Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Mol Carcinog ; 54(10): 1205-13, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25044025

RESUMO

DNAJB6 is a member of the heat shock protein 40 (Hsp40) family. We here investigated the clinical correlation and biological role of DNAJB6 overexpression in colorectal cancer (CRC). The expression of DNAJB6 protein was examined in 200 cases of colorectal adenocarcinomas by immunohistochemistry (IHC) technology. Gene transfection and RNA interference were performed to determine the effect of DNAJB6 expression on the invasion of CRC cells and to explore the underlying molecular mechanisms in vitro and in vivo. Overexpression of DNAJB6 was found in 39% (78/200) of the CRC tissues, especially in tumors at pT4 as compared with at pT1-3 (P = 0.02). A Kaplan-Meier survival analysis revealed a correlation between DNAJB6 expression and overall survival (OS) times (P = 0.003). Multivariate analysis confirmed that DNAJB6 overexpression was an independent prognostic factor for CRC (P = 0.002). RNA interference-mediated silencing of the DNAJB6 gene inhibited the invasion of CRC cells in vitro were accompanied by a significant reduction in the protein levels of IQ-domain GTPase-activating protein 1 (IQGAP1) and phosphorylated ERK (pERK). An in vivo assay showed that inhibition of DNAJB6 expression decreased the lung metastases of CRC cells. IHC analysis of serial sections showed that there was a positive correlation between DNAJB6 and IQGAP1 expression in primary CRC tissues (P = 0.013). The data suggest that DNAJB6 plays an important oncogenic role in CRC cell invasion by up-regulating IQGAP1 and activating the ERK signaling pathway and that DNAJB6 may be used as a prognostic marker for CRC.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Proteínas de Choque Térmico HSP40/genética , Sistema de Sinalização das MAP Quinases/genética , Chaperonas Moleculares/genética , Invasividade Neoplásica/genética , Proteínas do Tecido Nervoso/genética , Transdução de Sinais/genética , Proteínas Ativadoras de ras GTPase/genética , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Células HCT116 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Fosforilação/genética , Prognóstico , Interferência de RNA/fisiologia , Regulação para Cima/genética
2.
Mol Carcinog ; 53(7): 514-25, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23359356

RESUMO

Atypical protein kinase Cι (PKCι) has been identified as an oncoprotein in esophageal squamous cell carcinomas. However, the mechanisms underlying the role of PKCι in this disease remain unclear. In the present work, we found that inhibition of PKCι expression by RNAi induced apoptosis via the down-regulation of ß-catenin in esophageal cancer cells. Furthermore, we found that PKCι regulated ß-catenin in an autophagy dependent way. Since down-regulation of ß-catenin induced by knockdown of PKCι could be rescued by autophagy inhibition; knockdown of PKCι activated autophagy and promoted the recruitment of ß-catenin into autophagosome. These results suggested that PKCι positively regulated ß-catenin through negatively regulated autophagy and depletion of PKCι promoted apoptosis via autophagic degradation of ß-catenin in esophageal cancer cells. These data provide new insights into PKCι signaling in human cancer.


Assuntos
Apoptose/genética , Autofagia/genética , Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Isoenzimas/genética , Proteína Quinase C/genética , beta Catenina/genética , Proteína 5 Relacionada à Autofagia , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/genética , Proteínas Associadas aos Microtúbulos/genética , Interferência de RNA , RNA Interferente Pequeno/genética , beta Catenina/biossíntese , beta Catenina/metabolismo
3.
Acta Pharmacol Sin ; 35(9): 1199-206, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25152024

RESUMO

AIM: Mig-2 (also known as Kindlin-2 and FERMT2) is an important regulator of integrin activation and cell-extracellular matrix adhesion, and involved in carcinogenesis and tumor progression. The aim of this study was to investigate the role of mig-2 in cisplatin-induced apoptosis of human glioma cells in vitro. METHODS: The expression of mig-2 was modulated in human glioma H4, HS 683 and U-87 MG cells by transfection with a plasmid carrying mig-2 or mig-2 siRNA. Cisplatin-induced apoptosis was detected using Annexin V/PI staining and flow cytometry, as well as MTS analyses. The expression of apoptosis-related or signaling proteins was examined using Western blotting analysis. H4 cells were transfected with plasmids carrying mig-2 mutants to determine the functional domain of mig-2. RESULTS: In the 3 glioma cell lines tested, overexpression of mig-2 significantly attenuated cisplatin-induced apoptosis, whereas knock-down of mig-2 potentiated the apoptosis. The mechanisms of action of mig-2 were further addressed in H4 cells: overexpression of mig-2 markedly reduced cleaved caspase-9, caspase-8, caspase-3 and PARP, as well as p-JNK and p-p38, and increased p-AKT in cisplatin-treated H4 cells, whereas mig-2 siRNA reversely changed these apoptosis-related and signaling proteins. Furthermore, pretreatment with JNK inhibitor SP600125 and p38 inhibitor SB203580, or with AKT inhibitor LY294002 abolished the effects of mig-2 on cisplaxtin-induced apoptosis. In H4 cells, GFP-mig-2 F3 plasmid that contained only the F3 subdomain showed the same efficiency in attenuating cisplatin-induced apoptosis, as the mig-2 wild-type vector did, whereas GFP-mig-2 (1-541) plasmid that lacked the F3 subdomain was inactive. CONCLUSION: Mig-2 significantly attenuates the antitumor action of cisplatin against human glioma cells in vitro through AKT/JNK and AKT/p38 signaling pathways. The F3 subdomain of mig-2 is necessary and sufficient for this effect.


Assuntos
Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Glioma/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral , Humanos , Transdução de Sinais/efeitos dos fármacos
4.
Gastroenterology ; 142(3): 521-530.e3, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22108192

RESUMO

BACKGROUND & AIMS: Aberrant activation of the signal transducer and activator of transcription (Stat)3 and overexpression of polo-like kinase (PLK)1 each have been associated with cancer pathogenesis. The mechanisms and significance of dysregulation of Stat3 and PLK1 in carcinogenesis and cancer progression are unclear. We investigated the relationship between Stat3 and PLK1 and the effects of their dysregulation in esophageal squamous cell carcinoma (ESCC) cells. METHODS: We used immunoblot, quantitative reverse-transcription polymerase chain reaction, immunochemistry, chromatin immunoprecipitation, mobility shift, and reporter assays to investigate the relationship between Stat3 and PLK1. We used colony formation, fluorescence-activated cell sorting, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling, and xenograft tumor assays to determine the effects of increased activation of Stat3 and PLK1 in proliferation and survival of ESCC cells. RESULTS: Stat3 directly activated transcription of PLK1 in esophageal cancer cells and mouse embryonic fibroblast cell NIH3T3. PLK1 then potentiated the expression of Stat3; ß-catenin was involved in PLK1-dependent transcriptional activation of Stat3. This mutual regulation between Stat3 and PLK1 was required for proliferation of esophageal cancer cells and resistance to apoptosis in culture and as tumor xenografts in mice. Furthermore, phosphorylation of Stat3 and overexpression of PLK1 were correlated in a subset of ESCC. CONCLUSIONS: Stat3 and PLK1 control each other's transcription in a positive feedback loop that contributes to the development of ESCC. Increased activity of Stat3 and overexpression of PLK1 promote survival and proliferation of ESCC cells in culture and in mice.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Neoplasias Esofágicas/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Antineoplásicos/farmacologia , Western Blotting , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Separação Celular/métodos , Sobrevivência Celular , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Ativação Enzimática , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Retroalimentação Fisiológica , Feminino , Citometria de Fluxo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Nus , Células NIH 3T3 , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Pteridinas/farmacologia , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/genética , Transdução de Sinais , Fatores de Tempo , Ativação Transcricional , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/metabolismo , Quinase 1 Polo-Like
5.
Clin Sci (Lond) ; 124(3): 203-14, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22963397

RESUMO

There have been multiple lines of evidence suggesting that autophagy selectively targets signalling proteins and regulates cancer cell signalling in addition to bulk clearance of long-lived proteins and organelles. Protein degradation through autophagy requires receptor protein LC3B to sequester the substrates into the autophagosome. In the present study, we screened LC3B (light-chain 3B)-binding partners and identified autophagic substrates in cancer cells. With lung cancer NCI-H1975 and oesophageal cancer KYSE30 cell lines as models, we found that VPRBP (viral protein R-binding protein) was a novel LC3B-binding protein through GST (glutathione transferase)-LC3B pull-down combined with LC-MS/MS (liquid chromatography-tandem MS) methods. Co-immunoprecipitation assay showed that VPRBP-LC3/p62 were in the same protein complex as the two cell lines. Induction of autophagy led to a down-regulation of VPRPB, which could be rescued by the inhibition of autophagy degradation by BFA1 (bafilomycin A1) and by the disruption of autophagy through ATG5-knockdown. We also found that induction of autophagy promotes VPRBP-LC3/p62 interaction. Immunohistochemical examination of human NSCLC (non-small cell lung cancer) tissues showed that VPRBP was positively correlated with p62 and negatively correlated with LC3B. Moreover, p62 and VPRBP were associated with poor prognosis in lung ADC (adenocarcinoma) (p62, P=0.019; VPRBP, P=0.005). Patients with low expression of both p62 and VPRBP showed the best prognosis.


Assuntos
Autofagia , Proteínas de Transporte/metabolismo , Proliferação de Células , Neoplasias Pulmonares/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibióticos Antineoplásicos/farmacologia , Western Blotting , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Feminino , Humanos , Peróxido de Hidrogênio/farmacologia , Imuno-Histoquímica , Imunoprecipitação , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade , Oxidantes/farmacologia , Prognóstico , Ligação Proteica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases , Interferência de RNA , Proteína Sequestossoma-1 , Sirolimo/farmacologia , Ubiquitina-Proteína Ligases
6.
Zhonghua Yi Xue Za Zhi ; 93(18): 1426-31, 2013 May 14.
Artigo em Chinês | MEDLINE | ID: mdl-24025511

RESUMO

OBJECTIVE: To explore the role and mechanism of microRNA185 (miR-185) on proliferation, migration and invasion of esophageal squamous cell carcinoma (ESCC). METHODS: Samples were obtained from 23 ESCC patients undergoing surgery whose were confirmed by pathological diagnosis of esophageal carcinoma from 2002 to 2012,at Department of Thoracic Surgery,Cancer Institute and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College. Real-time PCR was used to measure the expression of miR-185. The xCELLigence RTCA MP system and Transwell assay were performed to detect the effect of miR-185 on proliferation, migration and invasion of ESCC respectively. After transfecting of miR-185 mimic into KYSE150, the expression of Six1's downstream gene cyclin A1 was evaluated by real-time PCR. After transfection of miR-185 inhibitor into KYSE30, the expression of E-cadherin, a downstream protein of Six1, was observed under confocal microscope. RESULTS: The expression level of miR-185 was down-regulated in ESCC compared with adjacent normal tissue (0.006 vs 0.039,P = 0.016). After transfection of miR-185 mimic, miR-185 significantly inhibited proliferation, migration and invasion of ESCC.Transwell assay showed, in comparison with the control group, the number of KYSE150 metastatic and invasive cells was respectively decreased(146 ± 15 vs 64 ± 11, 110 ± 12 vs 67 ± 5, both P < 0.05). And the expression level of cyclin A1 decreased. After transfection of miR-185 inhibitor,the expression level of E-cadherin decreased. CONCLUSION: miR-185 may inhibit proliferation, migration and invasion of ESCC through its target gene Six1.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , MicroRNAs/genética , Antígenos CD , Caderinas/metabolismo , Movimento Celular , Proliferação de Células , Ciclina A1/metabolismo , Carcinoma de Células Escamosas do Esôfago , Proteínas de Homeodomínio/metabolismo , Humanos , Invasividade Neoplásica/genética , Transfecção , Células Tumorais Cultivadas
7.
BMC Cancer ; 12: 367, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22920630

RESUMO

BACKGROUND: Chromosomal and genomic aberrations are common features of human cancers. However, chromosomal numerical and structural aberrations, breakpoints and disrupted genes have yet to be identified in esophageal squamous cell carcinoma (ESCC). METHODS: Using multiplex-fluorescence in situ hybridization (M-FISH) and oligo array-based comparative hybridization (array-CGH), we identified aberrations and breakpoints in six ESCC cell lines. Furthermore, we detected recurrent breakpoints in primary tumors by dual-color FISH. RESULTS: M-FISH and array-CGH results revealed complex numerical and structural aberrations. Frequent gains occurred at 3q26.33-qter, 5p14.1-p11, 7pter-p12.3, 8q24.13-q24.21, 9q31.1-qter, 11p13-p11, 11q11-q13.4, 17q23.3-qter, 18pter-p11, 19 and 20q13.32-qter. Losses were frequent at 18q21.1-qter. Breakpoints that clustered within 1 or 2 Mb were identified, including 9p21.3, 11q13.3-q13.4, 15q25.3 and 3q28. By dual-color FISH, we observed that several recurrent breakpoint regions in cell lines were also present in ESCC tumors. In particular, breakpoints clustered at 11q13.3-q13.4 were identified in 43.3% (58/134) of ESCC tumors. Both 11q13.3-q13.4 splitting and amplification were significantly correlated with lymph node metastasis (LNM) (P = 0.004 and 0.022) and advanced stages (P = 0.004 and 0.039). Multivariate logistic regression analysis revealed that only 11q13.3-q13.4 splitting was an independent predictor for LNM (P = 0.026). CONCLUSIONS: The combination of M-FISH and array-CGH helps produce more accurate karyotypes. Our data provide significant, detailed information for appropriate uses of these ESCC cell lines for cytogenetic and molecular biological studies. The aberrations and breakpoints detected in both the cell lines and primary tumors will contribute to identify affected genes involved in the development and progression of ESCC.


Assuntos
Carcinoma de Células Escamosas/genética , Aberrações Cromossômicas , Neoplasias Esofágicas/genética , Rearranjo Gênico , Idoso , Carcinoma de Células Escamosas/patologia , Pontos de Quebra do Cromossomo , Cromossomos Humanos , Hibridização Genômica Comparativa , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago , Feminino , Amplificação de Genes , Deleção de Genes , Humanos , Hibridização in Situ Fluorescente , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Cariotipagem Espectral , Células Tumorais Cultivadas
8.
Acta Pharmacol Sin ; 33(10): 1301-10, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22983390

RESUMO

AIM: Filamin binding LIM protein 1, also known as migfilin, is a skeleton organization protein that binds to mitogen-inducible gene 2 at cell-extracellular matrix adhesions. The aim of this study was to investigate the role of migfilin in cisplatin-induced apoptosis in human glioma cells, to determine the functional domains of migfilin, and to elucidate the molecular mechanisms underlying the regulation of cisplatin-related chemosensitivity. METHODS: The human glioma cell lines Hs683, H4, and U-87 MG were transfected with pEGFP-C2-migfilin to elevate the expression level of migfilin. RNA interference was used to reduce the expression of migfilin. To determine the functional domains of migfilin, U-87 MG cells were transfected with plasmids of migfilin deletion mutants. After treatment with cisplatin (40 µmol/L) for 24 h, the cell viability was assessed using the MTS assay, and the cell apoptotic was examined using the DAPI staining assay and TUNEL analysis. Expression levels of apoptosis-related proteins were detected by Western blot analysis. RESULTS: Overexpression of migfilin significantly enhanced cisplatin-induced apoptosis in Hs683, H4, and U-87 MG cells, whereas downregulation of migfilin expression inhibited the chemosensitivity of these cell lines. The N-terminal region of migfilin alone was able to enhance the cisplatin-induced apoptosis. However, despite the existence of the N-terminal region, mutants of migfilin with any one of three LIM domains deleted led to a function loss. Furthermore, apoptotic proteins (PARP and caspase-3) and the anti-apoptotic protein Bcl-xL were modulated by the expression level of migfilin in combination with cisplatin. CONCLUSION: The LIM1-3 domains of migfilin play a key role in sensitizing glioma cells to cisplatin-induced apoptosis through regulation of apoptosis-related proteins.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Cisplatino/farmacologia , Proteínas do Citoesqueleto/metabolismo , Glioma , Apoptose/genética , Western Blotting , Caspase 3/genética , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/genética , Regulação para Baixo , Deleção de Genes , Glioma/metabolismo , Glioma/patologia , Proteínas de Fluorescência Verde/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Plasmídeos , RNA Interferente Pequeno/genética , Transfecção , Regulação para Cima , Proteína bcl-X/genética
9.
Genes Chromosomes Cancer ; 50(7): 518-26, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21484929

RESUMO

Risk assessment of esophageal squamous cell carcinoma (ESCC) is currently based on clinicopathological parameters. To identify genomic markers that can predict overall survival in ESCC, we performed array comparative genomic hybridization (array CGH) on a screening set of 35 tumor samples from ESCC patients. Prognosis association of the genes selected on the basis of the array CGH results was further validated by real-time PCR in two independent sample sets (n = 151 and 84). Genomic analysis revealed seven high-level amplifications and two homozygous deletions. Gain of 11q13.2 and loss of 7q34 and 18q21.1-q23 were associated with poor outcome. Gain of 11q13.2 was an independent prognostic factor and was selected for further validation. In both validation sets of samples, copy number increase of CPT1A in 11q13.2 was correlated with short overall survival (P = 0.015, n = 151 and P = 0.044, n = 84). Multivariate analysis confirmed that CPT1A gain provided prognostic information in ESCC (HR, 1.643; 95% CI: 1.076-2.509; P = 0.022; HR, 2.488; 95% CI: 1.235-5.013; P = 0.011). Immunohistochemistry showed significant correlation between strong expression of CPT1A protein and poor outcome of ESCC patients (P = 0.018, n = 73). Our data suggest that gain of CPT1A may be a candidate prognostic factor.


Assuntos
Carcinoma de Células Escamosas/genética , Carnitina O-Palmitoiltransferase/genética , Neoplasias Esofágicas/genética , Genoma Humano , Idoso , Biomarcadores Tumorais , Carcinoma de Células Escamosas/patologia , Sobrevivência Celular/genética , Hibridização Genômica Comparativa , Neoplasias Esofágicas/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade
10.
Zhonghua Zhong Liu Za Zhi ; 34(12): 905-9, 2012 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-23336376

RESUMO

OBJECTIVE: To investigate the expression of BRCA1 in esophageal squamous cell carcinoma (ESCC) tissues and evaluate its correlation with clinicopathological features as well as the prognosis of ESCC patients. METHODS: The expression of BRCA1 was detected by immunohistochemistry (IHC) in 201 specimens of T3 stage ESCC tissues and corresponding adjacent normal tissues using tissue microarray. The correlation between BRCA1 expression and clinicopathological features of ESCC was determined by chi-square analysis. The cumulative survival rate was analyzed by Kaplan-Meier method. RESULTS: The positive rate of BRCA1 expression in ESCC tissues was significantly higher than that in adjacent normal tissues [88.6% (178/201) vs. 36.8% (74/201), P < 0.001]. There was a significant correlation between the expression of BRCA1 and lymph node metastasis. In the tumors with positive lymph nodes, strong positive expression of BRCA1 was found in 45.0% (49/109), while only 19.6% (18/92) in tumors without lymph node metastasis, showing a significant difference (P < 0.001). A close relationship was also found between the expression of BRCA1 and gross typing of tumors (P < 0.05). The expression of BRCA1 was not significantly correlated with gender, age, tumor location, differentiation, and tumor thrombus (P > 0.05). The results of Kaplan-Meier analysis indicated that ESCC patients with a higher positive rate of BRCA1 expression have a poorer prognosis (P < 0.05). CONCLUSIONS: The expression of BRCA1 is related to the occurrence and development of esophageal carcinoma. BRCA1 protein may serve as a new potential biomarker in estimating the biological behavior of ESCC.


Assuntos
Proteína BRCA1/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/cirurgia , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/cirurgia , Carcinoma de Células Escamosas do Esôfago , Feminino , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Taxa de Sobrevida
11.
Mol Cancer Res ; 7(1): 12-22, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19147533

RESUMO

The gene of SKP2, located on chromosome 5p13, plays a critical role in cell cycle progression, especially at the G(1)-S transition, putatively through its control of several cell cycle regulator proteins including p27(kip1), p21(cip1), p57(kip2), p130, cyclin E, and c-Myc. Previous studies in this laboratory revealed that gain of chromosome 5p was often seen in esophageal squamous cell carcinoma (ESCC). In the present study, we examined the amplification status and expression level of SKP2 in ESCC and investigated its clinicopathologic significance. Amplification and elevated expression of SKP2 correlated significantly with tumor stage and positive lymph node metastasis (P < 0.05). The SKP2 protein expression level as determined by immunohistochemical staining showed a significant inverse correlation with p27 protein. In vivo assay showed that inhibition of SKP2 expression also decreased tumor growth and lung metastasis of ESCC cells. At the molecular level, knockdown of SKP2 by RNA interference inhibited cell migration and invasion ability. Knockdown of SKP2 expression sensitized cancer cells to anoikis, and a wobble mutant of SKP2 that is resistant to SKP2 small interfering RNA can rescue this effect. Expression level of pAkt decreased after SKP2 knockdown. Treatment of cells with phosphoinositidyl 3-kinase inhibitor (LY294002) and constitutively activator (insulin-like growth factor I) had significant effects on the anoikis of SKP2 RNA interference cells. These results show for the first time that SKP2 is amplified and overexpressed in ESCC. Elevated expression of SKP2 protected cancer cells from anoikis, and this effect was mediated, at least in part, by the phosphoinositidyl 3-kinase-Akt pathway.


Assuntos
Anoikis/genética , Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Amplificação de Genes , Metástase Linfática/genética , Metástase Neoplásica/genética , Proteínas Quinases Associadas a Fase S/genética , Sequência de Bases , Carcinoma de Células Escamosas/patologia , Movimento Celular/genética , Primers do DNA , Neoplasias Esofágicas/patologia , Humanos , Dados de Sequência Molecular , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Plasmídeos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Interferência de RNA , RNA Neoplásico/genética
12.
Zhonghua Zhong Liu Za Zhi ; 32(10): 748-51, 2010 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-21163064

RESUMO

OBJECTIVE: China has the highest incidence and mortality of esophageal carcinoma in the world, and the esophageal squamous cell carcinoma (ESCC) is the major type. In this study, the authors investigated the expression of Aurora-A in stage T3 esophageal squamous cell carcinomas (ESCC) with positive and negative lymph node metastasis, and analyzed its relationship with prognosis of ESCC patients. METHODS: ESCC tissue arrays including 212 specimens had been constructed. The expression of Aurora-A in both ESCC tissues and adjacent normal tissues was determined by immunohistochemical staining. The correlation between Aurora-A protein levels and lymph node status in ESCC and survival rate were statistically analyzed. RESULTS: The positive expression of Aurora-A was 74.07% (140/189) in tumor tissues and 18.52% (35/189) in adjacent normal tissues, showing a significant difference between them (χ(2) = 105.162, P < 0.05). In tumors with positive lymph nodes, strong positive expression of Aurora-A was found in 42.99% (46/107), while only 7.37% (7/95) in tumors with negative lymph nodes, with a statistically significant difference (χ(2) = 36.132, P < 0.05). The cumulative survival rate of the patients with strong Aurora-A-positive tumors was significantly lower than that in patients with Aurora-A-negative tumors (P = 0.0042, P < 0.05). CONCLUSION: The positive expression of Aurora-A in ESCC tissues is much higher than that in adjacent normal tissues. The expression of Aurora-A is higher in lymph node-positive tumors than in the lymph node-negative ones. There is a significantly longer cumulative survival rate in patients with negative Aurora-A expression than that in patients with strong positive Aurora-A expression.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Aurora Quinases , Feminino , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Taxa de Sobrevida
13.
Zhonghua Yi Xue Za Zhi ; 90(44): 3156-9, 2010 Nov 30.
Artigo em Chinês | MEDLINE | ID: mdl-21211351

RESUMO

OBJECTIVE: To study the expression of non-coding RNA HULC in different tumor cell lines and its effects on the expression of neighboring genes. METHODS: The expression levels of HULC were detected in different tumor cell lines. And the neighboring genes of HULC were searched by bioinformatics. Then its effects on the neighboring genes were examined by real-time polymerase chain reaction (PCR). RESULTS: The expression levels of HULC RNA were detected in the 293T, EC9706, KYSE150, HCT116, SMMC-7721, HepG2, H446 and H1299 cell lines. Non-coding RNA HULC was specifically highly up-regulated in hepatocarcinoma cell line HepG2. Little coding genes existed around HULC gene (< 1000 bp) and the nearest coding gene was SLC35B3 at 200 000 bp away from the location of HULC gene. The SMMC-7721 cell line had a low endogenous expression of HULC gene. In comparison with SMMC-7721 cell line, the expression of SLC35B3 gene in HepG2 cell line was higher [(0.477 ± 0.040)% vs (0.129 ± 0.004)%, P < 0.01]. The exogenous expression of HULC had little effects on SLC35B3 gene. But knocking down the expression of HULC gene by siRNA attenuated the expression of SLC35B3 [siA (0.283 ± 0.007)%, siB (0.387 ± 0.015)%, control (0.477 ± 0.042)%]. CONCLUSION: The specifically up-regulated non-coding gene HULC in HepG2 cell line has some effects on the expression of its neighboring gene SLC35B3.


Assuntos
Proteínas de Transporte de Ânions/genética , Carcinoma Hepatocelular/genética , RNA não Traduzido/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos
14.
Int J Cancer ; 124(3): 578-88, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19004025

RESUMO

PLK1 is essential for the maintenance of genomic stability during mitosis. In our study, we found that overexpression of PLK1 was an independent prognostic factor (RR=4.253, p=0.020) and significantly correlated with survivin, an antiapoptotic protein, in esophageal squamous cell carcinoma (ESCC). Reverse transcription-polymerase chain reaction and fluorescence in situ hybridization (FISH) revealed upregulation of PLK1 mRNA and amplification of PLK1 gene, respectively. Depletion of PLK1 activated the intrinsic apoptotic pathway, which was substantiated by loss of mitochondrial membrane potential, reduction of Mcl-1 and Bcl-2 as well as activation of caspase-9. Coimmunoprecipitation and confocal microscopy displayed that PLK1 was associated with survivin and PLK1 depletion led to downregulation of survivin. Cotransfection of survivin constructs could partially reverse PLK1-depletion-induced apoptosis. These data suggest that PLK1 might be a useful prognostic marker and a potential therapeutic target for ESCC. Survivin is probably involved in antiapoptotic function of PLK1.


Assuntos
Apoptose/fisiologia , Carcinoma de Células Escamosas/metabolismo , Proteínas de Ciclo Celular/biossíntese , Neoplasias Esofágicas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Western Blotting , Carcinoma de Células Escamosas/mortalidade , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/mortalidade , Neoplasias Esofágicas/patologia , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Imunoprecipitação , Hibridização in Situ Fluorescente , Proteínas Inibidoras de Apoptose , Estimativa de Kaplan-Meier , Masculino , Potencial da Membrana Mitocondrial/fisiologia , Microscopia Confocal , Pessoa de Meia-Idade , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Survivina , Análise Serial de Tecidos , Quinase 1 Polo-Like
15.
Genes Chromosomes Cancer ; 47(2): 127-36, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17990328

RESUMO

DNA amplification is one of the mechanisms to activate genes that are implicated in neoplastic transformation and gain of chromosome band 3q26 is a common event in squamous cell carcinomas. The aim of the present work was to identify the specific target gene from four candidates (MDS1, PRKCI, ECT2, and PIK3CA) located on 3q26 amplification in esophageal squamous cell carcinomas (ESCCs). To assess the prevalence of copy number gains of putative genes, fluorescence in situ hybridization (FISH) was applied on 108 ESCCs and 9 ESCC cell lines. Our data showed that MDS1 and PRKCI were more frequently gained. Positive correlation was found only for PRKCI between amplification and tumor size (P = 0.043), lymph node metastasis (P = 0.015) and clinical stage (P = 0.002). PRKCI gene amplification was highly correlated with protein overexpression (P = 0.009), suggesting that gene amplification is one important mechanism involved in PRKCI overexpression. To investigate further the role of PRKCI alteration in esophageal tumors, a tissue microarray containing samples from 180 ESCCs was used for immunohistochemistry analysis. Statistical analysis revealed that PRKCI overexpression was correlated with lymph node metastasis (P = 0.002) and higher stage (P = 0.004). Performing multivariate logistic regression analysis, a significant association between PRKCI overexpression and presence of lymph node metastasis was found, which was independent of T-stage of the primary tumors (P = 0.004). Our results indicate that PRKCI is an attractive target in the 3q26 amplicon and that it may serve as a molecular marker for metastasis and occult advanced tumor stages in ESCC.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/enzimologia , Cromossomos Humanos Par 3/genética , Neoplasias Esofágicas/enzimologia , Amplificação de Genes , Isoenzimas/genética , Proteína Quinase C/genética , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Progressão da Doença , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Feminino , Dosagem de Genes , Humanos , Isoenzimas/biossíntese , Isoenzimas/metabolismo , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Proteína Quinase C/biossíntese , Proteína Quinase C/metabolismo
16.
Cancer Res ; 79(13): 3281-3293, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31053633

RESUMO

Metabolic activities are often accompanied by cell-cycle progression, yet known connections between these two processes remain limited. Here, we identified the isocitrate dehydrogenase 3ß (IDH3ß) as a novel substrate of anaphase-promoting complex/cyclosome (APC/C)-CDH1 and an important regulator of the cell cycle. In esophageal squamous cell carcinoma (ESCC), IDH3ß was posttranslationally upregulated in late G1 phase, and overexpression of IDH3ß accelerated G1-S transition, contributing to the promotion of cell proliferation in vitro and in vivo. α-Ketoglutarate (α-KG), a crucial metabolite in tricarboxylic acid (TCA) cycle, was dependent on IDH3ß level and partially accounted for IDH3ß-mediated cell growth. IDH3ß expression increased PFKFB3 protein levels and enhanced glucose uptake, and high expression of IDH3ß correlated with poor survival in patients with ESCC, suggesting a potential application of IDH3ß in prognosis. Overall, our results highlight a new molecular connection between cell-cycle regulation and the TCA cycle in ESCC. SIGNIFICANCE: These findings show that IDH3ß is an APC/C-CDH1 substrate and is expressed in a cell-cycle-dependent manner, highlighting novel molecular cross-talk between the TCA cycle and cell cycle in cancer cells.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/13/3281/F1.large.jpg.


Assuntos
Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Antígenos CD/metabolismo , Caderinas/metabolismo , Ciclo Celular , Proliferação de Células , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/secundário , Isocitrato Desidrogenase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase/genética , Animais , Antígenos CD/genética , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Caderinas/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Isocitrato Desidrogenase/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Metástase Neoplásica , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Cancer Lett ; 459: 145-155, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31173854

RESUMO

Radiotherapy is one of the standard therapies for esophageal squamous cell carcinoma (ESCC), but the efficacy is far from desirable. Large scale genome sequencing reveals PI3Kα is frequently hyper-activated in ESCC. We found that ESCC cells harboring alterations in PI3K pathway were more resistant to radiation and combination of a clinical PI3Kα-selective inhibitor CYH33 and radiation synergistically inhibited cell proliferation in 14 ESCC cell lines. Radiation induced phosphorylation of FOXO1 and Akt, which sensitized ESCC cells to PI3Kα inhibitors. Both S1PR3 and DNA-PK contributed to radiation-induced Akt phosphorylation, which were revealed to be collectively dependent on PI3Kα. By contrast, constitutively active Akt abrogated the synergism between PI3Kα inhibitors and radiation. PI3Kα inhibition enhanced radiation-induced DNA damage, G2/M arrest and apoptosis. Combination of CYH33 and radiation significantly inhibited the growth of xenografts derived from ESCC patients, which was accompanied with abrogation of radiation-induced phosphorylation of Akt and filtration of M2-like macrophages. Taken together, combination of CYH33 and radiation possesses synergism in ESCC, which provides promising rationale to test this combinatorial regimen in ESCC patients.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/radioterapia , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/radioterapia , Morfolinas/farmacologia , Piperazinas/farmacologia , Pirróis/farmacologia , Radiossensibilizantes/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Técnicas de Cocultura , Dano ao DNA , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/metabolismo , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Proteína Forkhead Box O1/metabolismo , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação/efeitos da radiação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Distribuição Aleatória , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Células THP-1 , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cancer Res ; 79(5): 941-953, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30530815

RESUMO

Various cancer stem cell (CSC) biomarkers have been identified for hepatocellular carcinoma (HCC), but little is known about the implications of heterogeneity and shared molecular networks within the CSC population. Through miRNA profile analysis in an HCC cohort (n = 241) for five groups of CSC+ HCC tissues, i.e., EpCAM+, CD90+, CD133+, CD44+, and CD24+ HCC, we identified a 14-miRNA signature commonly altered among these five groups of CSC+ HCC. miR-192-5p, the top-ranked CSC miRNA, was liver-abundant and -specific and markedly downregulated in all five groups of CSC+ HCC from two independent cohorts (n = 613). Suppressing miR-192-5p in HCC cells significantly increased multiple CSC populations and CSC-related features through targeting PABPC4. Both TP53 mutation and hypermethylation of the mir-192 promoter impeded transcriptional activation of miR-192-5p in HCC cell lines and primary CSC+ HCC. This study reveals the circuit from hypermethylation of the mir-192 promoter through the increase in PABPC4 as a shared genetic regulatory pathway in various groups of primary CSC+ HCC. This circuit may be the driver that steers liver cells toward hepatic CSC cells, leading to hepatic carcinogenesis. SIGNIFICANCE: miR-192-5p and its regulatory pathway is significantly abolished in multiple groups of HCC expressing high levels of CSC markers, which may represent a key event for hepatic carcinogenesis.


Assuntos
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Células-Tronco Neoplásicas/patologia , Animais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Estudos de Coortes , Metilação de DNA , Regulação para Baixo , Redes Reguladoras de Genes , Células HEK293 , Células Hep G2 , Xenoenxertos , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/biossíntese , Células-Tronco Neoplásicas/metabolismo , Proteínas de Ligação a Poli(A)/genética , Proteínas de Ligação a Poli(A)/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais/genética
19.
Int J Cancer ; 122(2): 260-6, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17935133

RESUMO

Esophagin/SPRR3 is one of the cornified-envelope structural precursor proteins, which is expressed during epithelia cell differentiation. In 1996, another research group discovered, and our own laboratory subsequently confirmed, frequent and dramatic decreased Esophagin/SPRR3 expression in esophageal squamous cell carcinoma (ESCC). However, the role of Esophagin/SPRR3 in tumorigenesis of esophageal epithelium remains undetermined. In this study, we demonstrate that expression of Esophagin/SPRR3 is frequently downregulated in ESCC. In contrast, no correlations between downregulation of Esophagin/SPRR3 expression and clinicopathologic characteristics were observed. Diminished Esophagin/SPRR3 expression was present in dysplastic epithelia, suggesting that Esophagin/SPRR3 alteration could represent an early event in squamous carcinogenesis of the esophagus. Exogenous expression of Esophagin/SPRR3 significantly suppressed the ability of ESCC cells to form colonies in plastic and soft agar, as well as tumor formation in vivo. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end label assay and immunofluorescence analysis of the active form of Caspase 3 indicated that dysregulated apoptosis might contribute to reduced tumorigenicity. In particular, upregulation of CDK11p46 protein was observed in ESCC cells expressing Esophagin/SPRR3, but not in control cells, indicating that Esophagin/SPRR3-induced apoptosis may be due, at least in part, to increased expression of CDK11p46 protein. These findings suggest that Esophagin/SPRR3 may play a role in the maintenance of normal esophageal epithelial homeostasis, and that aberrant expression of Esophagin/SPRR3 may contribute to the tumorigenesis of ESCC.


Assuntos
Apoptose , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Regulação Neoplásica da Expressão Gênica , Animais , Diferenciação Celular , Transformação Celular Neoplásica , Células Epiteliais/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia de Fluorescência , Fenótipo , Conformação Proteica
20.
J Mol Med (Berl) ; 85(8): 863-75, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17318615

RESUMO

Esophageal squamous cell carcinoma (ESCC) is one of the leading causes of cancer death in China. In the present study, proteins in tumors and adjacent normal esophageal tissues from 41 patients with ESCC were extracted, and two-dimensional electrophoresis (2-DE) was performed using the pH 3-10 and 4-7 immobilized pH gradient strips. The protein spots expressed differentially between tumors and normal tissues were identified by matrix-assisted laser desorption/ionization and liquid chromatography electrospray/ionization ion trap mass spectrometry. A total of 22 proteins differentially expressed between ESCC and normal esophageal tissues were identified, in which 17 proteins were upregulated and 5 downregulated in tumors. Biological functions of these proteins are related to cell signal transduction, cell proliferation, cell motility, glycolysis, regulation of transcription, oxidative stress processes, and protein folding. Some of the proteins obtained were confirmed by Western blotting and immunohistochemical staining. We showed that high expression of calreticulin and 78-kDa glucose-regulated protein (GRP78) were correlated with poor prognosis by Kaplan-Meier analysis and log rank analysis. Zinc finger protein 410, annexin V, similar to the ubiquitin-conjugating enzyme E2 variant 1 isoform c, mutant hemoglobin beta chain, TPM4-ALK fusion oncoprotein type 2, similar to heat shock congnate 71-kDa protein, GRP78, and pyruvate kinase M2 (M2-PK) were for the first time observed to be dysregulated in human ESCC tissues. The proteins here identified will contribute to the understanding of the tumorigenesis and progression of Chinese ESCC and may potentially provide useful markers for diagnosis or targets for therapeutic intervention and drug development.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Proteoma/análise , Proteômica/métodos , Povo Asiático , Western Blotting , Carcinoma de Células Escamosas/etnologia , China , Cromatografia Líquida , Eletroforese em Gel Bidimensional , Chaperona BiP do Retículo Endoplasmático , Neoplasias Esofágicas/etnologia , Humanos , Imuno-Histoquímica , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA