Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Cell ; 9(1): 1-2, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16413463

RESUMO

The status and interrelationship of p53 family members are critical elements in tumor progression. An intriguing paper in this issue of Cancer Cell (Rocco et al., 2006) reveals a new twist in the interactions between p63 and p73 following DNA damage, underscoring a role for p73 in the proapoptotic regulation of Puma, Noxa, and Bcl-2 in head and neck squamous cell carcinomas (HNSCC). These data define a pathway in which deltaNp63alpha promotes survival in squamous epithelial malignancy by repressing a p73-dependent proapoptotic transcriptional program, suggesting that p63 levels and p73 status may be key determinants of tumor response in patients with HNSCC.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Carcinoma de Células Escamosas/patologia , Genes Supressores de Tumor , Genes bcl-2 , Genes p53 , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Fatores de Transcrição , Proteína Tumoral p73 , Proteínas Supressoras de Tumor
2.
BMC Cancer ; 13: 196, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23594791

RESUMO

BACKGROUND: The transition from normal epithelium to adenoma and, to invasive carcinoma in the human colon is associated with acquired molecular events taking 5-10 years for malignant transformation. We discovered CCAT1, a non-coding RNA over-expressed in colon cancer (CC), but not in normal tissues, thereby making it a potential disease-specific biomarker. We aimed to define and validate CCAT1 as a CC-specific biomarker, and to study CCAT1 expression across the adenoma-carcinoma sequence of CC tumorigenesis. METHODS: Tissue samples were obtained from patients undergoing resection for colonic adenoma(s) or carcinoma. Normal colonic tissue (n = 10), adenomatous polyps (n = 18), primary tumor tissue (n = 22), normal mucosa adjacent to primary tumor (n = 16), and lymph node(s) (n = 20), liver (n = 8), and peritoneal metastases (n = 19) were studied. RNA was extracted from all tissue samples, and CCAT1 expression was analyzed using quantitative real time-PCR (qRT-PCR) with confirmatory in-situ hybridization (ISH). RESULTS: Borderline expression of CCAT1 was identified in normal tissue obtained from patients with benign conditions [mean Relative Quantity (RQ) = 5.9]. Significant relative CCAT1 up-regulation was observed in adenomatous polyps (RQ = 178.6 ± 157.0; p = 0.0012); primary tumor tissue (RQ = 64.9 ± 56.9; p = 0.0048); normal mucosa adjacent to primary tumor (RQ = 17.7 ± 21.5; p = 0.09); lymph node, liver and peritoneal metastases (RQ = 11,414.5 ± 12,672.9; 119.2 ± 138.9; 816.3 ± 2,736.1; p = 0.0001, respectively). qRT-PCR results were confirmed by ISH, demonstrating significant correlation between CCAT1 up-regulation measured using these two methods. CONCLUSION: CCAT1 is up-regulated across the colon adenoma-carcinoma sequence. This up-regulation is evident in pre-malignant conditions and through all disease stages, including advanced metastatic disease suggesting a role in both tumorigenesis and the metastatic process.


Assuntos
Adenocarcinoma/genética , Adenoma/genética , Colo/metabolismo , Neoplasias do Colo/genética , Neoplasias Hepáticas/genética , Neoplasias Peritoneais/genética , RNA Longo não Codificante/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/secundário , Adenoma/metabolismo , Adenoma/patologia , Adulto , Idoso , Western Blotting , Estudos de Casos e Controles , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Seguimentos , Humanos , Técnicas Imunoenzimáticas , Hibridização In Situ , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Peritoneais/metabolismo , Neoplasias Peritoneais/secundário , Prognóstico , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
3.
Apoptosis ; 17(9): 989-97, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22460505

RESUMO

Recently, thioridazine (10-[2-(1-methyl-2-piperidyl) ethyl]-2-methylthiophenothiazine), a well-known anti-psychotic agent was found to have anti-cancer activity in cancer cells. However, the molecular mechanism of the agent in cellular signal pathways has not been well defined. Thioridazine significantly increased early- and late-stage apoptotic fraction in cervical and endometrial cancer cells, suggesting that suppression of cell growth by thioridazine was due to the induction of apoptosis. Cell cycle analysis indicated thioridazine induced the down-regulation of cyclin D1, cyclin A and CDK4, and the induction of p21 and p27, a cyclin-dependent kinase inhibitor. Additionally, we compared the influence of thioridazine with cisplatin used as a control, and similar patterns between the two drugs were observed in cervical and endometrial cancer cell lines. Furthermore, as expected, thioridazine successfully inhibited phosphorylation of Akt, phosphorylation of 4E-BP1 and phosphorylation of p70S6K, which is one of the best characterized targets of the mTOR complex cascade. These results suggest that thioridazine effectively suppresses tumor growth activity by targeting the PI3K/Akt/mTOR/p70S6K signaling pathway.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias do Endométrio/metabolismo , Tioridazina/farmacologia , Neoplasias do Colo do Útero/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Caspase 3/metabolismo , Proteínas de Ciclo Celular , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular , Ciclina A/biossíntese , Ciclina D1/biossíntese , Quinase 4 Dependente de Ciclina/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Regulação para Baixo , Feminino , Células HeLa , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
4.
Cancer Cell ; 1(4): 369-79, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12086851

RESUMO

The P53 homolog p63 encodes multiple proteins with transactivating, apoptosis-inducing, and oncogenic activities. We showed that p63 is amplified and that DeltaNp63 isotypes are overexpressed in squamous cell carcinoma (SCC) and enhance oncogenic growth in vitro and in vivo. Moreover, p53 associated with DeltaNp63alpha and mediated its degradation. Here, we report that DeltaNp63 associates with the B56alpha regulatory subunit of protein phosphatase 2A (PP2A) and glycogen synthase kinase 3beta (GSK3beta), leading to a dramatic inhibition of PP2A-mediated GSK3beta reactivation. The inhibitory effect of DeltaNp63 on GSK3beta mediates a decrease in phosphorylation levels of beta-catenin, which induces intranuclear accumulation of beta-catenin and activates beta-catenin-dependent transcription. Our results suggest that DeltaNp63 isotypes act as positive regulators of the beta-catenin signaling pathway, providing a basis for their oncogenic properties.


Assuntos
Proteínas Sanguíneas/metabolismo , Neoplasias Ósseas/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Carcinoma de Células Escamosas/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Fosfoproteínas , Transativadores , Proteínas Sanguíneas/genética , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Divisão Celular/fisiologia , Núcleo Celular/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/farmacologia , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/farmacologia , Genes Supressores de Tumor , Quinase 3 da Glicogênio Sintase , Quinases da Glicogênio Sintase , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Luciferases/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/farmacologia , Fosforilação , Plasmídeos , Testes de Precipitina , Proteína Fosfatase 2 , Transdução de Sinais , Fatores de Transcrição , Proteínas Supressoras de Tumor , Técnicas do Sistema de Duplo-Híbrido , beta Catenina
5.
Biochem Biophys Res Commun ; 404(4): 958-62, 2011 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-21185263

RESUMO

Aberrant signaling of the Ras-Raf-MEK-ERK (MAP kinase) pathway driven by the mutant kinase BRAF(V600E), as a result of the BRAF(T1799A) mutation, plays a fundamental role in thyroid tumorigenesis. This study investigated the therapeutic potential of a BRAF(V600E)-selective inhibitor, PLX4032 (RG7204), for thyroid cancer by examining its effects on the MAP kinase signaling and proliferation of 10 thyroid cancer cell lines with wild-type BRAF or BRAF(T1799A) mutation. We found that PLX4032 could effectively inhibit the MAP kinase signaling, as reflected by the suppression of ERK phosphorylation, in cells harboring the BRAF(T1799A) mutation. PLX4032 also showed a potent and BRAF mutation-selective inhibition of cell proliferation in a concentration-dependent manner. PLX4032 displayed low IC(50) values (0.115-1.156µM) in BRAF(V600E) mutant cells, in contrast with wild-type BRAF cells that showed resistance to the inhibitor with high IC(50) values (56.674-1349.788µM). Interestingly, cells with Ras mutations were also sensitive to PLX4032, albeit moderately. Thus, this study has confirmed that the BRAF(T1799A) mutation confers cancer cells sensitivity to PLX4032 and demonstrated its specific potential as an effective and BRAF(T1799A) mutation-selective therapeutic agent for thyroid cancer.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Indóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Sulfonamidas/farmacologia , Neoplasias da Glândula Tireoide/genética , Substituição de Aminoácidos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Indóis/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Sulfonamidas/uso terapêutico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Vemurafenib
6.
Nat Med ; 10(4): 374-81, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15034568

RESUMO

Genomic amplification at 20q11-13 is a common event in human cancers. We isolated a germline translocation breakpoint at 20q11 from a bladder cancer patient. We identified CDC91L1, the gene encoding CDC91L1 (also called phosphatidylinositol glycan class U (PIG-U), a transamidase complex unit in the glycosylphosphatidylinositol (GPI) anchoring pathway), as the only gene whose expression was affected by the translocation. CDC91L1 was amplified and overexpressed in about one-third of bladder cancer cell lines and primary tumors, as well as in oncogenic uroepithelial cells transformed with human papillomavirus (HPV) E7. Forced overexpression of CDC91L1 malignantly transformed NIH3T3 cells in vitro and in vivo. Overexpression of CDC91L1 also resulted in upregulation of the urokinase receptor (uPAR), a GPI-anchored protein, and in turn increased STAT-3 phosphorylation in bladder cancer cells. Our findings suggest that CDC91L1 is an oncogene in bladder cancer, and implicate the GPI anchoring system as a potential oncogenic pathway and therapeutic target in human cancers.


Assuntos
Oncogenes , Neoplasias da Bexiga Urinária/genética , Animais , Cromossomos Humanos Par 20 , Clonagem Molecular , Imunofluorescência , Humanos , Hibridização in Situ Fluorescente , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Translocação Genética
7.
Genomics ; 96(2): 67-72, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20382216

RESUMO

While the methylation machinery of mammalian cells has been shown to be capable of both maintenance and de novo methylation at CpNpG sites, CpNpG methylation in the human genome has not been demonstrated. Here, we report the first observation of 5-methylcytosines in CpNpG triplets in the human genome. We identify the existence of CpNpG methylation in a number of genes which contain trinucleotide repeat regions, including the androgen receptor (AR). We further analyzed DNA extracted from primary tissue samples and found the same pattern of CpNpG methylation. To confirm our results, we performed Southern blot analysis by analyzing the cleavage sites of restriction enzymes within exon 1 of the AR gene and found direct evidence of the presence of 5mCs in CpNpG triplets in the human genome. Our results also suggest that this methylation pattern may be due to the human DNA methyltransferases DNMT1 and DNMT3A. Although the functional significance needs to be tested further, the discovery of inheritable CpNpG methylation in the human genome may have important implications in our understanding of gene regulation and of the development of various diseases, including cancer.


Assuntos
5-Metilcitosina/análise , Metilação de DNA , Genoma Humano/genética , Repetições de Trinucleotídeos/genética , Sequência de Bases , Southern Blotting , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Primers do DNA/genética , Genes/genética , Humanos , Dados de Sequência Molecular , Receptores Androgênicos/genética , Análise de Sequência de DNA
8.
Int J Cancer ; 127(12): 2965-73, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21351275

RESUMO

We examined the therapeutic potential of a novel MEK inhibitor, RDEA119, and its synergism with the mTOR inhibitor, temsirolimus, in thyroid cancer cell lines. RDEA119 potently inhibited the proliferation of the 4 cell lines that harbored BRAF mutation but had no or modest effects on the other 4 cells that harbored wild-type BRAF (IC(50) of 0.034-0.217 µM vs. 1.413-34.120 µM). This inhibitory effect of RDEA119 in selected cell lines OCUT1 (BRAF V600E(+), PIK3CA H1047R(+)) and SW1376 (BRAF V600E(+)) was enhanced by combination with the mTOR inhibitor, temsirolimus. The PTEN-deficient cell FTC133 was highly sensitive to temsirolimus but insensitive to RDEA119, and simultaneous treatment with the latter enhanced the sensitivity of the cell to the former. The KAT18 (wild-type) cell was not sensitive to either drug alone but became sensitive to the combination of the 2 drugs. The drug synergy was confirmed by combination index and isobologram analyses. RDEA119 and temsirolimus also showed synergistic effects on autophagic death of OCUT1 and KAT18 cells selectively tested. Dramatic synergistic effects of the 2 drugs were also seen on the growth of FTC133 xenograft tumors in nude mice. Overall, the effects of the 2 drugs on cell proliferation or autophagic death, either alone or in combination, were more pronounced in cells that harbored genetic alterations in the MAP kinase and PI3K/Akt pathways. Thus, these results demonstrated the important therapeutic potential of the novel MEK inhibitor RDEA119 and its synergism with temsirolimus in thyroid cancer.


Assuntos
Difenilamina/análogos & derivados , MAP Quinase Quinase Quinases/antagonistas & inibidores , Mutação/genética , Proteínas Proto-Oncogênicas B-raf/genética , Sirolimo/análogos & derivados , Sulfonamidas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Neoplasias da Glândula Tireoide/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Difenilamina/farmacologia , Sinergismo Farmacológico , Citometria de Fluxo , Humanos , Camundongos , Camundongos Nus , Sirolimo/farmacologia , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Clin Cancer Res ; 14(12): 3754-60, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559593

RESUMO

PURPOSE: Prostate cancer is a major cause of cancer death among men and the development of new biomarkers is important to augment current detection approaches. EXPERIMENTAL DESIGN: We identified hypermethylation of the ssDNA-binding protein 2 (SSBP2) promoter as a potential DNA marker for human prostate cancer based on previous bioinformatics results and pharmacologic unmasking microarray. We then did quantitative methylation-specific PCR in primary prostate cancer tissues to confirm hypermethylation of the SSBP2 promoter, and analyzed its correlation with clinicopathologic data. We further examined SSBP2 expression in primary prostate cancer and studied its role in cell growth. RESULTS: Quantitative methylation-specific PCR results showed that the SSBP2 promoter was hypermethylated in 54 of 88 (61.4%) primary prostate cancers versus 0 of 23 (0%) in benign prostatic hyperplasia using a cutoff value of 120. Furthermore, we found that expression of SSBP2 was down-regulated in primary prostate cancers and cancer cell lines. Hypermethylation of the SSBP2 promoter and its expression were closely associated with higher stages of prostate cancer. Reactivation of SSBP2 expression by the demethylating agent 5-aza-2'-deoxycytidine in prostate cancer cell lines confirmed epigenetic inactivation as one major mechanism of SSBP2 regulation. Moreover, forced expression of SSBP2 inhibited prostate cancer cell proliferation in the colony formation assay and caused cell cycle arrest. CONCLUSION: SSBP2 inhibits prostate cancer cell proliferation and seems to represent a novel prostate cancer-specific DNA marker, especially in high stages of human prostate cancer.


Assuntos
Adenocarcinoma/genética , Proliferação de Células , Metilação de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Neoplasias da Próstata/genética , Adenocarcinoma/patologia , Idoso , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sequência de Bases , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/fisiologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Análise Mutacional de DNA , Decitabina , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Estadiamento de Neoplasias , Hiperplasia Prostática/genética , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas
10.
Cancer Res ; 67(9): 4123-9, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17456585

RESUMO

Promoter hypermethylation is one of the common mechanisms leading to gene silencing in various human cancers. Using a combination of pharmacologic unmasking and microarray techniques, we identified 59 candidate hypermethylated genes, including LOXL1, a lysyl oxidase-like gene, in human bladder cancer cells. We further showed that LOXL1 and LOXL4 are commonly silenced genes in human bladder cancer cells, and this silence is predominantly related to promoter methylation. We also found LOXL1 and LOXL4 gene methylation and loss of expression in primary bladder tumors. In addition, somatic mutations were identified in LOXL4, but not in LOXL1 in bladder cancer. Moreover, reintroduction of LOXL1 and LOXL4 genes into human bladder cancer cells leads to a decrease of colony formation ability. Further studies indicated that the overexpression of LOXL1 and LOXL4 could antagonize Ras in activating the extracellular signal-regulated kinase (ERK) signaling pathway. Thus, our current study suggests for the first time that lysyl oxidase-like genes can act as tumor suppressor genes and exert their functions through the inhibition of the Ras/ERK signaling pathway in human bladder cancer.


Assuntos
Aminoácido Oxirredutases/genética , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Neoplasias da Bexiga Urinária/genética , Proteínas ras/antagonistas & inibidores , Aminoácido Oxirredutases/metabolismo , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Linhagem Celular Tumoral , Citoplasma/enzimologia , Metilação de DNA/efeitos dos fármacos , Decitabina , Epigênese Genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Enzimológica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Humanos , Sistema de Sinalização das MAP Quinases , Mutação , Células-Tronco Neoplásicas/enzimologia , Células-Tronco Neoplásicas/patologia , Proteína-Lisina 6-Oxidase , Neoplasias da Bexiga Urinária/enzimologia , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Proteínas ras/metabolismo
11.
Biochem Biophys Res Commun ; 370(1): 38-43, 2008 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-18346456

RESUMO

The pathogenesis of breast cancer involves multiple genetic and epigenetic events. In this study, we report an epigenetic alteration of DFNA5 in human breast cancer. DFNA5 gene was silenced in breast cancer cell lines that were methylated in the DFNA5 promoter, and restored by treatment with the demethylating agent, 5-aza-dC, and gene knock-down of DFNA5 increased cellular invasiveness in vitro. The mRNA expression of DFNA5 in breast cancer tissues was down-regulated as compared to normal tissues. Moreover, the DFNA5 promoter was found to be methylated in primary tumor tissues with high frequency (53%, 18/34). Quantitative methylation-specific PCR of DFNA5 clearly discriminated primary breast cancer tissues from normal breast tissues (15.3%, 2/13). Moreover, methylation status of DFNA5 was correlated with lymph node metastasis in breast cancer patients. Our data implicate DFNA5 promoter methylation as a novel molecular biomarker in human breast cancer.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/patologia , Metilação de DNA , Epistasia Genética , Regulação Neoplásica da Expressão Gênica , Receptores de Estrogênio/genética , Adulto , Idoso , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Humanos , Metástase Linfática , Pessoa de Meia-Idade , Invasividade Neoplásica , RNA Mensageiro , Receptores de Estrogênio/antagonistas & inibidores
12.
Biochem Biophys Res Commun ; 366(2): 321-7, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18042467

RESUMO

Phosphorylation pathway has been identified as an important step in membrane trafficking for AQP5. We generated stably transfected BEAS-2B human bronchial epithelial cells with various over-expression constructs on permeable support. In stable cells with wild-type AQP5 and S156A (AQP5 mutant targeting PKA consensus sequence), AQP5 expression was predominantly polarized to the apical membrane, whereas stable cells with N185D (AQP5 mutant targeting second NPA motif), mainly localized to the cytoplasm. Treatment with H89 and/or chlorophenylthio-cAMP (cpt-cAMP) did not affect membrane expression of AQP5 in any of three stable cells. In cells with wild-type AQP5 and N185D, AQP5s were phosphorylated by PKA, while phosphorylation of AQP5 was not detected in cells with S156A. These results indicate that, in AQP5, serine156 may be phosphorylated by PKA, but membrane expression of AQP5 may not be regulated by PKA phosphorylation. We conclude that AQP5 membrane targeting can include more than one mechanism besides cAMP dependent phosphorylation.


Assuntos
Aquaporina 5/metabolismo , Brônquios/metabolismo , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Células Epiteliais/metabolismo , Transporte Proteico/fisiologia , Linhagem Celular , Humanos , Fosforilação
13.
Mod Pathol ; 21(8): 979-91, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18487995

RESUMO

The glycosylphosphatidylinositol transamidase complex (GPIT) consists of five subunits: PIG-U, PIG-T, GPAA1, PIG-S and GPI8, and is important in attaching GPI anchors to target proteins. On the basis of our previous reports incriminating PIG-U as an oncogene in bladder cancer and PIG-T and GPAA1 as oncogenes in breast cancer, we evaluated the expression pattern of the GPIT subunits in 19 different human cancers at both mRNA and protein levels. In general, our results demonstrate a more frequent expression of GPIT subunits in cancers than in normal. Among the 19 anatomic sites compared; breast, ovary and uterus showed consistent evidence of overexpression of specific GPIT subunits. There was also overexpression of PIG-U and GPI8 in lymphoma. In addition, non-small cell lung carcinoma showed significant overexpression of the GPIT subunits as compared to small cell lung carcinoma and normal lung tissue. Also, deregulation of specific GPIT subunits was seen in various other cancers. Forced overexpression of two GPIT subunits; PIG-S and GPI8 alone or in combination induced increased proliferation and invasion of breast cancer cells. Collectively, our study defines a trend involving the deregulated expression and the functional contribution of the GPIT subunits in various cancers with potential implications in diagnosis, prognosis and therapeutic intervention.


Assuntos
Aciltransferases/genética , Aciltransferases/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Neoplasias/enzimologia , Neoplasias/genética , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Invasividade Neoplásica , Neoplasias/patologia
14.
Cancer Lett ; 264(1): 54-62, 2008 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-18423983

RESUMO

Overexpression of several aquaporins has been reported in different types of human cancer but the role of AQPs in human carcinogenesis has not yet been clearly defined. Here, we demonstrate that ectopic expression of human AQP5 (hAQP5), a water channel expressed in lung, salivary glands, and kidney, induces many phenotypic changes characteristic of transformation both in vitro and in vivo. Furthermore, the cell proliferative ability of AQP5 appears to be dependent upon the phosphorylation of a cAMP-protein kinase (PKA) consensus site located in a cytoplasmic loop of AQP5. In addition, phosphorylation of the PKA consensus site was found to be phosphorylated preferentially in tumors. These findings altogether indicate that hAQP5 plays an important role in human carcinogenesis and, furthermore, provide an attractive therapeutic target.


Assuntos
Aquaporina 5/metabolismo , Transformação Celular Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Aquaporina 5/efeitos dos fármacos , Aquaporina 5/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Humanos , Camundongos , Células NIH 3T3 , Neoplasias/genética , Fosforilação , Proteínas Proto-Oncogênicas/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Regulação para Cima/genética
15.
BJU Int ; 101(1): 113-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17941920

RESUMO

OBJECTIVE: To investigate cell division cycle 91-like 1 (CDC91L1; also called phosphatidylinositol glycan class U, PIG-U) expression in bladder cancer at both the mRNA and protein levels, and to study its clinical and prognostic significance, as CDC91L1 was recently identified as a new oncogene in human bladder cancer and its role in the biological behaviour of bladder cancer is largely unknown. PATIENTS AND METHODS: In all, 73 bladder tumours and 14 samples of normal bladder urothelium were studied by reverse-transcription polymerase chain reaction (PCR), real-time quantitative PCR and immunohistochemistry. RESULTS: The normalized CDC91L1 mRNA copy number in tumours was significantly greater than in normal controls (P < 0.05). There was overexpression of CDC91L1 mRNA in 30.1% (22/73) of the bladder tumours compared with the normal urothelium. At the protein level, 75.3% (55/73) of the bladder tumours and two of 14 of the normal urothelium had high expression of CDC91L1 protein, which is statistically significant (P < 0.001). The correlation between CDC91L1 protein and tumour grade, and muscle invasion of tumour was significant (both P < 0.05). In addition to tumour extent and tumour grade, CDC91L1 protein was an independent predictor of recurrence for superficial bladder cancer and had a trend to predict tumour progression. CONCLUSIONS: CDC91L1 (PIG-U) plays a role in the development of bladder urothelial cell carcinoma. CDC91L1 protein might be a potential biomarker for prediction of recurrence and a therapeutic target in bladder cancer.


Assuntos
Aciltransferases/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias da Bexiga Urinária/patologia , Estudos de Casos e Controles , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Recidiva Local de Neoplasia/prevenção & controle , Estadiamento de Neoplasias , Prognóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Urotélio/patologia
16.
Mol Cell Biol ; 25(14): 6077-89, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15988020

RESUMO

p63 is a member of the p53 tumor suppressor gene family, which regulates downstream target gene expression by binding to sequence-specific response elements similar to those of p53. By using oligonucleotide expression microarray analysis and analyzing the promoters of p63-induced genes, we have identified novel p63-specific response elements (p63-REs) in the promoter regions of EVPL and SMARCD3. These p63-REs exhibit characteristic differences from the canonical p53-RE (RRRCWWGYYY) in both the core-binding element (CWWG) as well as the RRR and/or YYY stretches. Luciferase assays on mutagenized promoter constructs followed by electromobility shift analysis showed that p53 preferentially activates and binds to the RRRCATGYYY sequence, whereas p63 preferentially activates RRRCGTGYYY. Whereas EVPL protein is highly expressed in epithelial cells of the skin and pharynx in the p63+/+ mouse, it is undetectable in these tissues in the p63-/- mouse. Our results indicate that p63 can regulate expression of specific target genes such as those involved in skin, limb, and craniofacial development by preferentially activating distinct p63-specific response elements.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/genética , Fosfoproteínas/metabolismo , Precursores de Proteínas/genética , Elementos de Resposta/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Desenvolvimento Ósseo/genética , Proteínas Cromossômicas não Histona , Proteínas de Ligação a DNA , Genes Reporter , Genes Supressores de Tumor , Humanos , Proteínas de Membrana/análise , Camundongos , Dados de Sequência Molecular , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Faringe/crescimento & desenvolvimento , Faringe/imunologia , Fosfoproteínas/genética , Regiões Promotoras Genéticas/genética , Precursores de Proteínas/análise , Pele/crescimento & desenvolvimento , Pele/imunologia , Transativadores/genética , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor
17.
Cancer Res ; 66(20): 9829-36, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17047043

RESUMO

Based on the oncogenic role of phosphatidylinositol glycan (PIG) class U in human tumors, we explored the role of two additional subunits of the glycosylphosphatidylinositol (GPI) transamidase complex in human breast cancer. We found that PIG class T (PIG-T) and GPI anchor attachment 1 (GPAA1) were overexpressed in breast cancer cell lines and primary tumors. Forced expression of PIG-T and GPAA1 transformed NIH3T3 cells in vitro and increased tumorigenicity and invasion of these cells in vivo. Suppression of PIG-T expression in breast cancer cell lines led to inhibition of anchorage-independent growth. Moreover, we found that PIG-T and GPAA1 expression levels positively correlated with paxillin phosphorylation in invasive breast cancer cell lines. Furthermore, suppression of PIG-T and GPAA1 expression led to a decrease in paxillin phosphorylation with a concomitant decrease in invasion ability. These results suggest that the GPI transamidase complex is composed of a group of proto-oncogenes that individually or as a group contribute to breast cancer growth. This aberrant growth is mediated, at least partially, by phosphorylation of paxillin, contributing to invasion and progression of breast cancer.


Assuntos
Aciltransferases/biossíntese , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Glicosilfosfatidilinositóis/biossíntese , Glicoproteínas de Membrana/biossíntese , Oncogenes , Aciltransferases/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Clonagem Molecular , Amplificação de Genes , Dosagem de Genes , Glicosilfosfatidilinositóis/genética , Humanos , Glicoproteínas de Membrana/genética , Invasividade Neoplásica , Paxilina/metabolismo , Fosforilação , Subunidades Proteicas
18.
Trends Biotechnol ; 36(6): 586-593, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-28755977

RESUMO

Plasma is an ionized gas that is typically formed under high-temperature laboratory conditions. Recent progress in atmospheric plasmas has led to cold atmospheric plasma (CAP) devices with ion temperatures close to room temperature. The unique chemical and physical properties of CAP have led to its use in various biomedical applications including cancer therapy. CAP exhibits a spontaneous transition from a spatially homogeneous state to a modifiable pattern that is subject to self-organization. In this Opinion article, we discuss some new applications for plasma in cancer therapy based on plasma self-organization, which enables adaptive features in plasma-based therapeutic systems.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Melanoma/terapia , Gases em Plasma/uso terapêutico , Medicina de Precisão/métodos , Neoplasias Cutâneas/terapia , Animais , Antineoplásicos Alquilantes/farmacologia , Aquaporinas/genética , Aquaporinas/metabolismo , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Espécies Reativas de Nitrogênio/agonistas , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Temozolomida/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Mol Cancer ; 6: 74, 2007 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18028549

RESUMO

BACKGROUND: GPI anchor attachment is catalyzed by the GPI transamidase (GPIT) complex. GAA1, PIG-T and PIG-U are the three of five GPIT subunits. Previous studies demonstrated amplification and overexpression of GPIT subunits in bladder and breast cancer with oncogenic function. We performed an analysis of these subunits in head and neck squamous cell carcinoma (HNSCC). RESULTS: To evaluate GAA1, PIG-T and PIG-U in HNSCC, we used quantitative PCR (QPCR) and quantitative RT-PCR (QRT-PCR) to determine the copy number of those genes in primary tumors and the matching lymphocytes in 28 patients with HNSCC and quantified RNA expression of those genes in 16 primary HNSCC patients and 4 normal control tissue samples. GAA1 showed a significant increase in normalized mRNA expression, 2.11 (95% CI: 1.43, 2.79), in comparison to that of normal controls, 0.43 (95% CI: -0.76, 1.61), p = 0.014 (Mann-Whitney test). The mean genomic copy number of GAA1 was significantly increased in HNSCC, 0.59 (95% CI: 0.50, 0.79), in comparison to lymphocyte DNA, 0.35 (95% CI: 0.30, 0.50), p = 0.001 (paired t-test). CONCLUSION: An increased expression level and elevated copy number for GAA1 suggest a role for this GPI anchor subunit in HNSCC.


Assuntos
Aciltransferases/genética , Carcinoma de Células Escamosas/genética , Neoplasias de Cabeça e Pescoço/genética , Estudos de Casos e Controles , Linhagem Celular , Dosagem de Genes/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Glicoproteínas de Membrana/genética , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Cancer Res ; 65(3): 758-66, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15705872

RESUMO

HSP70, a stress response protein, is known to be a determinant of cell death and cell transformation. We show that different isoforms of p63 have different transcriptional activities on hsp70 genes. DeltaNp63alpha, an abundantly expressed isoform of p63, activates (in vitro and in vivo), whereas TAp63gamma down-regulates the expression of hsp70. We further show that the transactivation domain at the NH(2) terminus of p63 represses, whereas the COOH terminus activates hsp70 transcription. In addition, DeltaNp63alpha regulates transcription of the hsp70 gene through its interaction with the CCAAT binding factor and NF-Y transcription factors which are known to form a complex with the CCAAT box located in the hsp70 promoter. Moreover, DeltaNp63alpha expression correlates with HSP70 expression in all head and neck cancer cell lines. Finally, we show colocalization of DeltaNp63alpha and HSP70 in the epithelium and coexpression of both proteins in 41 primary head and neck cancers. Our study provides strong evidence for the physiologic association between DeltaNp63alpha and hsp70 in human cancer, thus further supporting the oncogenic potential of DeltaNp63alpha.


Assuntos
Proteínas de Choque Térmico HSP70/genética , Neoplasias/genética , Fosfoproteínas/fisiologia , Transativadores/fisiologia , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Fator de Ligação a CCAAT/metabolismo , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Proteínas de Choque Térmico HSP70/biossíntese , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Neoplasias/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Osteossarcoma/genética , Osteossarcoma/metabolismo , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Estrutura Terciária de Proteína , Transativadores/biossíntese , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição , Ativação Transcricional , Proteínas Supressoras de Tumor , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA