Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Cell Death Dis ; 6: e1599, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25590809

RESUMO

Nicotinamide phosphoribosyltransferase (NAMPT) inhibitors (e.g., FK866) target the most active pathway of NAD(+) synthesis in tumor cells, but lack tumor-selectivity for use as a single agent. Reducing NAD(+) pools by inhibiting NAMPT primed pancreatic ductal adenocarcinoma (PDA) cells for poly(ADP ribose) polymerase (PARP1)-dependent cell death induced by the targeted cancer therapeutic, ß-lapachone (ß-lap, ARQ761), independent of poly(ADP ribose) (PAR) accumulation. ß-Lap is bioactivated by NADPH:quinone oxidoreductase 1 (NQO1) in a futile redox cycle that consumes oxygen and generates high levels of reactive oxygen species (ROS) that cause extensive DNA damage and rapid PARP1-mediated NAD(+) consumption. Synergy with FK866+ß-lap was tumor-selective, only occurring in NQO1-overexpressing cancer cells, which is noted in a majority (∼85%) of PDA cases. This treatment strategy simultaneously decreases NAD(+) synthesis while increasing NAD(+) consumption, reducing required doses and treatment times for both drugs and increasing potency. These complementary mechanisms caused profound NAD(P)(+) depletion and inhibited glycolysis, driving down adenosine triphosphate levels and preventing recovery normally observed with either agent alone. Cancer cells died through an ROS-induced, µ-calpain-mediated programmed cell death process that kills independent of caspase activation and is not driven by PAR accumulation, which we call NAD(+)-Keresis. Non-overlapping specificities of FK866 for PDA tumors that rely heavily on NAMPT-catalyzed NAD(+) synthesis and ß-lap for cancer cells with elevated NQO1 levels affords high tumor-selectivity. The concept of reducing NAD(+) pools in cancer cells to sensitize them to ROS-mediated cell death by ß-lap is a novel strategy with potential application for pancreatic and other types of NQO1+ solid tumors.


Assuntos
Naftoquinonas/farmacologia , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Poli Adenosina Difosfato Ribose/metabolismo , Acrilamidas/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Sinergismo Farmacológico , Metabolismo Energético/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Humanos , NAD(P)H Desidrogenase (Quinona)/metabolismo , Nicotinamida Fosforribosiltransferase/metabolismo , Neoplasias Pancreáticas/enzimologia , Piperidinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Pancreáticas
2.
Oncogene ; 34(1): 129-134, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24362532

RESUMO

Epidermal growth factor receptor (EGFR)vIII is the most common EGFR mutant found in glioblastoma (GBM). EGFRvIII does not bind ligand, is highly oncogenic and is usually coexpressed with EGFR wild type (EGFRwt). EGFRvIII activates Met, and Met contributes to EGFRvIII-mediated oncogenicity and resistance to treatment. Here, we report that addition of EGF results in a rapid loss of EGFRvIII-driven Met phosphorylation in glioma cells. Met is associated with EGFRvIII in a physical complex. Addition of EGF results in a dissociation of the EGFRvIII-Met complex with a concomitant loss of Met phosphorylation. Consistent with the abrogation of Met activation, addition of EGF results in the inhibition of EGFRvIII-mediated resistance to chemotherapy. Thus, our study suggests that ligand in the milieu of EGFRvIII-expressing GBM cells is likely to influence the EGFRvIII-Met interaction and resistance to treatment, and highlights a novel antagonistic interaction between EGFRwt and EGFRvIII in glioma cells.


Assuntos
Neoplasias Encefálicas/metabolismo , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/química , Fator de Crescimento Epidérmico/metabolismo , Glioblastoma/tratamento farmacológico , Humanos , Fenótipo , Fosforilação , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Temozolomida
3.
Oncogene ; 34(30): 3908-16, 2015 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25284588

RESUMO

Metastatic progression, including extravasation and micrometastatic outgrowth, is the main cause of cancer patient death. Recent studies suggest that cancer cells reprogram their metabolism to support increased proliferation through increased glycolysis and biosynthetic activities, including lipogenesis pathways. However, metabolic changes during metastatic progression, including alterations in regulatory gene expression, remain undefined. We show that transforming growth factor beta 1 (TGFß1)-induced epithelial-to-mesenchymal transition (EMT) is accompanied by coordinately reduced enzyme expression required to convert glucose into fatty acids, and concomitant enhanced respiration. Overexpressed Snail1, a transcription factor mediating TGFß1-induced EMT, was sufficient to suppress carbohydrate-responsive-element-binding protein (ChREBP, a master lipogenic regulator), and fatty acid synthase (FASN), its effector lipogenic gene. Stable FASN knockdown was sufficient to induce EMT, stimulate migration and extravasation in vitro. FASN silencing enhanced lung metastasis and death in vivo. These data suggest that a metabolic transition that suppresses lipogenesis and favors energy production is an essential component of TGFß1-induced EMT and metastasis.


Assuntos
Transição Epitelial-Mesenquimal , Fator de Crescimento Transformador beta1/fisiologia , Animais , Metabolismo dos Carboidratos , Linhagem Celular Tumoral , Movimento Celular , Repressão Enzimática , Ácido Graxo Sintase Tipo I/genética , Ácido Graxo Sintase Tipo I/metabolismo , Feminino , Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Metabolismo dos Lipídeos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Neoplasias , Fatores de Transcrição da Família Snail , Fatores de Transcrição/fisiologia
4.
Oncogene ; 33(33): 4253-64, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-24077285

RESUMO

EGFRvIII is a key oncogene in glioblastoma (GBM). EGFRvIII results from an in-frame deletion in the extracellular domain of EGFR, does not bind ligand and is thought to be constitutively active. Although EGFRvIII dimerization is known to activate EGFRvIII, the factors that drive EGFRvIII dimerization and activation are not well understood. Here we present a new model of EGFRvIII activation and propose that oncogenic activation of EGFRvIII in glioma cells is driven by co-expressed activated EGFR wild type (EGFRwt). Increasing EGFRwt leads to a striking increase in EGFRvIII tyrosine phosphorylation and activation while silencing EGFRwt inhibits EGFRvIII activation. Both the dimerization arm and the kinase activity of EGFRwt are required for EGFRvIII activation. EGFRwt activates EGFRvIII by facilitating EGFRvIII dimerization. We have previously identified HB-EGF, a ligand for EGFRwt, as a gene induced specifically by EGFRvIII. In this study, we show that HB-EGF is induced by EGFRvIII only when EGFRwt is present. Remarkably, altering HB-EGF recapitulates the effect of EGFRwt on EGFRvIII activation. Thus, increasing HB-EGF leads to a striking increase in EGFRvIII tyrosine phosphorylation while silencing HB-EGF attenuates EGFRvIII phosphorylation, suggesting that an EGFRvIII-HB-EGF-EGFRwt feed-forward loop regulates EGFRvIII activation. Silencing EGFRwt or HB-EGF leads to a striking inhibition of EGFRvIII-induced tumorigenicity, while increasing EGFRwt or HB-EGF levels resulted in accelerated EGFRvIII-mediated oncogenicity in an orthotopic mouse model. Furthermore, we demonstrate the existence of this loop in human GBM. Thus, our data demonstrate that oncogenic activation of EGFRvIII in GBM is likely maintained by a continuous EGFRwt-EGFRvIII-HB-EGF loop, potentially an attractive target for therapeutic intervention.


Assuntos
Neoplasias Encefálicas/metabolismo , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Animais , Linhagem Celular Tumoral , Receptores ErbB/genética , Retroalimentação Fisiológica , Regulação Neoplásica da Expressão Gênica , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fosforilação , Multimerização Proteica , Processamento de Proteína Pós-Traducional , Ativação Transcricional
5.
Oncogene ; 32(4): 479-90, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-22391565

RESUMO

Inadvertent mammalian tissue exposures to low doses of ionizing radiation (IR) after radiation accidents, remediation of radioactive-contaminated areas, space travel or a dirty bomb represent an interesting trauma to an organism. Possible low-dose IR-induced bystander effects could impact our evaluation of human health effects, as cells within tissue are not equally damaged after doses of IR ≤10 cGy. To understand tissue responses after low IR doses, we generated a reporter system using the human clusterin promoter fused to firefly luciferase (hCLUp-Luc). Secretory clusterin (sCLU), an extracellular molecular chaperone, induced by low doses of cytotoxic agents, clears cell debris. Low-dose IR (≥2 cGy) exposure induced hCLUp-Luc activity with peak levels at 96 h, consistent with endogenous sCLU levels. As doses increased (≥1 Gy), sCLU induction amplitudes increased and time-to-peak response decreased. sCLU expression was stimulated by insulin-like growth factor-1, but suppressed by p53. Responses in transgenic hCLUp-Luc reporter mice after low IR doses showed that specific tissues (that is, colon, spleen, mammary, thymus and bone marrow) of female mice induced hCLUp-Luc activity more than male mice after whole body (≥10 cGy) irradiation. Tissue-specific, non-linear dose- and time-responses of hCLUp-Luc and endogenous sCLU levels were noted. Colon maintained homeostatic balance after 10 cGy. Bone marrow responded with delayed, but prolonged and elevated expression. Intraperitoneal administration of α-transforming growth factor (TGF)ß1 (1D11), but not control (13C4) antibodies, immediately following IR exposure abrogated CLU induction responses. Induction in vivo also correlated with Smad signaling by activated TGFß1 after IR. Mechanistically, media with elevated sCLU levels suppressed signaling, blocked apoptosis and increased survival of TGFß1-exposed tumor or normal cells. Thus, sCLU is a pro-survival bystander factor that abrogates TGFß1 signaling and most likely promotes wound healing.


Assuntos
Clusterina/genética , Raios gama , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Transformador beta1/genética , Proteína Supressora de Tumor p53/genética , Irradiação Corporal Total , Animais , Apoptose/genética , Medula Óssea/metabolismo , Medula Óssea/efeitos da radiação , Linhagem Celular Tumoral , Clusterina/metabolismo , Colo/metabolismo , Colo/efeitos da radiação , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/efeitos da radiação , Feminino , Células HCT116 , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Células MCF-7 , Masculino , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais/efeitos da radiação , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteína Supressora de Tumor p53/metabolismo
6.
Oncogene ; 30(35): 3745-54, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21460853

RESUMO

Secretory clusterin (sCLU) is a stress-induced, pro-survival glycoprotein elevated in early-stage cancers, in particular in APC/Min-defective colon cancers. sCLU is upregulated after exposure to various cytotoxic agents, including ionizing radiation (IR), leading to a survival advantage. We found that stimulation of insulin-like growth factor-1 (IGF-1) and IGF-1R protein kinase signaling was required for sCLU induction after IR exposure. Here, we show that activation of Ataxia telangiectasia-mutated kinase (ATM) by endogenous or exogenous forms of DNA damage was required to relieve basal repression of IGF-1 transcription by the p53/NF-YA complex, leading to sCLU expression. Although p53 levels were stabilized and elevated after DNA damage, dissociation of NF-YA, and thereby p53, from the IGF-1 promoter resulted in IGF-1 induction, indicating that NF-YA was rate limiting. Cells with elevated endogenous DNA damage (deficient in H2AX, MDC1, NBS1, mTR or hMLH1) or cells exposed to DNA-damaging agents had elevated IGF-1 expression, resulting in activation of IGF-1R signaling and sCLU induction. In contrast, ATM-deficient cells were unable to induce sCLU after DNA damage. Our results integrate DNA damage resulting from genetic instability, IR, or chemotherapeutic agents, to ATM activation and abrogation of p53/NF-YA-mediated IGF-1 transcriptional repression, that induces IGF-1-sCLU expression. Elucidation of this pathway should uncover new mechanisms for cancer progression and reveal new targets for drug development to overcome resistance to therapy.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Clusterina/genética , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Instabilidade Genômica , Fator de Crescimento Insulin-Like I/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Fator de Ligação a CCAAT/metabolismo , Proteínas de Ciclo Celular/genética , Linhagem Celular , Proteínas de Ligação a DNA/genética , Humanos , Neoplasias/patologia , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/genética
7.
Breast Cancer Res Treat ; 130(1): 41-8, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21153485

RESUMO

We sequenced the 5' UTR of the estrogen-related receptor gamma gene (ERR-γ) in ~500 patient and volunteer samples and found that longer alleles of the (AAAG)(n) microsatellite were statistically and significantly more likely to exist in the germlines of breast cancer patients when compared to healthy volunteers. This microsatellite region contains multiple binding sites for a number of transcription factors, and we hypothesized that the polymorphic AAAG-containing sequence in the 5' UTR region of ERR-γ might modulate expression of ERR-γ. We found that the 369 bp PCR product containing the AAAG repeat drove expression of a reporter gene in estrogen receptor positive breast cancer cells. Our results support a role for the 5' UTR region in ERR-γ expression, which is potentially mediated via binding to the variable tandem AAAG repeat, the length of which correlates with breast cancer pre-disposition. Our study indicates that the AAAG tetranucleotide repeat polymorphism in ERR-γ gene 5' UTR region may be a new biomarker for genetic susceptibility to breast cancer.


Assuntos
Regiões 5' não Traduzidas , Alelos , Neoplasias da Mama/genética , Predisposição Genética para Doença , Repetições de Microssatélites , Regiões Promotoras Genéticas , Receptores de Estrogênio/genética , Animais , Sequência de Bases , Biomarcadores Tumorais/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Genes Reporter , Genótipo , Humanos , Dados de Sequência Molecular , Polimorfismo Genético , Receptores de Estrogênio/metabolismo
8.
J Steroid Biochem Mol Biol ; 103(3-5): 721-5, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17224269

RESUMO

Prostate cancer is the most commonly diagnosed cancer in the majority of western countries. Due to their antiproliferative and proapoptotic activity, vitamin D analogues have been introduced recently as an experimental therapy for prostate cancer. Clusterin (CLU) is a glycoprotein that has two known isoforms generated in human cells. A nuclear form of CLU protein (nCLU) is pro-apoptotic, and a secretory form (sCLU) is pro-survival. In this study, we analyzed whether proapoptotic and antiproliferative effects of 1,25(OH)(2)D(3) on LNCaP prostate cancer cells are modulated by expression of sCLU. Using colony forming assay, we studied the effect of treatment with different doses of 1,25(OH)(2)D(3) (10(-6), 10(-7), 10(-10)M) on proliferation of LNCaP cells that were stable transfected and over-express sCLU (LNT-1) as compared to empty vector-transfected cells (LN/C). We also measured apoptosis using TUNEL assay. sCLU over-expression protected against both antiproliferative (30%) and proapoptotic (15%) effects of 1,25(OH)(2)D(3), although this effect was statistically not significant. In conclusion, our findings demonstrate that expression of sCLU modulates growth regulatory effects of 1,25(OH)(2)D(3) in prostate cancer indicating that CLU interferes with vitamin D signalling pathways.


Assuntos
Apoptose/efeitos dos fármacos , Calcitriol/farmacologia , Clusterina/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Clusterina/genética , Humanos , Masculino , Neoplasias da Próstata/genética , Sensibilidade e Especificidade
9.
J Mol Histol ; 37(5-7): 183-8, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17048076

RESUMO

The glycoprotein clusterin (CLU), has two known isoforms generated in human cells. A nuclear form of CLU protein (nCLU) is pro-apoptotic, while a secretory form (sCLU) is pro-survival. Both forms are implicated in various cell functions, including DNA repair, cell cycle regulation, and apoptotic cell death. CLU expression has been associated with tumorigenesis and the progression of various malignancies. In response to DNA damage, cell survival can be enhanced by activation of DNA repair mechanisms, while simultaneously stimulating energy-expensive cell cycle checkpoints that delay the cell cycle progression to allow more time for DNA repair. This review summarizes our current understanding of the role of clusterin in DNA repair, apoptosis, and cell cycle control and the relevance.


Assuntos
Ciclo Celular , Clusterina/fisiologia , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Modelos Genéticos , Neoplasias/etiologia , Neoplasias/genética , Apoptose , Clusterina/genética , Humanos , Neoplasias/metabolismo , Isoformas de Proteínas/fisiologia
10.
Cancer Res ; 61(22): 8290-7, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11719462

RESUMO

Our previous data demonstrated that cells deficient in MutL homologue-1 (MLH1) expression had a reduced and shorter G(2) arrest after high-dose-rate ionizing radiation (IR), suggesting that the mismatch re pair (MMR) system mediates this cell cycle checkpoint. We confirmed this observation using two additional isogenetically matched human MLH1 (hMLH1)-deficient and -proficient human tumor cell systems: human ovarian cancer cells, A2780/CP70, with or without ectopically expressed hMLH1, and human colorectal carcinoma cells, RKO, with or without azacytidine treatment to reexpress hMLH1. We also examined matched MutS homologue-2 (hMSH2)-deficient and -proficient human endometrial carcinoma HEC59 cell lines to determine whether hMSH2, and MMR in general, is involved in IR-related G(2) arrest responses. As in MLH1-deficient cells, cells lacking hMSH2 demonstrated a similarly altered G(2) arrest in response to IR (6 Gy). These differences in IR-induced G(2) arrest between MMR-proficient and -deficient cells were found regardless of whether synchronized cells were irradiated in G(0)/G(1) or S phase, indicating that MMR indeed dramatically affects the G(2)-M checkpoint arrest. However, unlike the MMR-dependent damage tolerance response to 6-thioguanine exposures, no significant difference in the clonogenic survival of MMR-deficient cells compared with MMR-proficient cells was noted after high-dose-rate IR. In an attempt to define the signal transduction mechanisms responsible for MMR-mediated G(2) arrest, we examined the levels of tyrosine 15 phosphorylation of cdc2 (phospho-Tyr15-cdc2), a key regulator of the G(2)-M transition. Increased phospho-Tyr15-cdc2 levels were observed in both MMR-proficient and -deficient cell lines after IR. However, the levels of the phospho-Tyr15-cdc2 rapidly decreased in MMR (hMLH1 or hMSH2)-deficient cell lines at times coincident with progress from the IR-induced G(2) arrest through M phase. Thus, differences in the levels of phospho-Tyr15-cdc2 after high-dose-rate IR correspond temporally with the observed differences in the IR-induced G(2) arrest, suggesting that MMR proteins may exert their effect on IR-induced G(2) arrest by signaling the cdc2 pathway. Although MMR status does not significantly affect the survival of cells after high-dose-rate IR, it seems to regulate the G(2)-M checkpoint and might affect overall mutation rates.


Assuntos
Pareamento Incorreto de Bases , Proteína Quinase CDC2/fisiologia , Reparo do DNA/fisiologia , Fase G2/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Proteína Quinase CDC2/metabolismo , Proteínas de Transporte , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Fase G2/efeitos dos fármacos , Fase G2/efeitos da radiação , Humanos , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/deficiência , Proteínas Nucleares , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosforilação , Fase S/efeitos dos fármacos , Fase S/fisiologia , Fase S/efeitos da radiação , Transdução de Sinais/fisiologia , Transdução de Sinais/efeitos da radiação , Tioguanina/farmacologia , Células Tumorais Cultivadas
11.
Semin Radiat Oncol ; 11(4): 352-72, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11677660

RESUMO

Cellular responses to ionizing radiation (IR) include (a) activation of signal transduction enzymes; (b) stimulation of DNA repair, most notably DNA double strand break (DSB) repair by homologous or nonhomologous recombinatorial pathways; (c) activation of transcription factors and subsequent IR-inducible transcript and protein changes; (d) cell cycle checkpoint delays in G(1), S, and G(2) required for repair or for programmed cell death of severely damaged cells; (e) activation of zymogens needed for programmed cell death (although IR is a poor inducer of such responses in epithelial cells); and (f) stimulation of IR-inducible proteins that may mediate bystander effects influencing signal transduction, DNA repair, angiogenesis, the immune response, late responses to IR, and possibly adaptive survival responses. The overall response to IR depends on the cell's inherent genetic background, as well as its ability to biochemically and genetically respond to IR-induced damage. To improve the anti-tumor efficacy of IR, our knowledge of these pleiotropic responses must improve. The most important process for the survival of a tumor cell following IR is the repair of DNA double strand breaks (DSBs). Using yeast two-hybrid analyses along with other molecular and cellular biology techniques, we cloned transcripts/proteins that are involved in, or presumably affect, nonhomologous DNA double strand break end-joining (NHEJ) repair mediated by the DNA-PK complex. Using Ku70 as bait, we isolated a number of Ku-binding proteins (KUBs). We identified the first X-ray-inducible transcript/protein (xip8, Clusterin (CLU)) that associates with DNA-PK. A nuclear form of CLU (nCLU) prevented DNA-PK-mediated end joining, and stimulated cell death in response to IR or when overexpressed in the absence of IR. Structure-function analyses using molecular and cellular (including green fluorescence-tagged protein trafficking) biology techniques showed that nCLU appears to be an inactive protein residing in the cytoplasm of epithelial cells. Following IR injury, nCLU levels increase and an as yet undefined posttranslational modification appears to alter the protein, exposing nuclear localization sequences (NLSs) and coiled-coil domains. The modified protein translocates to the nucleus and triggers cell death, presumably through its interaction specifically with Ku70. Understanding nCLU responses, as well as the functions of the KUBs, will be important for understanding DSB repair. Knowledge of DSB repair may be used to improve the antitumor efficacy of IR, as well as other chemotherapeutic agents.


Assuntos
Reparo do DNA/fisiologia , DNA de Neoplasias/metabolismo , Neoplasias/radioterapia , Sobrevivência Celular , Dano ao DNA , Reparo do DNA/genética , DNA de Neoplasias/genética , Humanos , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Radiação Ionizante , Transdução de Sinais
12.
Exp Cell Res ; 267(1): 95-106, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11412042

RESUMO

beta-Lapachone (beta-lap) induces apoptosis in various cancer cells, and its intracellular target has recently been elucidated in breast cancer cells. Here we show that NAD(P)H:quinone oxidoreductase (NQO1/xip3) expression in human prostate cancer cells is a key determinant for apoptosis and lethality after beta-lap exposures. beta-Lap-treated, NQO1-deficient LNCaP cells were significantly more resistant to apoptosis than NQO1-expressing DU-145 or PC-3 cells after drug exposures. Formation of an atypical 60-kDa PARP cleavage fragment in DU-145 or PC-3 cells was observed after 10 microM beta-lap treatment and correlated with apoptosis. In contrast, LNCaP cells required 25 microM beta-lap to induce similar responses. Atypical PARP cleavage in beta-lap-treated cells was not affected by 100 microM zVAD-fmk; however, coadministration of dicoumarol, a specific inhibitor of NQO1, reduced beta-lap-mediated cytotoxicity, apoptosis, and atypical PARP cleavage in NQO1-expressing cells. Dicoumarol did not affect the more beta-lap-resistant LNCaP cells. Stable transfection of LNCaP cells with NQO1 increased their sensitivity to beta-lap, enhancing apoptosis compared to parental LNCaP cells or vector-alone transfectants. Dicoumarol increased survival of beta-lap-treated NQO1-expressing LNCaP transfectants. NQO1 activity, therefore, is a key determinant of beta-lap-mediated apoptosis and cytotoxicity in prostate cancer cells.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose , NAD(P)H Desidrogenase (Quinona)/metabolismo , Naftoquinonas/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Camptotecina/farmacologia , Caspases/metabolismo , Dicumarol/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , NAD(P)H Desidrogenase (Quinona)/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Transformação Genética , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
13.
Cancer Res ; 61(13): 5193-201, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11431359

RESUMO

DNA mismatch repair (MMR) is an efficient system for the detection and repair of mismatched and unpaired bases in DNA. Deficiencies in MMR are commonly found in both hereditary and sporadic colorectal cancers, as well as in cancers of other tissues. Because fluorinated thymidine analogues (which through their actions might generate lesions recognizable by MMR) are widely used in the treatment of colorectal cancer, we investigated the role of MMR in cellular responses to 5-fluorouracil and 5-fluoro-2'-deoxyuridine (FdUrd). Human MLH1(-) and MMR-deficient HCT116 colon cancer cells were 18-fold more resistant to 7.5 microM 5-fluorouracil (continuous treatment) and 17-fold more resistant to 7.5 microM FdUrd in clonogenic survival assays compared with genetically matched, MLH1(+) and MMR-proficient HCT116 3-6 cells. Likewise, murine MLH1(-) and MMR-deficient CT-5 cells were 3-fold more resistant to a 2-h pulse of 10 microM FdUrd than their MLH1(+) and MMR-proficient ME-10 counterparts. Decreased cytotoxicity in MMR-deficient cells after treatment with various methylating agents and other base analogues has been well reported and is believed to reflect a tolerance to DNA damage. Synchronized HCT116 3-6 cells treated with a low dose of FdUrd had a 2-fold greater G(2) cell cycle arrest compared with MMR-deficient HCT116 cells, and asynchronous ME-10 cells demonstrated a 4-fold greater G(2) arrest after FdUrd treatment compared with CT-5 cells. Enhanced G(2) arrest in MMR-proficient cells in response to other agents has been reported and is believed to allow time for DNA repair. G(2) cell cycle arrest as determined by propidium iodide staining was not a result of mitotic arrest, but rather a true G(2) arrest, as indicated by elevated cyclin B1 levels and a lack of staining with mitotic protein monoclonal antibody 2. Additionally, p53 and GADD45 levels were induced in FdUrd-treated HCT116 3-6 cells. DNA double-strand break (DSB) formation was 2-fold higher in MMR-proficient HCT116 3-6 cells after FdUrd treatment, as determined by pulsed-field gel electrophoresis. The formation of DSBs was not the result of enhanced apoptosis in MMR-proficient cells. FdUrd-mediated cytotoxicity was caused by DNA-directed and not RNA-directed effects, because administration of excess thymidine (and not uridine) prevented cytotoxicity, cell cycle arrest, and DSB formation. hMLH1-dependent responses to fluoropyrimidine treatment, which may involve the action of p53 and the formation of DSBs, clearly have clinical relevance for the use of this class of drugs in the treatment of tumors with MMR deficiencies.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Reparo do DNA/fisiologia , Floxuridina/farmacologia , Fluoruracila/farmacologia , Proteínas de Neoplasias/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Ciclina B/metabolismo , Ciclina B1 , Dano ao DNA , Resistencia a Medicamentos Antineoplásicos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fase G2/efeitos dos fármacos , Fase G2/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Mitose/efeitos dos fármacos , Mitose/fisiologia , Proteína 1 Homóloga a MutL , Proteínas de Neoplasias/deficiência , Proteínas Nucleares , Proteínas/metabolismo , Coloração e Rotulagem/métodos , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Proteínas GADD45
14.
J Biol Chem ; 276(22): 19150-9, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11279125

RESUMO

beta-Lapachone (beta-Lap) triggers apoptosis in a number of human breast and prostate cancer cell lines through a unique apoptotic pathway that is dependent upon NQO1, a two-electron reductase. Downstream signaling pathway(s) that initiate apoptosis following treatment with beta-Lap have not been elucidated. Since calpain activation was suspected in beta-Lap-mediated apoptosis, we examined alterations in Ca(2+) homeostasis using NQO1-expressing MCF-7 cells. beta-Lap-exposed MCF-7 cells exhibited an early increase in intracellular cytosolic Ca(2+), from endoplasmic reticulum Ca(2+) stores, comparable to thapsigargin exposures. 1,2-Bis-(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester, an intracellular Ca(2+) chelator, blocked early increases in Ca(2+) levels and inhibited beta-Lap-mediated mitochondrial membrane depolarization, intracellular ATP depletion, specific and unique substrate proteolysis, and apoptosis. The extracellular Ca(2+) chelator, EGTA, inhibited later apoptotic end points (observed >8 h, e.g. substrate proteolysis and DNA fragmentation), suggesting that later execution events were triggered by Ca(2+) influxes from the extracellular milieu. Collectively, these data suggest a critical, but not sole, role for Ca(2+) in the NQO1-dependent cell death pathway initiated by beta-Lap. Use of beta-Lap to trigger an apparently novel, calpain-like-mediated apoptotic cell death could be useful for breast and prostate cancer therapy.


Assuntos
Cálcio/metabolismo , Morte Celular , Naftoquinonas/metabolismo , Transdução de Sinais , 4-Nitroquinolina-1-Óxido/farmacologia , Trifosfato de Adenosina/metabolismo , Apoptose , Western Blotting , Neoplasias da Mama/metabolismo , Divisão Celular , Quelantes/farmacologia , Citosol/metabolismo , Fragmentação do DNA , Relação Dose-Resposta a Droga , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Retículo Endoplasmático/metabolismo , Feminino , Citometria de Fluxo , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Potenciais da Membrana , Microscopia Confocal , Mitocôndrias/metabolismo , Modelos Biológicos , Naftoquinonas/farmacologia , Neoplasias da Próstata/metabolismo , Quinolonas/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas
15.
Proc Natl Acad Sci U S A ; 97(11): 5907-12, 2000 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-10823943

RESUMO

Clusterin [CLU, a.k.a. TRPM-2, SGP-2, or ionizing radiation (IR)-induced protein-8 (XIP8)] was implicated in apoptosis, tissue injury, and aging. Its function remains elusive. We reisolated CLU/XIP8 by yeast two-hybrid analyses using as bait the DNA double-strand break repair protein Ku70. We show that a delayed (2-3 days), low-dose (0.02-10 Gy) IR-inducible nuclear CLU/XIP8 protein coimmunoprecipitated and colocalized (by confocal microscopy) in vivo with Ku70/Ku80, a DNA damage sensor and key double-strand break repair protein, in human MCF-7:WS8 breast cancer cells. Overexpression of nuclear CLU/XIP8 or its minimal Ku70 binding domain (120 aa of CLU/XIP8 C terminus) in nonirradiated MCF-7:WS8 cells dramatically reduced cell growth and colony-forming ability concomitant with increased G(1) cell cycle checkpoint arrest and increased cell death. Enhanced expression and accumulation of nuclear CLU/XIP8-Ku70/Ku80 complexes appears to be an important cell death signal after IR exposure.


Assuntos
Antígenos Nucleares , Morte Celular/fisiologia , DNA Helicases , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Glicoproteínas/fisiologia , Chaperonas Moleculares , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae , Adenocarcinoma/patologia , Motivos de Aminoácidos , Neoplasias da Mama/patologia , Núcleo Celular/metabolismo , Clusterina , Dano ao DNA , DNA Complementar/genética , DNA de Neoplasias/efeitos da radiação , Proteína Quinase Ativada por DNA , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Biblioteca Gênica , Genes Reporter , Glicoproteínas/química , Glicoproteínas/genética , Proteínas de Fluorescência Verde , Humanos , Autoantígeno Ku , Proteínas Luminescentes/genética , Microscopia Confocal , Ligação Proteica , Precursores de Proteínas/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Saccharomyces cerevisiae/genética , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
16.
Exp Cell Res ; 255(2): 144-55, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10694431

RESUMO

beta-Lapachone (beta-lap) effectively killed MCF-7 and T47D cell lines via apoptosis in a cell-cycle-independent manner. However, the mechanism by which this compound activated downstream proteolytic execution processes were studied. At low concentrations, beta-lap activated the caspase-mediated pathway, similar to the topoisomerase I poison, topotecan; apoptotic reactions caused by both agents at these doses were inhibited by zVAD-fmk. However at higher doses of beta-lap, a novel non-caspase-mediated "atypical" cleavage of PARP (i.e., an approximately 60-kDa cleavage fragment) was observed. Atypical PARP cleavage directly correlated with apoptosis in MCF-7 cells and was inhibited by the global cysteine protease inhibitors iodoacetamide and N-ethylmaleimide. This cleavage was insensitive to inhibitors of caspases, granzyme B, cathepsins B and L, trypsin, and chymotrypsin-like proteases. The protease responsible appears to be calcium-dependent and the concomitant cleavage of PARP and p53 was consistent with a beta-lap-mediated activation of calpain. beta-Lap exposure also stimulated the cleavage of lamin B, a putative caspase 6 substrate. Reexpression of procaspase-3 into caspase-3-null MCF-7 cells did not affect this atypical PARP proteolytic pathway. These findings demonstrate that beta-lap kills cells through the cell-cycle-independent activation of a noncaspase proteolytic pathway.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Cisteína Endopeptidases/metabolismo , Naftoquinonas/farmacologia , Ativação Enzimática/efeitos dos fármacos , Feminino , Humanos , Células Tumorais Cultivadas
17.
J Biol Chem ; 275(13): 9501-9, 2000 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-10734098

RESUMO

Activation of the transcription factor NF-kappaB by extracellular signals involves its release from the inhibitor protein IkappaBalpha in the cytoplasm and subsequent nuclear translocation. NF-kappaB can also be activated by the anticancer agent camptothecin (CPT), which inhibits DNA topoisomerase (Topo) I activity and causes DNA double-strand breaks during DNA replication to induce S phase-dependent cytotoxicity. Here we show that CPT activates NF-kappaB by a mechanism that is dependent on initial nuclear DNA damage followed by cytoplasmic signaling events. NF-kappaB activation by CPT is dramatically diminished in cytoplasts and in CEM/C2 cells expressing a mutant Topo I protein that fails to bind CPT. This response is intensified in S phase cell populations and is prevented by the DNA polymerase inhibitor aphidicolin. In addition, CPT activation of NF-kappaB involves degradation of cytoplasmic IkappaBalpha by the ubiquitin-proteasome pathway in a manner that depends on the IkappaB kinase complex. Finally, inhibition of NF-kappaB activation augments CPT-induced apoptosis. These findings elucidate the progression of signaling events that initiates in the nucleus with CPT-Topo I interaction and continues in the cytoplasm resulting in degradation of IkappaBalpha and nuclear translocation of NF-kappaB to attenuate the apoptotic response.


Assuntos
Camptotecina/farmacologia , Núcleo Celular/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Dano ao DNA , Proteínas I-kappa B , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Apoptose , Sequência de Bases , Núcleo Celular/metabolismo , Cisteína Endopeptidases/metabolismo , Citoplasma/metabolismo , Primers do DNA , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Complexos Multienzimáticos/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Complexo de Endopeptidases do Proteassoma , Fase S/efeitos dos fármacos , Topotecan/farmacologia , Ubiquitinas/metabolismo
18.
J Biol Chem ; 275(8): 5416-24, 2000 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-10681517

RESUMO

beta-Lapachone activates a novel apoptotic response in a number of cell lines. We demonstrate that the enzyme NAD(P)H:quinone oxidoreductase (NQO1) substantially enhances the toxicity of beta-lapachone. NQO1 expression directly correlated with sensitivity to a 4-h pulse of beta-lapachone in a panel of breast cancer cell lines, and the NQO1 inhibitor, dicoumarol, significantly protected NQO1-expressing cells from all aspects of beta-lapachone toxicity. Stable transfection of the NQO1-deficient cell line, MDA-MB-468, with an NQO1 expression plasmid increased apoptotic responses and lethality after beta-lapachone exposure. Dicoumarol blocked both the apoptotic responses and lethality. Biochemical studies suggest that reduction of beta-lapachone by NQO1 leads to a futile cycling between the quinone and hydroquinone forms, with a concomitant loss of reduced NAD(P)H. In addition, the activation of a cysteine protease, which has characteristics consistent with the neutral calcium-dependent protease, calpain, is observed after beta-lapachone treatment. This is the first definitive elucidation of an intracellular target for beta-lapachone in tumor cells. NQO1 could be exploited for gene therapy, radiotherapy, and/or chemopreventive interventions, since the enzyme is elevated in a number of tumor types (i.e. breast and lung) and during neoplastic transformation.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Neoplasias da Mama/enzimologia , Divisão Celular/efeitos dos fármacos , Naftoquinonas/farmacologia , Quinona Redutases/metabolismo , Apoptose/efeitos dos fármacos , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Redutases do Citocromo/metabolismo , Citocromo-B(5) Redutase , Dicumarol/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Humanos , Modelos Biológicos , NAD/metabolismo , NADH NADPH Oxirredutases/metabolismo , NADPH-Ferri-Hemoproteína Redutase , Proteínas/metabolismo , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo , Vitamina K/farmacologia
19.
FASEB J ; 14(2): 379-90, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10657994

RESUMO

Regulation of transcriptional responses in growth-arrested human cells under conditions that promote potentially lethal damage repair after ionizing radiation (IR) is poorly understood. Sp1/retinoblastoma control protein (RCP) DNA binding increased within 30 min and peaked at 2-4 h after IR (450-600 cGy) in confluent radioresistant human malignant melanoma (U1-Mel) cells. Increased phosphorylation of Sp1 directly corresponded to Sp1/RCP binding and immediate-early gene induction, whereas pRb remained hypophosphorylated. Transfection of U1-Mel cells with the human papillomavirus E7 gene abrogated Sp1/RCP induction and G(0)/G(1) cell cycle checkpoint arrest responses, increased apoptosis and radiosensitivity, and augmented genetic instability (i.e., increased polyploidy cells) after IR. Increased NF-kappaB DNA binding in U1-Mel cells after IR treatment lasted much longer (i.e., >20 h). U1-Mel cells overexpressing dominant-negative IkappaBalpha S32/36A mutant protein were significantly more resistant to IR exposure and retained both G(2)/M and G(0)/G(1) cell cycle checkpoint responses without significant genetic instability (i.e., polyploid cell populations were not observed). Nuclear p53 protein levels and DNA binding activity increased only after high doses of IR (>1200 cGy). Disruption of p53 responses in U1-Mel cells by E6 transfection also abrogated G(0)/G(1) cell cycle checkpoint arrest responses and increased polyploidy after IR, but did not alter radiosensitivity. These data suggest that abrogation of individual components of this coordinate IR-activated transcription factor response may lead to divergent alterations in cell cycle checkpoints, genomic instability, apoptosis, and survival. Such coordinate transcription factor activation in human cancer cells is reminiscent of prokaryotic SOS responses, and further elucidation of these events should shed light on the initial molecular events in the chromosome instability phenotype.-Yang, C.-R., Wilson-Van Patten, C., Planchon, S. M., Wuerzberger-Davis, S. M., Davis, T. W., Cuthill, C., Miyamoto, S., Boothman, D. A. Coordinate modulation of Sp1, NF-kappa B, and p53 in confluent human malignant melanoma cells after ionizing radiation.


Assuntos
Melanoma Experimental/radioterapia , NF-kappa B/metabolismo , Fator de Transcrição Sp1/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ciclo Celular/efeitos da radiação , Relação Dose-Resposta à Radiação , Raios gama , Humanos , Melanoma Experimental/metabolismo , Tolerância a Radiação , Proteína do Retinoblastoma , Raios X
20.
Ann N Y Acad Sci ; 922: 274-92, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11193903

RESUMO

The efficacy of topoisomerase (Topo) I-active drugs may be improved by better understanding the molecular and cellular responses of tumor compared to normal cells after genotoxic insults. Ionizing radiation (IR) + Topo I-active drugs (e.g., Topotecan) caused synergistic cell killing in various human cancer cells, even in cells from highly radioresistant tumors. Topo I poisons had to be added either during or immediately after IR. Synergy was caused by DNA lesion modification mechanisms as well as by concomitant stimulation of two pathways of cell death: necrosis (IR) + apoptosis (Topo I poisons). Cumulative data favor a mechanism of synergistic cell killing caused by altered DNA lesion modification and enhanced apoptosis. However, alterations in cell cycle regulation may also play a role in the synergy between these two agents in certain human cancers. We recently showed that NF-kappa B, a known anti-apoptotic factor, was activated in various cancer cells after poisoning Topo I using clinically active drugs. NF-kappa B activation was dependent on initial nuclear DNA damage followed by cytoplasmic signaling events. Cytoplasmic signaling leading to NF-kappa B activation after Topo I poisons was diminished in cytoplasts (lacking nuclei) and in CEM/C2 cells that expressed a mutant Topo I protein that did not interact with Topo I-active drugs. NF-kappa B activation was intensified in S-phase and blocked by aphidicolin, suggesting that activation was a result of double-strand break formation due to Topo I poisoning and DNA replication. Dominant-negative I kappa B expression augmented Topo I poison-mediated apoptosis. Elucidation of molecular signal transduction pathways after Topo I drug-IR combinations may lead to improved radiotherapy by blocking anti-apoptotic NF-kappa B responses. Recent data also indicate that synergy caused by IR + Topo I poisons is different from radiosensitization by beta-lapachone (beta-lap), a "reported" Topo I and II-alpha poison in vitro. In fact, beta-lap does not kill cells by poisoning either Topo I or II-alpha in vivo. Instead, the compound is "activated" by an IR (damage)-inducible enzyme, NAD(P)H:quinone oxidoreductase (NQO1), a gene cloned as x-ray-inducible transcript #3, xip3. Unlike the lesion modification pathway induced by IR + Topo I drugs, beta-lap kills cells via NQO1 futile cycle metabolism. Downstream apoptosis caused by beta-lap appears to be noncaspase-mediated, involving calpain or a calpain-like protease. Thus, although Topo I poisons or beta-lap in combination with IR both synergistically kill cancer cells, the mechanisms are very different.


Assuntos
Inibidores Enzimáticos/farmacologia , Naftoquinonas/farmacologia , Radiossensibilizantes/farmacologia , Inibidores da Topoisomerase I , Animais , Bovinos , DNA Topoisomerases Tipo I/fisiologia , Humanos , NAD(P)H Desidrogenase (Quinona)/biossíntese , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/fisiologia , NF-kappa B/fisiologia , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA