Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 217
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 37(5): 983-989, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28302628

RESUMO

OBJECTIVE: To investigate the relationship between 3 markers of apoptosis, that is, FADD (Fas-associated death domain-containing protein), caspase-3, and caspase-8, and incidence of coronary events (CEs) in a population-based cohort study. APPROACH AND RESULTS: In vitro experiments were performed to assess the response of the apoptotic biomarkers after Fas stimulation of peripheral blood mononuclear cells. The experiments showed significantly increased releases of FADD, caspase-3, and caspase-8 after Fas stimulation. The relationship between FADD, caspase-3, and caspase-8, respectively, and incidence of CEs was studied in 4284 subjects from the population-based Malmö Diet and Cancer Study. Cox' proportional hazards regression was used to examine the association between the apoptotic biomarkers and incidence of CE over a mean follow-up of 19 years. A total of 381 individuals had CE during the follow-up. High FADD at baseline was significantly associated with incident CE. In the highest compared with the lowest quartile of FADD, the risk factor adjusted hazards ratio for CE was 1.82 (95% confidence interval, 1.35-2.46; P for trend <0.001). A significant association was also found between caspase-8 and CE; the hazards ratio (Q4 versus Q1) was 1.90 (95% confidence interval, 1.39-2.60; P for trend <0.001) after adjustment for risk factors. No association was found between caspase-3 and CEs. CONCLUSIONS: High levels of FADD and caspase-8, but not caspase-3, were associated with increased incidence of CE in subjects from the general population. The in vitro experiments support the view that these biomarkers could reflect activation of the extrinsic apoptotic pathway.


Assuntos
Apoptose , Caspase 3/sangue , Caspase 8/sangue , Doença das Coronárias/sangue , Doença das Coronárias/epidemiologia , Proteína de Domínio de Morte Associada a Fas/sangue , Biomarcadores/sangue , Células Cultivadas , Doença das Coronárias/diagnóstico por imagem , Doença das Coronárias/patologia , Feminino , Humanos , Incidência , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/enzimologia , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prognóstico , Modelos de Riscos Proporcionais , Fatores de Risco , Suécia/epidemiologia , Fatores de Tempo , Regulação para Cima , Receptor fas/farmacologia
2.
Mol Cell ; 64(2): 236-250, 2016 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-27746017

RESUMO

Caspase-8 activation can be triggered by death receptor-mediated formation of the death-inducing signaling complex (DISC) and by the inflammasome adaptor ASC. Caspase-8 assembles with FADD at the DISC and with ASC at the inflammasome through its tandem death effector domain (tDED), which is regulated by the tDED-containing cellular inhibitor cFLIP and the viral inhibitor MC159. Here we present the caspase-8 tDED filament structure determined by cryoelectron microscopy. Extensive assembly interfaces not predicted by the previously proposed linear DED chain model were uncovered, and were further confirmed by structure-based mutagenesis in filament formation in vitro and Fas-induced apoptosis and ASC-mediated caspase-8 recruitment in cells. Structurally, the two DEDs in caspase-8 use quasi-equivalent contacts to enable assembly. Using the tDED filament structure as a template, structural analyses reveal the interaction surfaces between FADD and caspase-8 and the distinct mechanisms of regulation by cFLIP and MC159 through comingling and capping, respectively.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/química , Caspase 8/química , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/química , Proteína de Domínio de Morte Associada a Fas/química , Proteínas Virais/química , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Sítios de Ligação , Proteínas Adaptadoras de Sinalização CARD , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Caspase 8/genética , Caspase 8/metabolismo , Microscopia Crioeletrônica , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/genética , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Domínio Efetor de Morte , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Expressão Gênica , Humanos , Células Jurkat , Plasmídeos/química , Plasmídeos/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transfecção , Proteínas Virais/genética , Proteínas Virais/metabolismo , Receptor fas/farmacologia
3.
Cell Death Dis ; 7: e2209, 2016 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-27124583

RESUMO

Glioblastoma (GBM) is one of the most aggressive types of cancer with limited therapeutic options and unfavorable prognosis. Stemness and non-classical epithelial-to-mesenchymal transition (ncEMT) features underlie the switch from normal to neoplastic states as well as resistance of tumor clones to current therapies. Therefore, identification of ligand/receptor systems maintaining this privileged state is needed to devise efficient cancer therapies. In this study, we show that the expression of CD95 associates with stemness and EMT features in GBM tumors and cells and serves as a prognostic biomarker. CD95 expression increases in tumors and with tumor relapse as compared with non-tumor tissue. Recruitment of the activating PI3K subunit, p85, to CD95 death domain is required for maintenance of EMT-related transcripts. A combination of the current GBM therapy, temozolomide, with a CD95 inhibitor dramatically abrogates tumor sphere formation. This study molecularly dissects the role of CD95 in GBM cells and contributes the rational for CD95 inhibition as a GBM therapy.


Assuntos
Neoplasias Encefálicas/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Recidiva Local de Neoplasia/genética , Receptor fas/genética , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/classificação , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Combinação de Medicamentos , Sinergismo Farmacológico , Glioblastoma/classificação , Glioblastoma/mortalidade , Glioblastoma/patologia , Humanos , Imunoglobulina G/farmacologia , Recidiva Local de Neoplasia/classificação , Recidiva Local de Neoplasia/mortalidade , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Cultura Primária de Células , Prognóstico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Análise de Sobrevida , Temozolomida , Receptor fas/metabolismo , Receptor fas/farmacologia
4.
Anticancer Drugs ; 26(7): 716-27, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25850884

RESUMO

Glioblastoma is a disease characterized by rapid invasive tumour growth. Studies on the proapoptotic CD95/CD95L signalling pathway recently suggested a significant contribution of CD95 signalling towards the high degree of motility in glioma cells. Apogenix has developed APG101, a clinical phase II compound designed to bind and neutralize CD95L, and thus to interfere with CD95/CD95L-based signalling. APG101 has shown clinical efficacy in a controlled randomized phase II trial in patients with recurrent glioma. Because APG101 is not cytotoxic to tumour cells in vitro, we postulated that the anti-invasive function of APG101 is the main mechanism of action for this compound. Using three-dimensional spheroid invasion assays in vitro and in murine brain tissue cultures, we found that knockdown of endogenous CD95L reduced the invasive phenotype in our two glioblastoma model cell lines U87-MG and U251-MG. Invasion was restored in CD95L knockdown cells upon the addition of soluble recombinant CD95L and this effect was inhibited by APG101. We conclude that CD95L from autocrine and paracrine sources contributes towards the invasive phenotype of glioblastoma cells and that APG101 acts as a suppressor of proinvasive signalling by the CD95/CD95L pathway in glioblastoma.


Assuntos
Neoplasias Encefálicas/patologia , Proteína Ligante Fas/metabolismo , Glioblastoma/patologia , Imunoglobulina G/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Esferoides Celulares/efeitos dos fármacos , Receptor fas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Resistencia a Medicamentos Antineoplásicos , Técnicas de Silenciamento de Genes , Glioblastoma/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , Transdução de Sinais , Esferoides Celulares/patologia , Esferoides Celulares/fisiologia , Receptor fas/genética , Receptor fas/farmacologia
5.
PLoS One ; 9(2): e90298, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587316

RESUMO

Eosinophils play a central role in asthma. The present study was performed to investigate the effect of tumour necrosis factor-α (TNF-α) on longevity of isolated human eosinophils. In contrast to Fas, TNF-α inhibited eosinophil apoptosis as evidenced by a combination of flow cytometry, DNA fragmentation assay and morphological analyses. The effect of TNF-α on eosinophil apoptosis was reversed by a TNF-α neutralising antibody. The anti-apoptotic effect of TNF-α was not due to autocrine release of known survival-prolonging cytokines interleukins 3 and 5 or granulocyte-macrophage-colony-stimulating factor as their neutralisation did not affect the effect of TNF-α. The anti-apoptotic signal was mediated mainly by the TNF-receptor 1. TNF-α induced phosphorylation and degradation of IκB and an increase in NF-κB DNA-binding activity. The survival-prolonging effect of TNF-α was reversed by inhibitors of NF-κB pyrrolidinedithiocarbamate and gliotoxin and by an inhibitor of IκB kinase, BMS-345541. TNF-α induced also an increase in AP-1 DNA-binding activity and the antiapoptotic effect of TNF-α was potentiated by inhibitors of AP-1, SR 11302 and tanshinone IIA and by an inhibitor of c-jun-N-terminal kinase, SP600125, which is an upstream kinase activating AP-1. Our results thus suggest that TNF-α delays human eosinophil apoptosis via TNF-receptor 1 and the resulting changes in longevity depend on yin-yang balance between activation of NF-κB and AP-1.


Assuntos
Asma/genética , Eosinófilos/metabolismo , NF-kappa B/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Transcrição AP-1/genética , Fator de Necrose Tumoral alfa/farmacologia , Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Asma/metabolismo , Asma/patologia , Sobrevivência Celular/efeitos dos fármacos , Eosinófilos/efeitos dos fármacos , Eosinófilos/patologia , Regulação da Expressão Gênica , Gliotoxina/farmacologia , Humanos , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Imidazóis/farmacologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Fosforilação , Cultura Primária de Células , Ligação Proteica , Pirrolidinas/farmacologia , Quinoxalinas/farmacologia , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Retinoides/farmacologia , Transdução de Sinais , Tiocarbamatos/farmacologia , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Receptor fas/farmacologia
6.
Blood ; 121(3): 556-65, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23203823

RESUMO

Graft-versus-host disease (GVHD) induced by transplant-derived T cells represents a major complication after allogeneic bone marrow transplantation (BMT). However, these T cells support engraftment, early T-cell immunity, and mediate the graft-versus-tumor (GVT) effect. Cytotoxic effector functions by transplanted T cells are predominantly mediated by the perforin/granzyme and the CD95/CD95L system. APG101, a novel recombinant human fusion protein consisting of the extracellular domain of CD95 and the Fc domain of an IgG1 antibody inhibited CD95L-induced apoptosis without interfering with T-cell function in vitro and was therefore tested for its ability to prevent GVHD in murine BMT models across minor or major histocompatibility barriers. Starting APG101 treatment either 1 day before or 6 days after transplantation effectively reduced clinical GVHD and rescued survival between 60% and 100% if GVHD was CD95L mediated. APG101 did not interfere with the GVT effect, because P815 mastocytoma and most importantly primary Bcr-Abl-transformed B-cell leukemias were completely eradicated by the alloantigen-specific T cells. Phenotype and homing of alloantigen-specific T cells or their perforin/granzyme-mediated cytotoxicity and proliferative capacity were not affected by APG101 treatment suggesting that APG101 therapy might be useful in GVHD prophylaxis without impairing T-cell function and most importantly preserving GVT activity.


Assuntos
Transplante de Medula Óssea/efeitos adversos , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Imunoglobulina G/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Receptor fas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Transplante de Medula Óssea/imunologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/imunologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Modelos Animais de Doenças , Feminino , Imunoglobulina G/imunologia , Imunofenotipagem , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neoplasias/imunologia , Proteínas Recombinantes de Fusão/imunologia , Transplante Homólogo , Receptor fas/imunologia
7.
J Neurotrauma ; 29(8): 1586-99, 2012 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-22260324

RESUMO

Spinal cord injury (SCI) is a devastating condition that currently lacks clinically-relevant and effective neuroprotective therapeutic options. Optimal therapeutic agents for clinical translation should show efficacy in a cervical compression/contusion model using a clinically-relevant post-injury therapeutic time window. To date, few compounds have met that rigorous standard. The objective of this work was to evaluate the efficacy of delayed post-injury administration of soluble Fas receptor (sFasR) via intrathecal catheter following acute cervical SCI in a clinically-relevant contusion/compression model. Female Wistar rats were given a C7-T1 moderately severe clip compression injury, followed by either 8-h or 24-h delayed treatment initiation. Long-term neurobehavioral analysis of motor recovery and neuropathic pain development was undertaken. The extent of oligodendrocyte and neuron survival was assessed in peri-lesional cord sections 8 weeks post-SCI. This was complemented by an evaluation of the level of tissue preservation at and adjacent to the site of injury. In animals treated with sFasR delayed 8 h post-injury, significant behavioral effects were observed, coinciding with enhanced cell survival, peri-lesional tissue sparing, and enhanced integrity of descending fiber tracts compared to control treatments. Animals treated with sFasR delayed by 24 h showed more modest improvements in behavioral recovery, and had consistent improvements in cell survival and tissue preservation. This work has shown for the first time that the Fas-mediated apoptotic pathway can be therapeutically targeted in a clinically-relevant time window post-SCI.


Assuntos
Degeneração Neural/tratamento farmacológico , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Receptor fas/uso terapêutico , Animais , Apoptose/fisiologia , Comportamento Animal/efeitos dos fármacos , Feminino , Degeneração Neural/etiologia , Degeneração Neural/metabolismo , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Neuralgia/metabolismo , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Medição da Dor , Ratos , Ratos Wistar , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/metabolismo , Fatores de Tempo , Receptor fas/farmacologia
8.
PLoS One ; 6(5): e19974, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21629700

RESUMO

Activation of an acid sphingomyelinase (aSMase) leading to a biosynthesis of GD3 disialoganglioside has been associated with Fas-induced apoptosis of lymphoid cells. The present study was undertaken to clarify the role of this enzyme in the generation of gangliosides during apoptosis triggered by Fas ligation. The issue was addressed by using aSMase-deficient and aSMase-corrected cell lines derived from Niemann-Pick disease (NPD) patients. Fas cross-linking elicited a rapid production of large amounts of complex a- and b-series species of gangliosides with a pattern and a chromatographic behavior as single bands reminiscent of brain gangliosides. The gangliosides were synthesized within the first ten minutes and completely disappeared within thirty minutes after stimulation. Noteworthy is the observation that GD3 was not the only ganglioside produced. The production of gangliosides and the onset of apoptotic hallmarks occurred similarly in both aSMase-deficient and aSMase-corrected NPD lymphoid cells, indicating that aSMase activation is not accountable for ganglioside generation. Hampering ganglioside production by inhibiting the key enzyme glucosylceramide synthase did not abrogate the apoptotic process. In addition, GM3 synthase-deficient lymphoid cells underwent Fas-induced apoptosis, suggesting that gangliosides are unlikely to play an indispensable role in transducing Fas-induced apoptosis of lymphoid cells.


Assuntos
Apoptose/efeitos dos fármacos , Gangliosídeos/metabolismo , Linfócitos/citologia , Linfócitos/metabolismo , Receptor fas/farmacologia , Western Blotting , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Citometria de Fluxo , Humanos , Linfócitos/efeitos dos fármacos , Esfingomielinas/metabolismo
9.
Blood ; 117(22): 5953-62, 2011 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-21478427

RESUMO

The most common form of neutrophil death is apoptosis. In the present study, we report surprising differences in the molecular mechanisms used for caspase activation between FAS/CD95-stimulated and TNF receptor 1 (TNFR1)-stimulated neutrophils. Whereas FAS-induced apoptosis was followed by caspase-8 activation and required Bid to initiate the mitochondrial amplification loop, TNF-α-induced apoptosis involved class IA PI3Ks, which were activated by MAPK p38. TNF-α-induced PI3K activation resulted in the generation of reactive oxygen species, which activated caspase-3, a mechanism that did not operate in neutrophils without active NADPH oxidase. We conclude that in neutrophils, proapoptotic pathways after TNFR1 stimulation are initiated by p38 and PI3K, but not by caspase-8, a finding that should be considered in anti-inflammatory drug-development strategies.


Assuntos
Apoptose , Neutrófilos/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/fisiologia , Caspase 3/metabolismo , Caspase 8/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Humanos , Potencial da Membrana Mitocondrial , Camundongos , Mitocôndrias/metabolismo , NADPH Oxidases/metabolismo , Neutrófilos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
J Biol Chem ; 285(26): 19997-20005, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20427266

RESUMO

Evidence accumulates that in clinically relevant cell death, both the intrinsic and extrinsic apoptotic pathway synergistically contribute to organ failure. In search for an inhibitor of apoptosis that provides effective blockage of these pathways, we analyzed viral proteins that evolved to protect the infected host cells. In particular, the cowpox virus protein crmA has been demonstrated to be capable of blocking key caspases of both pro-apoptotic pathways. To deliver crmA into eukaryotic cells, we fused the TAT protein transduction domain of HIV to the N terminus of crmA. In vitro, the TAT-crmA fusion protein was efficiently translocated into target cells and inhibited apoptosis mediated through caspase-8, caspase-9, and caspase-3 after stimulation with alpha-Fas, etoposide, doxorubicin, or staurosporine. The extrinsic apoptotic pathway was investigated following alpha-Fas stimulation. In vivo 90% of TAT-crmA-treated animals survived an otherwise lethal dose of alpha-Fas and showed protection from Fas-induced organ failure. To examine the intrinsic apoptotic pathway, we investigated the survival of mice treated with an otherwise lethal dose of doxorubicin. Whereas all control mice died within 31 days, 40% of mice that concomitantly received intraperitoneal injections of TAT-crmA survived. To test the ability to comprehensively block both the intrinsic and extrinsic apoptotic pathway in a clinically relevant setting, we employed a murine cardiac ischemia-reperfusion model. TAT-crmA reduced infarction size by 40% and preserved left ventricular function. In summary, these results provide a proof of principle for the inhibition of apoptosis with TAT-crmA, which might provide a new treatment option for ischemia-reperfusion injuries.


Assuntos
Apoptose/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Serpinas/metabolismo , Transdução de Sinais/fisiologia , Proteínas Virais/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Inibidores de Caspase , Caspases/metabolismo , Doxorrubicina/farmacologia , Etoposídeo/farmacologia , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Immunoblotting , Células Jurkat , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/prevenção & controle , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Serpinas/genética , Serpinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Análise de Sobrevida , Proteínas Virais/genética , Proteínas Virais/farmacologia , Receptor fas/farmacologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
11.
Endocrinology ; 151(2): 529-36, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19934374

RESUMO

Senescence marker protein-30 (SMP30) is an androgen-independent factor that decreases with age. We recently identified SMP30 as the lactone-hydrolyzing enzyme gluconolactonase (GNL), which is involved in vitamin C biosynthesis in animal species. To examine whether the age-related decrease in SMP30/GNL has effects on glucose homeostasis, we used SMP30/GNL knockout (KO) mice treated with L-ascorbic acid. In an ip glucose tolerance test at 15 wk of age, blood glucose levels in SMP30/GNL KO mice were significantly increased by 25% at 30 min after glucose administration compared with wild-type (WT) mice. Insulin levels in SMP30/GNL KO mice were significantly decreased by 37% at 30 min after glucose compared with WT mice. Interestingly, an insulin tolerance test showed a greater glucose-lowering effect in SMP30/GNL KO mice. High-fat diet feeding severely worsened glucose tolerance in both WT and SMP30/GNL KO mice. Morphometric analysis revealed no differences in the degree of high-fat diet-induced compensatory increase in beta-cell mass and proliferation. In the static incubation study of islets, insulin secretion in response to 20 mm glucose or KCl was significantly decreased in SMP30/GNL KO mice. On the other hand, islet ATP content at 20 mm in SMP30/GNL KO mice was similar to that in WT mice. Collectively, these data indicate that impairment of the early phase of insulin secretion due to dysfunction of the distal portion of the secretion pathway underlies glucose intolerance in SMP30/GNL KO mice. Decreased SMP30/GNL may contribute to the worsening of glucose tolerance that occurs in normal aging.


Assuntos
Proteínas de Ligação ao Cálcio/deficiência , Hidrolases de Éster Carboxílico/deficiência , Hidrolases de Éster Carboxílico/genética , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/fisiologia , Envelhecimento/genética , Envelhecimento/fisiologia , Ração Animal , Animais , Apoptose/efeitos dos fármacos , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Encéfalo/fisiologia , Divisão Celular/efeitos dos fármacos , Epididimo/anatomia & histologia , Teste de Tolerância a Glucose , Insulina/análise , Insulina/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/fisiologia , Pulmão/fisiologia , Masculino , Camundongos , Camundongos Knockout , Alvéolos Pulmonares/patologia , Fator de Necrose Tumoral alfa/farmacologia , Aumento de Peso , Receptor fas/farmacologia
12.
Ren Fail ; 31(5): 377-81, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19839838

RESUMO

UNLABELLED: Fas (APO-1/CD95) is a cell surface receptor that initiates apoptotic pathway. Fas-stimulated ROS generation may play important role in Fas-mediated apoptosis. The aim of this study was to evaluate the influence of interferon-alpha on oxidative stress parameters in Fas-induced renal apoptosis in mice kidney. SUBJECTS AND METHODS: One-month-old Balb C male mice were used for the study. The animals were divided in four groups: group 1 were the controls, group 2 mice were treated with anti-Fas antibody i.p., group 3 mice were treated with IFN-alpha, and group 4 mice were treated with both agents simultaneously. The mice were killed 48 h afterwards, and kidneys were homogenized. TBA reactive substances (TBARS), glutathione content, and reactive carbonyl group (RCG) were measured. RESULTS: The results showed a statistically significant increase of TBARS (p < 0.05) and RCG (p < 0.05) concentration in the group treated with anti-Fas antibody versus control. IFN-alpha decreased the concentration of TBARS and RCG after anti-Fas antibody administration (p < 0.05). There is no significant difference in glutathione content between investigated groups. CONCLUSION: IFN-alpha might be considered as a new target for therapeutic intervention in FasL/Fas induced renal injury.


Assuntos
Apoptose/efeitos dos fármacos , Interferon-alfa/farmacologia , Rim/efeitos dos fármacos , Receptor fas/farmacologia , Animais , Apoptose/fisiologia , Creatinina/sangue , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Injeções Intraperitoneais , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Probabilidade , Distribuição Aleatória , Valores de Referência , Estatísticas não Paramétricas , Ureia/sangue
13.
Exp Neurol ; 215(1): 48-52, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18950622

RESUMO

Estrogen is protective in experimental cerebral ischemia, yet the mechanism remains unclear. Fas-mediated apoptosis has been shown to be induced after cerebral ischemia and significantly contribute to ischemic brain damage. In this study, we tested if estrogen is protective against cerebral ischemia by suppressing Fas-mediated apoptosis. 17Beta-estradiol-treated and untreated ovariectomized (OVX) female mice were subjected to 2 h middle cerebral artery occlusion (MCAO). Expression of Fas and Fas-associated death domain (FADD) were measured at 3, 6 and 12 h of reperfusion by RT-PCR and Western blot, respectively. Post-ischemic activities of caspase-8 and -3 activities, the two downstream effectors of Fas-induced apoptosis, were also assayed at same time points by ELISA. Finally, Fas antibody-induced cell death in primary cortical neurons was assayed by fluorescence activated cell sorter (FACS) in the presence and absence of estradiol. Our data showed that estradiol-treated OVX female mice sustained smaller infarct compared to untreated OVX mice. Ischemia upregulated Fas and FADD expression, and increased caspase-8 and -3 activities in OVX female mouse cortex, which were significantly attenuated by estradiol. Estradiol also significantly inhibited Fas antibody-induced neuronal cell apoptosis. Our data suggests that inhibition of ischemia-induced Fas-mediated apoptosis is an important mechanism of neuroprotection by estrogen in cerebral ischemia.


Assuntos
Apoptose/efeitos dos fármacos , Infarto Encefálico/prevenção & controle , Estradiol/uso terapêutico , Infarto da Artéria Cerebral Média/complicações , Receptor fas/farmacologia , Análise de Variância , Animais , Infarto Encefálico/etiologia , Caspase 3/metabolismo , Caspase 8/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Modelos Animais de Doenças , Embrião de Mamíferos , Estradiol/farmacologia , Proteína de Domínio de Morte Associada a Fas/genética , Proteína de Domínio de Morte Associada a Fas/metabolismo , Feminino , Citometria de Fluxo/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/terapia , Neurônios/efeitos dos fármacos , Ovariectomia , Ratos
14.
Cancer Immunol Immunother ; 58(5): 699-708, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18807035

RESUMO

Several tumor immunotherapy approaches result in a low percentage of durable responses in selected cancers. We hypothesized that the insensitivity of cancer cells to immunotherapy may be related to an anti-apoptotic cancer cell milieu, which could be pharmacologically reverted through the inhibition of antiapoptotic Bcl-2 family proteins in cancer cells. ABT-737, a small molecule inhibitor of the antiapoptotic proteins Bcl-2, Bcl-w and Bcl-x(L), was tested for the ability to increase antitumor immune responses in two tumor immunotherapy animal models. The addition of systemic therapy with ABT-737 to the immunization of BALB/c mice with tumor antigen peptide-pulsed dendritic cells (DC) resulted in a significant delay in CT26 murine colon carcinoma tumor growth and improvement in survival. However, the addition of ABT-737 to either a vaccine strategy involving priming with TRP-2 melanoma antigen peptide-pulsed DC and boosting with recombinant Listeria monocytogenes expressing the same melanoma antigen, or the adoptive transfer of TCR transgenic cells, did not result in superior antitumor activity against B16 murine melanoma. In vitro studies failed to demonstrate increased cytotoxic lytic activity when testing the combination of ABT-737 with lymphokine activated killer (LAK) cells, or the death receptor agonists Fas, TRAIL-ligand or TNF-alpha against the CT26 and B16 cell lines. In conclusion, the Bcl-2 inhibitor ABT-737 sensitized cancer cells to the antitumor effect of antigen-specific immunotherapy in a vaccine model for the CT26 colon carcinoma in vivo but not in two immunotherapy strategies against B16 melanoma.


Assuntos
Compostos de Bifenilo/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Neoplasias do Colo/terapia , Imunoterapia/métodos , Melanoma Experimental/terapia , Proteínas de Neoplasias/antagonistas & inibidores , Nitrofenóis/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Sulfonamidas/uso terapêutico , Animais , Antígenos de Neoplasias/imunologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/imunologia , Neoplasias do Colo/imunologia , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imunoterapia Adotiva , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/imunologia , Células Matadoras Ativadas por Linfocina/transplante , Listeria monocytogenes/imunologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Piperazinas/uso terapêutico , Receptores de Morte Celular/agonistas , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Receptor fas/farmacologia
15.
Anticancer Res ; 28(2A): 593-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18506997

RESUMO

BACKGROUND: Acquired resistance to apoptosis is a critical event in tumour development and in insensitivity toward therapy. To investigate resistance mechanisms to Fas/CD95/Apo-1-induced apoptosis, a Fas ligand-resistant variant of the U937 cell line was generated. RESULTS: Selection for Fas resistance resulted in a partial cross-resistance to TRAIL and TNF-alpha. Activation of caspase-8 was found to be impaired and the expression of Fas was reduced. However, FADD expression and ligand-induced aggregation of Fas was intact. Inhibition of various signalling pathways with pharmacological inhibitors revealed that resistance to death receptor-mediated apoptosis was dependent on altered tyrosine phosphatase/kinase activities and de novo protein synthesis. Moreover, FLIP, an anti-apoptotic protein, was expressed to a higher extent in the resistant cells. CONCLUSION: We provide evidence that acquired resistance to Fas-induced apoptosis in U937 cells involves a discrete set of molecular mechanisms which also render the cells cross-resistant to other death ligands.


Assuntos
Caspase 8/metabolismo , Resistencia a Medicamentos Antineoplásicos , Receptor fas/farmacologia , Apoptose/efeitos dos fármacos , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Ativação Enzimática , Proteína Ligante Fas/metabolismo , Proteína de Domínio de Morte Associada a Fas/farmacologia , Humanos , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Células U937
16.
Oncol Rep ; 19(6): 1577-82, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18497968

RESUMO

A-kinase-anchoring protein 149 (AKAP149) is a member of a structurally diverse, though functionally similar anchoring protein family and is localized to the outer membrane of mitochondria and in the endoplasmic reticulum-nuclear envelope network. AKAP149 plays an important role in controlling the subcellular localization and temporal specificity of protein phosphorylation and mRNA metabolism by tethering kinases and phosphatases, such as protein kinase A and type I protein phosphatase, through its N-terminal protein-binding motifs and mRNAs via its C-terminal RNA-binding motifs. It is well recognized that caspases play a central role in transducing and amplifying the intracellular death signal and that apoptosis is executed as a consequence of caspase-mediated cleavage of multiple cellular substrates. The identification of novel death substrates and elucidation of the consequences of their proteolytic cleavages by caspases are therefore crucial for our understanding of cell death and other biological processes. Herein, we demonstrated that AKAP149 is a direct substrate of active caspase-3, -8 -and -10 in vitro and in vivo. 35S-labeled full-length AKAP149 was completely cleaved in vitro by active caspase-3, -8 and -10 into two fragments of approximately 105 and 45 kDa, while caspase-2 cleaved it partially and caspase-1 did not cleave it at all. AKAP149 was also cleaved by caspases during Fas- and staurosporine-induced apoptosis in Jurkat T and HeLa cells, which were blocked by specific inhibitors of caspase-3 and -8. The specific cleavage site for these caspases was mapped in vitro and in vivo to Asp582 at AKAP149, which is located between the protein kinase A regulatory subunit anchoring and KH RNA-binding domains. In addition, HeLa cells transiently overexpressing AKAP149 D582E mutant were resistant to staurosporine-induced HeLa cell apoptosis. Taken together, these data suggest that AKAP149 activity may be deregulated by caspase-dependent proteolysis during apoptotic cell death and may provide useful information for elucidating the apoptosis signaling pathways in detail.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Apoptose/fisiologia , Caspases/metabolismo , Proteínas de Ancoragem à Quinase A/genética , Ácido Aspártico/química , Ácido Aspártico/genética , Células HeLa , Humanos , Células Jurkat , Mutação/genética , Estaurosporina/farmacologia , Receptor fas/farmacologia
17.
Oncogene ; 27(18): 2513-24, 2008 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-17982483

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has emerged as a promising antineoplastic agent because of its ability to selectively kill tumoral cells. However, some cancer cells are resistant to TRAIL-induced apoptosis. We have previously demonstrated that in endometrial carcinoma cells such resistance is caused by elevated FLICE-inhibitory protein (FLIP) levels. The present study focuses on the mechanisms by which FLIP could be modulated to sensitize endometrial carcinoma cells to TRAIL-induced apoptosis. We find that inhibition of casein kinase (CK2) sensitizes endometrial carcinoma cells to TRAIL- and Fas-induced apoptosis. CK2 inhibition correlates with a reduction of FLIP protein, suggesting that CK2 regulates resistance to TRAIL and Fas by controlling FLIP levels. FLIP downregulation correlates with a reduction of mRNA and is prevented by addition of the MG-132, suggesting that CK2 inhibition results in a proteasome-mediated degradation of FLIP. Consistently, forced expression of FLIP restores resistance to TRAIL and Fas. Moreover, knockdown of either FADD or caspase-8 abrogates apoptosis triggered by inhibition of CK2, indicating that CK2 sensitization requires formation of functional DISC. Finally, because of the possible role of both TRAIL and CK2 in cancer therapy, we demonstrate that CK2 inhibition sensitizes primary endometrial carcinoma explants to TRAIL apoptosis. In conclusion, we demonstrate that CK2 regulates endometrial carcinoma cell sensitivity to TRAIL and Fas by regulating FLIP levels.


Assuntos
Antineoplásicos/farmacologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/biossíntese , Caseína Quinase II/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias do Endométrio/metabolismo , Proteínas de Neoplasias/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Receptor fas/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Caseína Quinase II/antagonistas & inibidores , Linhagem Celular , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Leupeptinas/farmacologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Células Tumorais Cultivadas , Receptor fas/uso terapêutico
18.
Am J Pathol ; 171(6): 1831-46, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17991716

RESUMO

Hepatitis C virus (HCV) is a major cause of chronic hepatitis that can lead to cirrhosis and hepatocellular carcinoma. To study the effects of HCV protein expression on host cells, we established conditional expression of the full-length open reading frame (ORF) of an infectious cDNA clone of HCV (genotype 1a, H77 strain) in the nontransformed human hepatocyte line cell HH4 using the ecdysone receptor regulatory system. Treatment with the ecdysone analog ponasterone-A induced tightly regulated and dose-dependent full-length HCV ORF expression and properly processed HCV proteins. HCV Core, NS3, and NS5A colocalized in perinuclear regions and associated with the early endosomal protein EEA1. HCV ORF expression caused marked growth inhibition, increased intracellular reactive oxygen species, up-regulation of glutamate-l-cysteine ligase activity, increased glutathione level, and activation of nuclear factor kappaB. Although it was not directly cytotoxic, HCV ORF expression sensitized HH4 cells to Fas at certain concentrations but not to tumor necrosis factor-related apoptosis-inducing ligand. HCV ORF expression in HH4 cells up-regulated genes involved in innate immune response/inflammation and oxidative stress responses and down-regulated cell growth-related genes. Expression of HCV ORF in host cells may contribute to HCV pathogenesis by producing oxidative stress and increasing the expression of genes related to the innate immune response and inflammation.


Assuntos
Hepacivirus/metabolismo , Hepatócitos/imunologia , Hepatócitos/virologia , Imunidade Inata/genética , Proteínas Virais/metabolismo , Apoptose/genética , Adesão Celular/genética , Ciclo Celular/genética , Proliferação de Células , Sobrevivência Celular/genética , Ecdisterona/análogos & derivados , Ecdisterona/farmacologia , Matriz Extracelular/genética , Expressão Gênica , Glutamato-Cisteína Ligase/metabolismo , Hepacivirus/genética , Hepatócitos/química , Humanos , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fases de Leitura Aberta/efeitos dos fármacos , Fases de Leitura Aberta/genética , Estresse Oxidativo/genética , Poliproteínas/genética , Poliproteínas/metabolismo , Transcrição Gênica , Regulação para Cima , Proteínas Virais/análise , Proteínas Virais/genética , Receptor fas/farmacologia
19.
Exp Mol Med ; 39(4): 439-49, 2007 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-17934331

RESUMO

Neutrophils are also known to acquire the characteristics of dendritic cells (DCs) under the appropriate conditions. In this study, neutrophils were cultivated in vitro in the presence or absence of compounds modulating their survival in an attempt to characterize the expression profile of the DC markers. Higher MHC-II, CD80, CD86, CD83, and CD40 expression levels were detected on the surface of the cultured neutrophils for 24 h than on the freshly isolated cells. The annexin V-positive cells showed a higher expression level of the DC markers than the annexin V-negative cells. The population of neutrophils double stained with annexin V and the DC markers increased after being incubated with agonistic anti-Fas Ab. LPS, the anti-apoptotic compound, decreased the CD86 and MHC-II expression levels but 50-60% of the DC marker-positive cells were detected in the annexin V-positive cells. In contrast, CD80, CD86, CD83, and HLA-DR mRNA levels increased in the GM-CSF-treated neutrophils but not in the anti-Fas Ab-treated neutrophils. T cell proliferation was inhibited by co-culturing them with anti-Fas Ab- or LPS-treated neutrophils at a high neutrophil:T cell ratio. However, the superantigen-mediated T cell proliferation was increased by the LPS-treated neutrophils but decreased by the anti-Fas Ab-treated neutrophils. There was a lower level of interferon-gamma production in the T cells co-cultured with anti-Fas Ab-treated neutrophils than with the LPS-treated neutrophils. This suggests that apoptotic neutrophils express DC markers on their surface and the differential expression of DC markers might have a detrimental effect on the immune reaction.


Assuntos
Antígenos de Diferenciação/biossíntese , Apoptose , Células Dendríticas/metabolismo , Neutrófilos/metabolismo , Apresentação de Antígeno , Antígenos CD/biossíntese , Células Cultivadas , Humanos , Lipopolissacarídeos/farmacologia , Ativação Linfocitária , Neutrófilos/fisiologia , Linfócitos T/imunologia , Receptor fas/farmacologia
20.
J Pharmacol Exp Ther ; 323(1): 31-8, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17620456

RESUMO

Simendans are novel agents used in the treatment of decompensated heart failure. They sensitize troponin C to calcium and open ATP-sensitive potassium channels and have been shown to reduce cardiac myocyte apoptosis. The aim of the present study was to evaluate whether simendans reduce pulmonary eosinophilia and regulate eosinophil apoptosis. Bronchoalveolar lavage (BAL) eosinophilia was evaluated in ovalbumin-sensitized mice. Effects of simendans on apoptosis in isolated human eosinophils were assessed by relative DNA fragmentation assay, annexin V-binding, and morphological analysis. Dextrosimendan [(+)-[[4-(1,4,5,6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl)phenyl)hydrazono]propanedinitrile] reduced ovalbumin-induced BAL-eosinophilia in sensitized mice. Levosimendan [(-)-[[4-(1,4,5,6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl)phenyl]hydrazono]propanedinitrile] and dextrosimendan reversed interleukin (IL)-5-afforded survival of human eosinophils by inducing apoptosis in vitro. Even high concentrations of IL-5 were not able to overcome the effect of dextrosimendan. Dextrosimendan further enhanced spontaneous apoptosis as well as that induced by CD95 ligation, without inducing primary necrosis. Dextrosimendan-induced DNA fragmentation was shown to be dependent on caspase and c-Jun NH2-terminal kinase activation, whereas extracellular signal-regulated kinase, p38 mitogen-activated kinase, and ATP-sensitive potassium channels seemed to play no role in its actions. Taken together, our results show that simendans possess antieosinophilic activity and may be useful for the treatment of eosinophilic inflammation.


Assuntos
Apoptose/efeitos dos fármacos , Eosinófilos/efeitos dos fármacos , Hidrazonas , Eosinofilia Pulmonar/tratamento farmacológico , Piridazinas , Animais , Líquido da Lavagem Broncoalveolar/citologia , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fragmentação do DNA/efeitos dos fármacos , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Eosinófilos/imunologia , Humanos , Hidrazonas/farmacologia , Hidrazonas/uso terapêutico , Interleucina-5/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/farmacologia , Canais de Potássio/metabolismo , Eosinofilia Pulmonar/imunologia , Piridazinas/farmacologia , Piridazinas/uso terapêutico , Simendana , Receptor fas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA