF had no effect on PAI-1 production or fibrin deposition. In contrast, PAI-1 contributed to fibrin deposition in livers of rats treated with SLD/LPS. In summary, PMNs, TNF and PAI-1 contribute to the liver injury induced by SLD/LPS cotreatment. TNF and PAI-1 independently contributed to PMN activation, which is critical to the pathogenesis of liver injury. Moreover, PAI-1 contributed to liver injury by promoting fibrin deposition.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Lipopolissacarídeos/toxicidade , Neutrófilos/efeitos dos fármacos , Sulindaco/toxicidade , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/metabolismo , Fibrina/metabolismo , Masculino , Neutrófilos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Proteínas/farmacologia , Coelhos , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
5.
Br J Pharmacol ; 162(7): 1521-33, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21175575

RESUMO

BACKGROUND AND PURPOSE: The use of non-steroidal anti-inflammatory drugs (NSAIDs) in the treatment of rheumatoid arthritis (RA) is limited by their toxicity. We evaluated the anti-inflammatory efficacy and safety of three novel modified NSAIDs, phospho-aspirin, phospho-ibuprofen and phospho-sulindac. EXPERIMENTAL APPROACH: We determined the anti-inflammatory effects and gastrointestinal safety of the phospho-NSAIDs in the rat adjuvant arthritis model and studied their mechanism of action in cultured cells, Cytokines were measured with elisa and activation of nuclear factor-κB (NF-κB) by immunohistochemistry. KEY RESULTS: All three phospho-NSAIDs showed less gastrointestinal toxicity than their parent compounds and demonstrated strong anti-inflammatory effects, essentially reversing joint inflammation and oedema. They have a broad but not uniform effect on the expression of relevant cytokines, in general decreasing IL-6 and IL-1ß and increasing IL-10 levels in rat plasma and cultured cells. Phospho-sulindac and phospho-ibuprofen but not phospho-aspirin suppressed PGE(2) production in vitro, whereas phospho-aspirin (in contrast to aspirin) showed the same effect in vivo. In joint tissues, phospho-aspirin inhibited NF-κB activation, and suppressed inflammation and bone resorption. Phospho-aspirin also inhibited Jurkat T cell proliferation. In general, phospho-aspirin had greater efficacy but different effects upon inflammatory mediators compared with aspirin. The chemical modification of the parent NSAIDs seems crucial for their safety and efficacy. CONCLUSIONS AND IMPLICATIONS: Phospho-aspirin, phospho-ibuprofen and phospho-sulindac were safer than their parent NSAIDs, were highly effective in rat adjuvant arthritis and inhibited many key mediators in the pathophysiology of RA. These novel compounds are promising candidate drugs for the treatment of RA and merit further evaluation.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Artrite Experimental/tratamento farmacológico , Animais , Anti-Inflamatórios não Esteroides/toxicidade , Artrite Experimental/imunologia , Aspirina/análogos & derivados , Aspirina/farmacologia , Aspirina/toxicidade , Linhagem Celular , Citocinas/biossíntese , Citocinas/genética , Dinoprostona/antagonistas & inibidores , Dinoprostona/biossíntese , Dinoprostona/metabolismo , Gastroenteropatias/induzido quimicamente , Humanos , Ibuprofeno/análogos & derivados , Ibuprofeno/farmacologia , Ibuprofeno/toxicidade , Células Jurkat , Camundongos , NF-kappa B/metabolismo , Células NIH 3T3 , Organofosfatos/farmacologia , Organofosfatos/toxicidade , Compostos Organofosforados/farmacologia , Compostos Organofosforados/toxicidade , Ratos , Sulindaco/análogos & derivados , Sulindaco/farmacologia , Sulindaco/toxicidade
6.
J Pharmacol Exp Ther ; 334(3): 693-702, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20498252

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) such as sulindac and indomethacin are a major cause of gastric erosions and ulcers. Induction of apoptosis by NSAIDs is an important mechanism involved. Understanding how NSAIDs affect genes that regulate apoptosis is useful for designing therapeutic or preventive strategies and for evaluating the efficacy of safer drugs being developed. We investigated whether growth arrest and DNA damage-inducible 45alpha (GADD45alpha), a stress signal response gene involved in regulation of DNA repair and induction of apoptosis, plays a part in NSAID-induced gastric mucosal injury and apoptosis in vivo in mice and in vitro in cultured human AGS and rat RGM-1 gastric epithelial cells. Intraperitoneal administration of sulindac and indomethacin both resulted in up-regulation of GADD45alpha expression and induction of significant injury and apoptosis in gastric mucosa of wild-type mice. GADD45alpha(-/-) mice were markedly more resistant to both sulindac- and indomethacin-induced gastric mucosal injury and apoptosis than wild-type mice. Sulindac sulfide and indomethacin treatments also concentration-dependently increased GADD45alpha expression and apoptosis in AGS and RGM-1 cells. Antisense suppression of GADD45alpha expression significantly reduced sulindac and indomethacin-induced activation of caspase-9 and apoptosis in AGS cells. Pretreatments with exogenous prostaglandins and small interfering RNA suppression of cyclooxygenase (COX)-1 and -2 did not affect up-regulation of GADD45alpha by sulindac sulfide and indomethacin in AGS cells. These findings indicate that GADD45alpha up-regulation is a COX-independent mechanism that is required for induction of severe gastric mucosal apoptosis and injury by NSAIDs, probably via a capase-9-dependent pathway of programmed cell death.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Proteínas de Ciclo Celular/antagonistas & inibidores , Mucosa Gástrica/patologia , Indometacina/toxicidade , Proteínas Nucleares/antagonistas & inibidores , Úlcera Gástrica/patologia , Úlcera Gástrica/prevenção & controle , Sulindaco/toxicidade , Animais , Elementos Antissenso (Genética)/farmacologia , Apoptose/efeitos dos fármacos , Caspase 9/metabolismo , Inibidores de Caspase , Proteínas de Ciclo Celular/biossíntese , Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 1/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Dano ao DNA , Dinoprostona/farmacologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/biossíntese , RNA Interferente Pequeno/farmacologia , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Úlcera Gástrica/induzido quimicamente , Regulação para Cima
7.
Toxicology ; 272(1-3): 32-8, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20371263

RESUMO

Among currently prescribed nonsteroidal anti-inflammatory drugs, sulindac (SLD) is associated with the greatest incidence of idiosyncratic hepatotoxicity in humans. Previously, an animal model of SLD-induced idiosyncratic hepatotoxicity was developed by cotreating rats with a nonhepatotoxic dose of LPS. Tumor necrosis factor-alpha (TNF) was found to be critically important to the pathogenesis. In this study, the mechanism of liver injury induced by SLD/LPS cotreatment was further explored. Protein carbonyls, products of oxidative stress, were elevated in liver mitochondria of SLD/LPS-cotreated rats. The results of analyzing gene expression in livers of rats before the onset of liver injury indicated that genes associated with oxidative stress were selectively regulated by SLD/LPS cotreatment. Antioxidant treatment with either ebselen or dimethyl sulfoxide attenuated SLD/LPS-induced liver injury. The role of oxidative stress was further investigated in vitro. SLD sulfide, the toxic metabolite of SLD, enhanced TNF-induced cytotoxicity and caspase 3/7 activity in HepG2 cells. SLD sulfide also increased dichlorofluorescein fluorescence, suggesting generation of reactive oxygen species (ROS). Hydrogen peroxide and TNF cotreatment of HepG2 cells caused greater cytotoxicity than either treatment alone. Either antioxidant tempol or a pancaspase inhibitor Z-VAD-FMK decreased cell death as well as caspase 3/7 activity induced by SLD sulfide/TNF coexposure. These results indicate that SLD/LPS treatment causes oxidative stress in livers of rats and suggest that ROS are important in SLD/LPS-induced liver injury in vivo. Furthermore, ROS contribute to the cytotoxic interaction of SLD and TNF by activating caspase 3/7.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Lipopolissacarídeos/metabolismo , Fígado/patologia , Estresse Oxidativo/genética , Sulindaco/metabolismo , Clorometilcetonas de Aminoácidos/antagonistas & inibidores , Animais , Anti-Inflamatórios não Esteroides/toxicidade , Linhagem Celular Tumoral , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Humanos , Lipopolissacarídeos/toxicidade , Fígado/metabolismo , Masculino , Mitocôndrias Hepáticas/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Sulindaco/toxicidade , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
8.
J Pharmacol Exp Ther ; 331(1): 114-21, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19638570

RESUMO

Sulindac (SLD) is a nonsteroidal anti-inflammatory drug (NSAID) that has been associated with a greater incidence of idiosyncratic hepatotoxicity in human patients than other NSAIDs. In previous studies, cotreatment of rats with SLD and a modestly inflammatory dose of lipopolysaccharide (LPS) led to liver injury, whereas neither SLD nor LPS alone caused liver damage. In studies presented here, further investigation of this animal model revealed that the concentration of tumor necrosis factor-alpha (TNF-alpha) in plasma was significantly increased by LPS at 1 h, and SLD enhanced this response. Etanercept, a soluble TNF-alpha receptor, reduced SLD/LPS-induced liver injury, suggesting a role for TNF-alpha. SLD metabolites in plasma and liver were determined by LC/MS/MS. Cotreatment with LPS did not increase the concentrations of SLD or its metabolites, excluding the possibility that LPS contributed to liver injury through enhanced exposure to SLD or its metabolites. The cytotoxicities of SLD and its sulfide and sulfone metabolites were compared in primary rat hepatocytes and HepG2 cells; SLD sulfide was more toxic in both types of cells than SLD or SLD sulfone. TNF-alpha augmented the cytotoxicity of SLD sulfide in primary hepatocytes and HepG2 cells. These results suggest that TNF-alpha can enhance SLD sulfide-induced hepatotoxicity, thereby contributing to liver injury in SLD/LPS-cotreated rats.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/sangue , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Sulindaco/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Animais , Linhagem Celular Tumoral , Doença Hepática Induzida por Substâncias e Drogas/patologia , Sinergismo Farmacológico , Humanos , Mediadores da Inflamação/sangue , Mediadores da Inflamação/toxicidade , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/toxicidade , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Ratos , Ratos Sprague-Dawley , Sulindaco/toxicidade , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/sangue
9.
Cancer Prev Res (Phila) ; 2(6): 572-80, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19470791

RESUMO

Nonsteroidal anti-inflammatory drugs such as sulindac have shown promising antineoplastic activity, although toxicity from cyclooxygenase (COX) inhibition and the suppression of prostaglandin synthesis limits their use for chemoprevention. Previous studies have concluded that the mechanism responsible for their antineoplastic activity may be COX independent. To selectively design out the COX inhibitory activity of sulindac sulfide (SS), in silico modeling studies were done that revealed the crucial role of the carboxylate moiety for COX-1 and COX-2 binding. These studies prompted the synthesis of a series of SS derivatives with carboxylate modifications that were screened for tumor cell growth and COX inhibitory activity. A SS amide (SSA) with a N,N-dimethylethyl amine substitution was found to lack COX-1 and COX-2 inhibitory activity, yet potently inhibit the growth of human colon tumor cell lines, HT-29, SW480, and HCT116 with IC(50) values of 2 to 5 micromol/L compared with 73 to 85 micromol/L for SS. The mechanism of growth inhibition involved the suppression of DNA synthesis and apoptosis induction. Oral administration of SSA was well-tolerated in mice and generated plasma levels that exceeded its in vitro IC(50) for tumor growth inhibition. In the human HT-29 colon tumor xenograft mouse model, SSA significantly inhibited tumor growth at a dosage of 250 mg/kg. Combined treatment of SSA with the chemotherapeutic drug, Camptosar, caused a more sustained suppression of tumor growth compared with Camptosar treatment alone. These results indicate that SSA has potential safety and efficacy advantages for colon cancer chemoprevention as well as utility for treating malignant disease if combined with chemotherapy.


Assuntos
Adenocarcinoma/patologia , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias do Colo/patologia , Animais , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/enzimologia , Linhagem Celular Tumoral/transplante , Ciclo-Oxigenase 1/química , Ciclo-Oxigenase 1/efeitos dos fármacos , Ciclo-Oxigenase 2/química , Ciclo-Oxigenase 2/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Inibidores de Ciclo-Oxigenase/toxicidade , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Irinotecano , Masculino , Dose Máxima Tolerável , Camundongos , Camundongos Nus , Modelos Moleculares , Proteínas de Neoplasias/análise , Conformação Proteica , Sulindaco/administração & dosagem , Sulindaco/análogos & derivados , Sulindaco/síntese química , Sulindaco/farmacocinética , Sulindaco/farmacologia , Sulindaco/uso terapêutico , Sulindaco/toxicidade , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Toxicol Sci ; 108(1): 184-93, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19074762

RESUMO

Sulindac (SLD) is a nonsteroidal anti-inflammatory drug (NSAID) that has been associated with a greater incidence of idiosyncratic hepatotoxicity in human patients than other NSAIDs. One hypothesis regarding idiosyncratic adverse drug reactions is that interaction of a drug with a modest inflammatory episode precipitates liver injury. In this study, we tested the hypothesis that lipopolysaccharide (LPS) interacts with SLD to cause liver injury in rats. SLD (50 mg/kg) or its vehicle was administered to rats by gavage 15.5 h before LPS (8.3 x 10(5) endotoxin unit/kg) or its saline vehicle (i.v.). Thirty minutes after LPS treatment, SLD or vehicle administration was repeated. Rats were killed at various times after treatment, and serum, plasma, and liver samples were taken. Neither SLD nor LPS alone caused liver injury. Cotreatment with SLD/LPS led to increases in serum biomarkers of both hepatocellular injury and cholestasis. Histological evidence of liver damage was found only after SLD/LPS cotreatment. As a result of activation of hemostasis induced by SLD/LPS cotreatment, fibrin and hypoxia were present in liver tissue before the onset of hepatotoxicity. Heparin treatment reduced hepatic fibrin deposition and hypoxia and protected against liver injury induced by SLD/LPS cotreatment. These results indicate that cotreatment with nontoxic doses of LPS and SLD causes liver injury in rats, and this could serve as a model of human idiosyncratic liver injury. The hemostatic system is activated by SLD/LPS cotreatment and plays an important role in the development of SLD/LPS-induced liver injury.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Hemostasia/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Sulindaco/toxicidade , Análise de Variância , Animais , Colestase/metabolismo , Relação Dose-Resposta a Droga , Fibrina/metabolismo , Heparina/farmacologia , Hipóxia/metabolismo , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Ratos , Sulindaco/administração & dosagem , Sulindaco/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
Life Sci ; 79(24): 2269-74, 2006 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-16952380

RESUMO

Hepatic tryptophan 2,3-dioxygenase (TDO) is one of the rate-limiting enzymes in tryptophan catabolism and plays an important role in regulating the physiological flux of tryptophan into relevant metabolic pathways. In this study, we determined the effect of the non-steroidal anti-inflammatory agents, tolmetin and sulindac, on rat liver TDO activity and the subsequent changes in the hippocampal and striatal neurotransmitter levels. The amount of melatonin produced by the pineal gland was also measured using high performance liquid chromatography (HPLC). Treatment of rats with tolmetin or sulindac (5 mg/kg/bd for 5 days) significantly inhibited liver TDO activity. The results show that whilst tolmetin and sulindac increase serotonin levels in the hippocampus, these agents also significantly reduce dopamine levels in the striatum. Tolmetin, but not sulindac, increased the amount of melatonin produced by the pineal gland. The results of this study suggest that whilst tolmetin and sulindac may be beneficial for patients suffering from depression, these agents also have the potential to induce adverse effects in patients suffering with neurological disorders such as Parkinson's disease.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Inibidores Enzimáticos/toxicidade , Fígado/efeitos dos fármacos , Sulindaco/toxicidade , Tolmetino/toxicidade , Triptofano Oxigenase/antagonistas & inibidores , Animais , Cromatografia Líquida de Alta Pressão , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/enzimologia , Dopamina/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Fígado/enzimologia , Masculino , Melatonina/análise , Melatonina/metabolismo , Glândula Pineal/química , Glândula Pineal/efeitos dos fármacos , Glândula Pineal/metabolismo , Ratos , Ratos Wistar , Serotonina/metabolismo , Triptofano Oxigenase/metabolismo
12.
Mol Cancer Ther ; 5(5): 1190-6, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16731751

RESUMO

The nonsteroidal anti-inflammatory drug sulindac is metabolized to sulindac sulfone (exisulind), an antineoplastic agent that inhibits growth and induces apoptosis in solid tumors. In colon cancer cells, the antineoplastic effects of exisulind have been attributed, in part, to induction of cyclic guanosine 3',5'-monophosphate (cGMP) signaling through inhibition of cGMP-specific phosphodiesterases, which elevates intracellular cGMP, and novel expression of cGMP-dependent protein kinase (PKG) Ibeta, the presumed downstream effector mediating apoptosis. Here, inhibition of proliferation and induction of cell death by exisulind was dissociated from cGMP signaling in human colon cancer cells. Accumulation of intracellular cGMP produced by an exogenous cell-permeant analogue of cGMP or a potent agonist of guanylyl cyclase C yielded cytostasis without cell death. Surprisingly, the antiproliferative effects of induced cGMP accumulation were paradoxically less than additive, rather than synergistic, when combined with exisulind. Further, although exisulind induced expression of PKG Ibeta, it did not elevate intracellular cGMP and its efficacy was not altered by inhibition or activation of PKG I. Rather, PKG I induced by exisulind may mediate desensitization of cytostasis induced by cGMP. Thus, cytotoxic effects of exisulind are independent of cGMP signaling in human colon cancer cells. Moreover, combination therapies, including exisulind and agents that induce cGMP signaling, may require careful evaluation in patients with colon cancer.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/metabolismo , Guanilato Ciclase/farmacologia , Transdução de Sinais , Sulindaco/análogos & derivados , Células CACO-2 , Morte Celular , Proliferação de Células/efeitos dos fármacos , GMP Cíclico/metabolismo , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Modelos Biológicos , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase , Receptores de Peptídeos , Transdução de Sinais/efeitos dos fármacos , Sulindaco/farmacologia , Sulindaco/toxicidade , Células Tumorais Cultivadas
13.
J Biol Chem ; 280(31): 28230-40, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-15946948

RESUMO

Expression of the gamma-glutamylcysteine synthetase heavy subunit (gamma-GCSh), which encodes the rate-limiting enzymes for glutathione biosynthesis, is regulated by many cytotoxic agents. Moreover, gamma-GCSh mRNA expression is elevated in colorectal cancer, but how gamma-GCSh expression is regulated is not completely understood. By using actinomycin D, which inhibits new RNA synthesis, we showed that treatment of human colorectal cancer cells with the prooxidant sulindac increased the half-life of gamma-GCSh mRNA. By using a tetracycline-regulated gamma-GCSh mRNA assay system, we systematically dissected the cis-acting sequence and trans-acting factors that regulate the stability of gamma-GCSh by cytotoxic prooxidants. We demonstrated that a HuR recognition sequence, AUUUA, in the 3'-untranslated region is responsible for the decay of gamma-GCSh mRNA. Oxidative stress enhanced cytoplasmic content of HuR. Overexpression of HuR by transfection stabilized gamma-GCSh mRNA, whereas overexpression of a dominant-negative HuR mutant suppressed the induced stability. Furthermore, prooxidant-induced gamma-GCSh mRNA stabilization and HuR binding were blocked by p38 mitogen-activated protein kinase inhibitors. We provide the first evidence that reduction-oxidation regulation of gamma-GCSh expression, itself a reduction-oxidation sensor and regulator, is mediated at least in part by the p38 mitogen-activated protein kinase signaling through the HuR RNA-binding protein.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glutamato-Cisteína Ligase/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Estresse Oxidativo/fisiologia , RNA Mensageiro/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais , Meia-Vida , Humanos , Oxidantes/toxicidade , Subunidades Proteicas/genética , Proteínas Recombinantes/metabolismo , Sulindaco/toxicidade
14.
Oncogene ; 24(12): 2104-9, 2005 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-15688007

RESUMO

P21WAF1/cip1 plays a critical role in regulating intestinal cell proliferation, maturation and tumorigenesis. Our previous work demonstrated that inactivation of a single p21 allele in Apc1638+/- mice was sufficient to accelerate Apc-initiated tumor formation, and that inactivation of only one p21 allele was also sufficient to abrogate duodenal tumor inhibition by sulindac, a nonsteroidal anti-inflammatory drug. To dissect the role of p21 in sulindac inhibition of intestinal tumor development in Apc1638+/- mice, we quantified p21 expression from Apc+/-, p21+/+, +/- or -/- mice fed sulindac. In Apc+/-, p21 wild-type mice fed the sulindac supplemental diet, both p21 mRNA and protein were significantly increased in the flat mucosa and tumors of the duodenum. However, p21 was not induced by sulindac in the duodenal flat mucosa and tumors of Apc+/- mice in which one or both p21 alleles had been inactivated. Further investigation revealed that the cytosine residues in a CpG cluster in the promoter region of the mouse p21 gene displayed hypermethylation in the Apc+/-, p21+/- mice. This suggested that although the p21+/- mice retained a wild-type allele, this allele was functionally modulated by hypermethylation, and that the inability of sulindac to inhibit tumor formation in Apc+/-, p21+/- mice is likely due to the inability to induce expression of the wild-type, but differentially methylated, p21 allele.


Assuntos
Antineoplásicos/toxicidade , Proteínas de Ciclo Celular/genética , Metilação de DNA , Regiões Promotoras Genéticas , Sulindaco/toxicidade , Animais , Inibidor de Quinase Dependente de Ciclina p21 , Fosfatos de Dinucleosídeos/genética , Deleção de Genes , Genes APC , Camundongos , Camundongos Knockout , RNA Mensageiro/genética
15.
Mol Cancer Ther ; 4(1): 51-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15657353

RESUMO

The nonsteroidal anti-inflammatory drug (NSAID) sulindac prevents experimental colon cancer and can regress precancerous polyps in humans. Sulindac sulfide inhibits cyclooxygenase (COX)-mediated prostaglandin synthesis and retards the growth of cultured colon cell lines primarily by inducing apoptosis. Given the known role of mitogen-activated protein kinase (MAPK) in signal transduction and the regulation of cell survival and death, we determined the effect of sulindac sulfide on MAPK activation, COX-2 expression, and apoptosis induction in HCA-7 human colon cancer cells. Sulindac sulfide treatment was associated with activation of ERKp44/42 and p38 MAPK in a dosage- and time-dependent manner, and also activated upstream MEK. Similar results were seen in HCT-15 cells and also with the selective COX-2 inhibitor NS398. ERKp44/42 and p38 activation were accompanied by an induction of COX-2 protein expression. Selective inhibitors of sulindac sulfide-induced ERKp44/42 (PD98059) and p38 MAPK (SB203580) activation also suppressed the induction of COX-2 by this NSAID. Furthermore, both MAPK inhibitors significantly augmented sulindac sulfide-induced apoptosis, as did suppression of constitutive COX-2 using antisense oligonucleotides. In conclusion, MEK/ERK and p38 MAPK activation mediate COX-2 induction by sulindac sulfide. Selective inhibitors of these MAPKs potentiate apoptosis induction by this NSAID, suggesting a novel strategy for the prevention or treatment of colorectal cancer.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , MAP Quinase Quinase 1/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Sulindaco/análogos & derivados , Sulindaco/toxicidade , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Antineoplásicos/toxicidade , Linhagem Celular Tumoral , Neoplasias do Colo , Inibidores de Ciclo-Oxigenase/toxicidade , Sinergismo Farmacológico , Flavonoides/toxicidade , Humanos , Nitrobenzenos/toxicidade , Sulfonamidas/toxicidade
16.
Biochem J ; 374(Pt 2): 481-8, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12793857

RESUMO

Naproxen, sulindac and salicylate, three NSAIDs (non-steroidal anti-inflammatory drugs), were cytotoxic to human colorectal cancer cells in culture. Toxicity was accompanied by significant depletion of intracellular polyamine content. Inhibition of ornithine decarboxylase (the first enzyme of the polyamine biosynthetic pathway), induction of polyamine oxidase and spermidine/spermine N(1)-acetyltransferase (the enzymes responsible for polyamine catabolism) and induction of polyamine export all contributed to the decreased intracellular polyamine content. Morphological examination of the cells showed typical signs of apoptosis, and this was confirmed by DNA fragmentation and measurement of caspase-3-like activity. Re-addition of spermidine to the cells partially prevented apoptosis and recovered the cell number. Thus polyamines appear to be an integral part of the signalling pathway mediating NSAID toxicity in human colorectal cancer cells, and may therefore also be important in cancer chemoprevention in humans.


Assuntos
Anti-Inflamatórios não Esteroides/antagonistas & inibidores , Anti-Inflamatórios não Esteroides/toxicidade , Neoplasias Colorretais/metabolismo , Poliaminas/farmacologia , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/metabolismo , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/química , Neoplasias Colorretais/patologia , Fragmentação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Corantes Fluorescentes/metabolismo , Humanos , Indóis/metabolismo , Substâncias Intercalantes/metabolismo , Naproxeno/administração & dosagem , Naproxeno/antagonistas & inibidores , Naproxeno/metabolismo , Naproxeno/toxicidade , Poliaminas/metabolismo , Putrescina/metabolismo , Putrescina/farmacologia , Putrescina/fisiologia , Salicilatos/administração & dosagem , Salicilatos/antagonistas & inibidores , Salicilatos/metabolismo , Salicilatos/toxicidade , Espermidina/metabolismo , Espermidina/farmacologia , Espermidina/fisiologia , Espermina/metabolismo , Espermina/farmacologia , Espermina/fisiologia , Sulindaco/administração & dosagem , Sulindaco/antagonistas & inibidores , Sulindaco/metabolismo , Sulindaco/toxicidade , Células Tumorais Cultivadas
17.
Ann N Y Acad Sci ; 1010: 273-7, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15033733

RESUMO

Transcription factor NF-kappaB plays a pivotal role in cancer cells in the resistance to apoptosis, since NF-kappaB is frequently activated in many primary carcinoma cells. Indeed, several NF-kappaB inhibitors are found to be promising anti-cancer agents. However, some anti-cancer agents activate NF-kappaB signals and may reduce their potential, including tumor necrosis factor (TNF)-alpha. Recently, the nonsteroidal anti-inflammatory drug (NSAID) sulindac and its metabolites have been shown to inhibit the NF-kappaB-mediated survival signals through inhibition of IKK-beta by their direct interaction. We thus investigate whether sulindac and its metabolite can augment TNF-alpha-mediated apoptosis in human carcinoma cells and be applicable for in vivo clinical usage. We here demonstrate that sulindac inhibited TNF-alpha-mediated NF-kappaB activation and greatly enhanced TNF-alpha-induced apoptosis in human gastric MKN45 and cervical HeLa carcinoma cell lines. The in vivo tumor growth of MKN45 cells was most strongly inhibited by a combination of TNF-alpha with sulindac compared with TNF-alpha or sulindac alone. Moreover, we demonstrate that sulindac sulfide further augmented TNF-alpha-mediated apoptosis. Our data strongly suggest that combination therapy of TNF-alpha with sulindac and its metabolites may sensitize cancer cells to TNF-alpha and augment its pro-apoptotic potential. Therefore, in combination with sulindac or its metabolites, TNF-alpha may become a potentially useful anti-cancer agent to suppress tumor.


Assuntos
Apoptose/efeitos dos fármacos , Sulindaco/toxicidade , Fator de Necrose Tumoral alfa/toxicidade , Apoptose/fisiologia , Linhagem Celular Tumoral , Feminino , Humanos , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Neoplasias do Colo do Útero
18.
Curr Opin Investig Drugs ; 1(3): 386-91, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11249724

RESUMO

Cell Pathways has developed exisulind (Aptosyn), an oral apoptosis modulator and cGMP phosphodiesterase inhibitor, for the potential treatment of several oncologic indications including precancerous adenomatous polyposis coli (APC), also known as familial adenomatous polyposis (FAP), precancerous sporadic colonic polyps, cervical dysplasia and the prevention of tumor recurrence in prostate and breast cancer. An NDA filing for the treatment of precancerous APC, for which the US FDA designated exisulind a Fast Track product in July 1998, was initially expected in March 1999 [291313]. However, in January of the same year the company stated that it anticipated a delay in the NDA filing. The decision was based on unsatisfactory phase III data [308912], [313124]. In June 1999, the company completed analysis of the phase III trial data [328000] and the NDA was submitted in August 1999. An NDA was accepted for review by the FDA in October 1999 for the treatment of APC [328000], [338007], [344721], after data from three additional trials were submitted to the FDA in support of the NDA. At this time phase II/III trials were also ongoing for prostate and breast cancer recurrence [287250], [326795]. Approval for the indication of FAP had been expected by the end of 2000 [365737] but in September 2000 the FDA completed its initial review and advised Cell Pathways that exisulind will not be approved at this time. Cell Pathways has received a 'non-approvable' letter and intends to advise the FDA of its intent to amend the NDA and to request a meeting to address the deficiencies in the NDA [383249], [383560]. The first of the three additional trials submitted to the FDA in October 1999 was a 6-month, open-label trial involving 48 of the patients who completed a phase II/III study of exisulind in early 1999. After 6 months of treatment with exisulind, 25 patients who had previously been taking placebo experienced a 50% reduction in polyp formation. The patients continuing treatment with exisulind exhibited a further 50% reduction from their already reduced rate of polyp formation [344991]. The second study was an extension study of 11 patients who participated in a 6-month, open-label, phase I/II, dose-ranging, safety and efficacy trial. As of October 1999, these patients were still on therapy and had been receiving exisulind for between 36 and 50 months. They had all experienced statistically significant reductions in polyp formation rates [344991]. The third study was a double-blind, placebo-controlled safety study of 26 patients. All patients exhibited a trend of reduced new polyp formation when compared to placebo. Exisulind was generally well-tolerated by all patients during the course of the studies [344991]. In July 2000, Cell Pathways signed a marketing and distribution agreement for Canada with Paladin Labs, allowing Paladin exclusive rights to commercialize exisulind within Canada [376072]. Also in July 2000, Cell Pathways announced that patents covering methods of identifying compounds that selectively stimulate apoptosis have been allowed in Europe and Japan. The patents describe the mechanism of action of Cell Pathways' SAANDs, including exisulind, and use of that knowledge in screening for new drugs [374888]. In January 1999, the company received US-05858694 covering the mechanism of action of exisulind [311504].


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Drogas em Investigação/farmacologia , Sulindaco/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Ensaios Clínicos como Assunto , Drogas em Investigação/síntese química , Drogas em Investigação/toxicidade , Humanos , Relação Estrutura-Atividade , Sulindaco/análogos & derivados , Sulindaco/síntese química , Sulindaco/toxicidade
19.
Chem Res Toxicol ; 10(5): 546-55, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9168252

RESUMO

Reactive metabolite-modified hepatic protein adducts have been proposed to play important roles in the mechanism(s) of hepatotoxicity of nonsteroidal anti-inflammatory drugs (NSAIDs). In the present study, immunochemical techniques have been used to compare the patterns of drug-protein adducts expressed in livers of mice given single doses of one or other of three different NSAIDs. These were diclofenac and sulindac, which are widely used but potentially hepatotoxic drugs, and ibuprofen, which is considered to be nonhepatotoxic. Specific polyclonal antisera were produced by immunization of rabbits with conjugates prepared by coupling each of the NSAIDs to the carrier protein keyhole limpet hemocyanin. Immunoblotting studies revealed dose-dependent formation of major 110 kDa polypeptide adducts in livers from mice sacrificed 6 h after administration of single doses of either diclofenac (0-300 mg/kg) or sulindac (0-100 mg/kg). Lower levels of several other adducts, of 140 and 200 kDa, were also expressed in livers from these animals. In contrast, livers from mice treated with ibuprofen (0-200 mg/kg) predominantly expressed a 60 kDa adduct and only relatively low levels of a 110 kDa adduct. The various adducts were shown by differential centrifugation to be concentrated in the nuclear fraction of liver homogenates. Those derived from diclofenac and sulindac were further localized, by Percoll density gradient centrifugation, to a subfraction which contained a high activity of the bile canalicular marker enzyme alkaline phosphatase. This suggests that they are concentrated in the bile canalicular domain of hepatocytes. The different patterns of adduct formation raise the possibility that formation of certain NSAID protein adducts, particularly 110 kDa adducts, has toxicological significance.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Fígado/química , Fígado/metabolismo , Proteínas/química , Proteínas/metabolismo , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/toxicidade , Diclofenaco/química , Diclofenaco/metabolismo , Diclofenaco/toxicidade , Feminino , Ibuprofeno/química , Ibuprofeno/metabolismo , Ibuprofeno/toxicidade , Soros Imunes/química , Immunoblotting , Imunoquímica , Fígado/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Coelhos , Sulindaco/química , Sulindaco/metabolismo , Sulindaco/toxicidade
20.
Oncogene ; 12(4): 893-901, 1996 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8632912

RESUMO

Nonsteroidal anti-inflammatory drugs lower the incidence of and mortality from colon cancer. Sulindac reduces the number and size of polyps in patients with familial adenomatous polyposis. We have shown that sulindac and sulindac sulfide reversibly reduce the proliferation rate of HT-29 colon cancer cells, alter their morphology, induce them to accumulate in the G0/G1 phase of the cell cycle, and sulindac sulfide induces cell death by apoptosis. In this study we confirmed that sulindac and sulindac sulfide prevent HT-29 cells from progressing from the G0/G1 into the S phase. This block in cell cycle progression is associated with an initial rise, then an abrupt decrease in the levels of p34cdc2 protein. Sulindac and sulindac sulfide decrease the levels of mitotic cyclins, induce the levels of p21WAF-1/cip1, and reduce the total levels of pRB, with a relative increase in the amount of the underphosphorylated form of pRB in a time- and concentration-dependent manner. In addition, these compounds reduce the levels of mutant p53. These responses are not associated with intestinal cell differentiation and occur independent of the ability of these compounds to induce apoptosis. We conclude that sulindac and sulindac sulfide reduce the levels of major components of the molecular cell cycle machinery and alter the levels of several tumor suppressor proteins in a manner consistent with cell cycle quiescence. These mechanisms may be operative in vivo to account, in part, for the anti-neoplastic effects of these compounds.


Assuntos
Anti-Inflamatórios não Esteroides/toxicidade , Antineoplásicos/toxicidade , Ciclo Celular/efeitos dos fármacos , Ciclinas/metabolismo , Genes do Retinoblastoma/efeitos dos fármacos , Genes p53/efeitos dos fármacos , Sulindaco/análogos & derivados , Sulindaco/toxicidade , Adenocarcinoma , Apoptose/efeitos dos fármacos , Western Blotting , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Neoplasias do Colo , Ciclinas/efeitos dos fármacos , Humanos , Mitose/efeitos dos fármacos , Proteína do Retinoblastoma/biossíntese , Proteína Supressora de Tumor p53/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA