Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 36(47): 6542-6554, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-28759046

RESUMO

Current anti-epidermal growth factor receptor (EGFR) therapy for oral cancer does not provide satisfactory efficacy due to drug resistance or reduced EGFR level. As an alternative candidate target for therapy, here we identified an oncogene, ROS1, as an important driver for oral squamous cell carcinoma (OSCC) metastasis. Among tumors from 188 oral cancer patients, upregulated ROS1 expression strongly correlated with metastasis to lung and lymph nodes. Mechanistic studies uncover that the activated ROS1 results from highly expressed ROS1 gene instead of gene rearrangement, a phenomenon distinct from other cancers. Our data further reveal a novel mechanism that reduced histone methyltransferase EZH2 leads to a lower trimethylation of histone H3 lysine 27 suppressive modification, relaxes chromatin, and promotes the accessibility of the transcription factor STAT1 to the enhancer and the intron regions of ROS1 target genes, CXCL1 and GLI1, for upregulating their expressions. Down-regulation of ROS1 in highly invasive OSCC cells, nevertheless, reduces cell proliferation and inhibits metastasis to lung in the tail-vein injection and the oral cavity xenograft models. Our findings highlight ROS1 as a candidate biomarker and therapeutic target for OSCC. Finally, we demonstrate that co-targeting of ROS1 and EGFR could potentially offer an effective oral cancer therapy.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/secundário , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células , Quimiocina CXCL1/metabolismo , Regulação para Baixo , Receptores ErbB/antagonistas & inibidores , Histonas/metabolismo , Humanos , Masculino , Metilação , Camundongos , Terapia de Alvo Molecular/métodos , Neoplasias Bucais/tratamento farmacológico , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Fator de Transcrição STAT1/metabolismo , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína GLI1 em Dedos de Zinco/metabolismo
2.
Cell Death Dis ; 5: e1409, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25188519

RESUMO

Btk and Etk/BMX are Tec-family non-receptor tyrosine kinases. Btk has previously been reported to be expressed primarily in B cells and has an important role in immune responses and B-cell malignancies. Etk has been shown previously to provide a strong survival and metastasis signal in human prostate cancer cells, and to confer androgen independence and drug resistance. While the role of Etk in prostate carcinogenesis is well established, the functions of Btk in prostate cancer have never been investigated, likely due to the perception that Btk is a hematopoietic, but not epithelial, kinase. Herein, we found that Btk is overexpressed in prostate cancer tissues and prostate cancer cells. The level of Btk in prostate cancer tissues correlates with cancer grades. Knockdown of Btk expression selectively inhibits the growth of prostate cancer cells, but not that of the normal prostate epithelial cells, which express very little Btk. Dual inhibition of Btk and Etk has an additive inhibitory effect on prostate cancer cell growth. To explore Btk and Etk as targets for prostate cancer, we developed a small molecule dual inhibitor of Btk and Etk, CTN06. Treatment of PC3 and other prostate cancer cells, but not immortalized prostate epithelial cells with CTN06 resulted in effective cell killing, accompanied by the attenuation of Btk/Etk signals. The killing effect of CTN06 is more potent than that of commonly used inhibitors against Src, Raf/VEGFR and EGFR. CTN06 induces apoptosis as well as autophagy in human prostate cancer cells, and is a chemo-sensitizer for docetaxel (DTX), a standard of care for metastatic prostate cancer patients. CTN06 also impeded the migration of human prostate cancer cells based on a 'wound healing' assay. The anti-cancer effect of CTN06 was further validated in vivo in a PC3 xenograft mouse model.


Assuntos
Proliferação de Células/efeitos dos fármacos , Imidazóis/toxicidade , Inibidores de Proteínas Quinases/toxicidade , Proteínas Tirosina Quinases/metabolismo , Quinoxalinas/toxicidade , Tirosina Quinase da Agamaglobulinemia , Animais , Antineoplásicos/uso terapêutico , Antineoplásicos/toxicidade , Autofagia/efeitos dos fármacos , Sítios de Ligação , Linhagem Celular Tumoral , Docetaxel , Humanos , Imidazóis/química , Imidazóis/uso terapêutico , Masculino , Camundongos , Camundongos Nus , Simulação de Acoplamento Molecular , Fosfolipase C gama/metabolismo , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/uso terapêutico , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinoxalinas/química , Quinoxalinas/uso terapêutico , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator de Transcrição STAT3/metabolismo , Taxoides/uso terapêutico , Taxoides/toxicidade , Transplante Heterólogo
3.
Oncogene ; 33(36): 4521-30, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-24662833

RESUMO

Macro-autophagy is associated with drug resistance in various cancers and can function as an adaptive response to maintain cell survival under metabolic stresses, including androgen deprivation. Androgen deprivation or treatment with androgen receptor (AR) signaling inhibitor (ARSI), Enzalutamide (MDV-3100, ENZA) or bicalutamide induced autophagy in androgen-dependent and in castration-resistant CaP (castration-resistant prostate cancer (CRPC)) cell lines. The autophagic cascade triggered by AR blockage, correlated with the increased light chain 3-II/I ratio and ATG-5 expression. Autophagy was observed in a subpopulation of C4-2B cells that developed insensitivity to ENZA after sustained exposure in culture. Using flow cytometry and clonogenic assays, we showed that inhibiting autophagy with clomipramine (CMI), chloroquine or metformin increased apoptosis and significantly impaired cell viability. This autophagic process was mediated by AMP-dependent protein kinase (AMPK) activation and the suppression of mammalian target of rapamycin (mTOR) through Raptor phosphorylation (Serine 792). Furthermore, small interfering RNA targeting AMPK significantly inhibited autophagy and promoted cell death in CaP cells acutely or chronically exposed to ENZA or androgen deprivation, suggesting that autophagy is an important survival mechanism in CRPC. Lastly, in vivo studies with mice orthotopically implanted with ENZA-resistant cells demonstrated that the combination of ENZA and autophagy modulators, CMI or metformin significantly reduced tumor growth when compared with control groups (P<0.005). In conclusion, autophagy is as an important mechanism of resistance to ARSI in CRPC. Antiandrogen-induced autophagy is mediated through the activation of AMPK pathway and the suppression of mTOR pathway. Blocking autophagy pharmacologically or genetically significantly impairs prostate cancer cell survival in vitro and in vivo, implying the therapeutics potential of autophagy inhibitors in the antiandrogen-resistance setting.


Assuntos
Antagonistas de Receptores de Andrógenos/uso terapêutico , Autofagia/efeitos dos fármacos , Cloroquina/farmacologia , Clomipramina/farmacologia , Metformina/farmacologia , Feniltioidantoína/análogos & derivados , Animais , Benzamidas , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos SCID , Nitrilas , Feniltioidantoína/uso terapêutico , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/patologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Oncogene ; 33(19): 2495-503, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-23728339

RESUMO

Src tyrosine kinase (Src) is implicated in the development of bone metastasis and castration resistance of prostate cancer. Src inhibitors are currently being tested in clinical trials for such diseases. Understanding the molecular and cellular actions of Src inhibitors holds the key to future improvement of this line of therapy. Here we describe the microRNA expression profiles modulated by two Src inhibitors and demonstrate that the miR-30 family members are the most prominently induced species. Consistent with its tumor suppressor role, miR-30 is downmodulated by oncogenic signals such as epidermal growth factor (EGF) and hepatocyte growth factor, and is generally underexpressed in prostate cancer specimens. A number of epithelial-to-mesenchymal transition (EMT)-associated genes are predicted targets of miR-30. Among these genes the Ets-related gene (ERG) is the most frequently overexpressed oncogene in prostate cancer activated by genomic fusion events between promoter upstream sequences of the TMPRSS2 and coding sequences of ERG. We showed by ERG 3' untranslated region reporter and mutagenesis assays that ERG is a direct target of miR-30. Overexpression of miR-30 in prostate cancer cells suppresses EMT phenotypes and inhibits cell migration and invasion. It also inhibits the in vitro and in vivo growth of VCaP cells, which depends on TMPRSS2-ERG for proliferation. TMPRSS2-ERG is generally regulated by androgen at the transcriptional level. Our finding reveals a new post-transcriptional mechanism of TMPRSS2-ERG regulation by Src and growth signals via miR-30 providing a rationale for targeting ERG-positive castration-resistant tumors with Src inhibitors.


Assuntos
Transição Epitelial-Mesenquimal/genética , MicroRNAs/genética , Neoplasias da Próstata/genética , Transdução de Sinais/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Imunofluorescência , Genes Supressores de Tumor , Xenoenxertos , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Neoplasias da Próstata/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Transativadores/genética , Transativadores/metabolismo , Regulador Transcricional ERG , Quinases da Família src/genética , Quinases da Família src/metabolismo
5.
Oncogene ; 32(35): 4130-8, 2013 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-23069658

RESUMO

Although prostate cancer (CaP) is the most frequently diagnosed malignant tumor in American men, the mechanisms underlying the development and progression of CaP remain largely unknown. Recent studies have shown that downregulation of the microRNA miR-124 occurs in several types of human cancer, suggesting a tumor suppressive function of miR-124. Until now, however, it has been unclear whether miR-124 is associated with CaP. In the present study, we completed a series of experiments to understand the functional role of miR-124 in CaP. We detected the expression level of miR-124 in clinical CaP tissues, evaluated the influence of miR-124 on the growth of CaP cells and investigated the mechanism underlying the dysregulation of miR-124. We found that (i) miR-124 directly targets the androgen receptor (AR) and subsequently induces an upregulation of p53; (ii) miR-124 is significantly downregulated in malignant prostatic cells compared to benign cells, and DNA methylation causes the reduced expression of miR-124; and (iii) miR-124 can inhibit the growth of CaP cells in vitro and in vivo. Data from this study revealed that loss of miR-124 expression is a common event in CaP, which may contribute to the pathogenesis of CaP. Our studies also suggest that miR-124 is a potential tumor suppressive gene in CaP, and restoration of miR-124 expression may represent a novel strategy for CaP therapy.


Assuntos
Proliferação de Células , Genes Supressores de Tumor/fisiologia , MicroRNAs/fisiologia , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Animais , Apoptose , Linhagem Celular Tumoral , Metilação de DNA , Regulação para Baixo , Humanos , Masculino , Camundongos , Neoplasias da Próstata/patologia , Neoplasias da Próstata/prevenção & controle , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Oncogene ; 31(24): 2907-18, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21986944

RESUMO

Male germ cell-associated kinase (MAK), a direct transcriptional target of androgen receptor (AR), is a co-activator of AR. In this study, we determined the activating mechanism of MAK and identified a previously unknown AR-independent role of MAK in mitosis. We found that MAK kinase activity requires dual phosphorylation of the conserved TDY motif and that the phosphorylation is dynamic during cell cycle. MAK associates with CDH1 (FZR1, fizzy/cell division cycle 20 related 1) and phosphorylates CDH1 at sites phosphorylated by cyclin-dependent kinases. When MAK is overexpressed, the binding of CDH1 to anaphase promoting complex/cyclosome decreased, resulting in an attenuation of anaphase-promoting complex/C ubiquitin ligase activity and the consequential stabilization of the CDH1 targets such as Aurora kinase A and Polo-like kinase 1. As such, overexpression of MAK leads to mitotic defects such as centrosome amplification and lagging chromosomes. Our immunohistochemistry result showed that MAK is overexpressed in prostate tumor tissues, suggesting a role of MAK in prostate carcinogenesis. Taken with our previous results, our data implicate MAK in both AR activation and chromosomal instability, acting in both early and late prostate cancer development.


Assuntos
Instabilidade Cromossômica , Mitose , Neoplasias da Próstata/enzimologia , Proteínas Serina-Treonina Quinases/biossíntese , Receptores Androgênicos/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Motivos de Aminoácidos , Ciclossomo-Complexo Promotor de Anáfase , Aurora Quinase A , Aurora Quinases , Proteínas Cdh1 , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Regulação para Baixo , Humanos , Imuno-Histoquímica , Masculino , Fosforilação , Próstata/citologia , Próstata/enzimologia , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Quinase 1 Polo-Like
7.
Oncogene ; 27(49): 6365-75, 2008 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-18679417

RESUMO

Prostate cancer is the most frequently diagnosed cancer in American men. We have previously demonstrated that Src mediates androgen-independent proliferation in prostate cancer. We sought to investigate the Src-mediated oncogenic pathways and tumor biology using AZD0530, a novel Src family kinase/Abl dual-kinase inhibitor that is entering phase II clinical trials. We show that while both Src and Abl are expressed in all prostate cancer cell lines, Src but not Abl is activated in the prostate. Furthermore, Src activation is inhibited by AZD0530 in a rapid and dose-dependent manner. We show that Src mediates cell proliferation in DU145 and PC3 cells at the G1 phase of cell cycle. Src inhibition resulted in decreased binding of beta-catenin to the promoters of G1 phase cell cycle regulators cyclin D1 and c-Myc. C-Myc may also be regulated at the protein level by extracellular signal-regulated kinase 1/2 and GSK3beta. Cell motility factors focal adhesion kinase, p130CAS and paxillin activation in DU145 and PC3 cells were also inhibited. Administration of AZD0530 in mice reduced orthotopic DU145 xenograft growth by 45%. We have further delineated the Src-mediated oncogenic growth and migration pathways in prostate cancer and established mechanistic rationale for Src inhibition as novel therapy in the treatment of prostate cancer.


Assuntos
Antineoplásicos/farmacologia , Benzodioxóis/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Quinazolinas/farmacologia , Quinases da Família src/antagonistas & inibidores , Antineoplásicos/química , Benzodioxóis/química , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteína Substrato Associada a Crk/metabolismo , Ciclina D , Ciclinas/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Masculino , Estrutura Molecular , Fosforilação/efeitos dos fármacos , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Quinazolinas/química , Proteínas Repressoras/metabolismo , Fatores de Tempo , beta Catenina/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo
8.
Oncogene ; 27(4): 499-505, 2008 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-17653089

RESUMO

We have previously demonstrated that human H2-relaxin can mediate androgen-independent growth of LNCaP through a mechanism that involves the activation of the androgen receptor (AR) signaling pathway. The goal of the current study is to elucidate the mechanism(s) by which H2-relaxin causes activation of the AR pathway. Our data indicate that there is cross-talk between AR and components of the Wnt signaling pathway. Addition of H2-relaxin to LNCaP cells resulted in increased phosphorylation of protein kinase B (Akt) and inhibitory phosphorylation of glycogen synthase kinase-3beta (GSK-3beta) with subsequent cytoplasmic accumulation of beta-catenin. Immunoprecipitation and immunocytochemical studies demonstrated that the stabilized beta-catenin formed a complex with AR, which was then translocated into the nucleus. Chromatin immunoprecipitation analysis determined that the AR/beta-catenin complex binds to the proximal region of the prostate-specific antigen promoter. Inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, using LY294002, prevented both H2-relaxin-mediated phosphorylation of Akt and GSK-3beta and translocation of beta-catenin/AR into the nucleus. Knockdown of beta-catenin levels using a beta-catenin-specific small interfering RNA inhibited H2-relaxin-induced AR activity. The combined data demonstrate that PI3K/Akt and components of the Wnt pathway can facilitate H2-relaxin-mediated activation of the AR pathway.


Assuntos
Receptores Androgênicos/metabolismo , Relaxina/fisiologia , beta Catenina/fisiologia , Núcleo Celular/efeitos dos fármacos , Cromonas/farmacologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Masculino , Modelos Biológicos , Morfolinas/farmacologia , Fosforilação , Regiões Promotoras Genéticas , Antígeno Prostático Específico/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas , beta Catenina/metabolismo
9.
Cancer Lett ; 254(2): 265-73, 2007 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-17449174

RESUMO

Because of the current controversy on the origin and clinical value of circulating KRAS codon 12 mutations in lung cancer, we screened 180 patients using a combined restriction fragment-length polymorphism and polymerase chain reaction (RFLP-PCR) assay. We detected KRAS mutations in 9% plasma samples and 0% matched lymphocytes. Plasma KRAS mutations correlated significantly with poor prognosis. We validated the positive results in a second laboratory by DNA sequencing and found matching codon 12 sequences in blood and tumor in 78% evaluable cases. These results support the notion that circulating KRAS mutations originate from tumors and are prognostically relevant in lung cancer.


Assuntos
Neoplasias Pulmonares/genética , Mutação , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , DNA de Neoplasias/sangue , DNA de Neoplasias/genética , Feminino , Humanos , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Prognóstico , Proteínas Proto-Oncogênicas p21(ras) , Análise de Sobrevida
10.
Virus Genes ; 27(3): 211-8, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14618081

RESUMO

Marek's disease virus latent protein MEQ (MDV Eco Q) is abundantly expressed and consistently detected in MDV-induced tumors and cell lines. Deletion mutants were constructed to study the domain structure of MEQ. Four deletion mutants were obtained in the basic regions of MEQ, namely basic region 1 (DeltaBR1), basic region 2 (DeltaBR2), basic regions 1 and 2 (DeltaBR1 and 2), and the C-terminal (bZIP) domain. The BR1 and BR2 are nuclear localization signals and either is sufficient to cause transport of MEQ into the nucleus. In addition, the BR2 is also responsible for MEQ's nucleolar localization. A monoclonal antibody (Mab 23B46) was produced using recombinant fowlpox virus (rFPV) expressing MEQ (rFPV/MEQ) as a source of protein. The isotype of Mab 23B46 is IgG1 and immunoprecipitated a band in rFPV/MEQ infected cells with molecular weight of 60 kDa specific to MEQ protein. We detected abundant expression of MEQ in (rFPV/MEQ), recombinant baculovirus (rBac) (rBac/MEQ), and lymphoid tumors induced by MDV. In order to delineate the epitope of MEQ reactive with Mab 23B46, we used four deletion mutants from the basic and bZIP domains. We found the deletions in the N-terminal region including BR1 (DeltaBR1), and (DeltaBR1 and 2) completely abolished the specific binding with Mab 23B46 as shown by Western blot analysis and immunofluoresence test. Deletion of BR2 (DeltaBR2) and the C-terminal (bZIP) domain had no effect on antibody binding. These data provide direct evidence that monoclonal antibody reactive epitope is localized in the BR1 domain of the molecule. Since both BR1 and BR2 domains contain sequences important for nuclear entry, we now have reagent to further study and elucidate the mechanism of MEQ's involvement in nuclear and nucleolar localization.


Assuntos
Herpesvirus Galináceo 2/genética , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/genética , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Anticorpos Antivirais , Antígenos Virais/química , Antígenos Virais/genética , Transformação Celular Viral , Células Cultivadas , Epitopos/química , Epitopos/genética , Genes Virais , Herpesvirus Galináceo 2/imunologia , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/virologia , Dados de Sequência Molecular , Sinais de Localização Nuclear , Proteínas Oncogênicas Virais/imunologia , Estrutura Terciária de Proteína , Ratos , Deleção de Sequência
11.
Angiogenesis ; 6(4): 289-93, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-15166497

RESUMO

Once metastatic, prostate cancer (CaP) treatment options are limited to androgen withdrawal. In this environment, the cells often develop an androgen independent state resulting in patient demise. It has been shown that during this transition, CaP cells transdifferentiate to neuroendocrine cells, which produce neuropeptides. These neuropeptides have a mitogenic effect on surrounding CaP cells. Previous observations suggest that endothelial cells may show a similar mitogenic response to neuropeptides, implicating angiogenesis in the progression of CaP. We stimulated human umbilical endothelial cells (HUVECs) with the neuropeptides bombesin and neurotensin and measured proliferation, migration, cell tube formation, and tyrosine kinase activation. In our studies, neurotensin and bombesin did not stimulate HUVEC proliferation, migration, nor tube formation. Although HUVECs express the non-receptor tyrosine kinases Fak, Src, and Etk which mediate neuropeptide signaling in CaP, they are not activated by neuropeptides in HUVECs.


Assuntos
Neovascularização Patológica , Neuropeptídeos/metabolismo , Neoplasias da Próstata/química , Western Blotting , Bombesina/metabolismo , Bombesina/farmacologia , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Masculino , Neuropeptídeos/farmacologia , Neurotensina/metabolismo , Neurotensina/farmacologia , Testes de Precipitina , Proteínas Tirosina Quinases/metabolismo , Fatores de Tempo , Veias Umbilicais/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Quinases da Família src/metabolismo
12.
Virus Genes ; 24(2): 173-80, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12018709

RESUMO

Marek's disease virus, a herpesvirus, and avian leukosis virus, subgroup J, a retrovirus, are oncogenic viruses of poultry. Both viruses may infect the same flock, the same bird and the same cell. In a double-infected cell, the retroviral DNA can integrate into the cellular or the Marek's disease virus (MDV) genome. The retroviral-long terminal repeat (LTR) integration into MDV was first described by Isfort et al., (Proc Natl Acad Sci 89, 991-995, 1992) following tissue culture co-infection. The recombinant virus isolated, RM1, had altered biological properties compared to the parental MDV (Witter R.L., Li D., Jones D., and Kung H.-J., Avian Dis 41, 407-421, 1997) . The issue of retroviral sequence integration into herpesviruses in vivo, in cases of double-virus infection is of wide significance in general virology and veterinary medicine; it also represents a special case of gene transposition. Using the avian system, we aimed to determine occurrence of such integrations in vivo. Chickens were experimentally co-infected with both avian leukosis virus (ALV) subgroup J and with MDV. To demonstrate the presence of the retroviral LTR in the MDV genome we applied the Hot Spot-combined PCR assay (Borenshtain R. and Davidson I., J Virol Meth 82, 119-127, 1999) that consisted of two consecutive steps of amplification. By that HS-cPCR assay, certain MDV genomic sites, defined as HS for integration were specifically amplified, the HS step, and then subjected to screening in an attempt to detect LTR inserts. The screening was achieved by amplification using heterologous primer sets, one for the MDV hot spot and the other for the retroviral LTR, the cPCR step. The products were Southern blotted and hybridized with MDV and ALV-LTR probes. Chimeric molecules were detected and evidenced by an intense signal in 3/10 chickens and weakly in other 3/10 birds. Detection was by LTR amplification, sequencing and multiple alignment to the ALV-J-LTR sequence. The present study indicated that chimeric molecules were produced in vivo.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/complicações , Mardivirus/genética , Doença de Marek/complicações , Sequências Repetidas Terminais/genética , Integração Viral , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/patogenicidade , Galinhas/virologia , Mardivirus/patogenicidade , Reação em Cadeia da Polimerase/métodos , Doenças das Aves Domésticas/virologia , Proteínas Recombinantes de Fusão/genética , Recombinação Genética , Análise de Sequência de DNA
13.
J Virol ; 75(23): 11298-306, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11689610

RESUMO

Human immunodeficiency virus infection in humans and simian immunodeficiency virus (SIV) infection in rhesus macaques (RM) leads to a generalized loss of immune responses involving perturbations in T-cell receptor (TCR) signaling. In contrast, naturally SIV-infected sooty mangabeys (SM) remain asymptomatic and retain immune responses despite relatively high viral loads. However, SIV infection in both RM and SM led to similar decreases in TCR-induced Lck phosphorylation. In this study, a protein tyrosine kinase (PTK) differential display method was utilized to characterize the effects of in vivo SIV infection on key signaling molecules of the CD4(+) T-cell signaling pathways. The CD4(+) T cells from SIV-infected RM, but not SIV-infected SM, showed chronic downregulation of baseline expression of MLK3, PRK, and GSK3, and symptomatically SIV-infected RM showed similar downregulation of MKK3. In vitro TCR stimulation with or without CD28 costimulation of CD4(+) T cells did not lead to the enhancement of gene transcription of these PTKs. While the CD4(+) T cells from SIV-infected RM showed a significant increase of the baseline and anti-TCR-mediated ROR2 transcription, SIV infection in SM led to substantially decreased anti-TCR-stimulated ROR2 transcription. TCR stimulation of CD4(+) T cells from SIV-infected RM (but not SIV-infected SM) led to the repression of CaMKKbeta and the induction of gene transcription of MLK2. Studies of the function of these molecules in T-cell signaling may lead to the identification of potential targets for specific intervention, leading to the restoration of T-cell responses.


Assuntos
Linfócitos T CD4-Positivos/enzimologia , Proteínas Tirosina Quinases/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/enzimologia , Vírus da Imunodeficiência Símia/patogenicidade , Animais , Sequência de Bases , Cercocebus atys , Clonagem Molecular , Primers do DNA , Ativação Linfocitária , Fosforilação , Proteínas Tirosina Quinases/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Transcrição Gênica
14.
Clin Cancer Res ; 7(11): 3640-8, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11705887

RESUMO

PURPOSE: There is a continuing need for genetically matched cell systems to model cellular behaviors that are frequently observed in aggressive breast cancers. EXPERIMENTAL DESIGN: We report here the isolation and initial characterization of a spontaneously arising variant of MCF-10A cells, NeoST, which provides a new model to study cell adhesion and signal transduction in breast cancer. RESULTS: NeoST cells recapitulate important biological and biochemical features of metastatic breast cancer, including anchorage-independent growth, invasiveness in three-dimensional reconstituted membranes, loss of E-cadherin expression, and increased tyrosine kinase activity. A comprehensive analysis of tyrosine kinase expression revealed overexpression or functional activation of the Axl, FAK, and EphA2 tyrosine kinases in transformed MCF-10A cells. CONCLUSIONS: MCF-10A and these new derivatives provide a genetically matched model to study defects in cell adhesion and signaling that are relevant to cellular behaviors that often typify aggressive breast cancer cells.


Assuntos
Neoplasias da Mama/patologia , Adesão Celular/fisiologia , Comunicação Celular/fisiologia , Matriz Extracelular/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/fisiopatologia , Divisão Celular/fisiologia , Linhagem Celular Transformada , Tamanho Celular/fisiologia , Humanos , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Fatores de Tempo
15.
Mol Cell Biol ; 21(24): 8385-97, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11713275

RESUMO

The bombesin/gastrin-releasing peptide (GRP) family of neuropeptides has been implicated in various in vitro and in vivo models of human malignancies including prostate cancers. It was previously shown that bombesin and/or neurotensin (NT) acts as a survival and migratory factor(s) for androgen-independent prostate cancers. However, a role in the transition from an androgen-dependent to -refractory state has not been addressed. In this study, we investigate the biological effects and signal pathways of bombesin and NT on LNCaP, a prostate cancer cell line which requires androgen for growth. We show that both neurotrophic factors can induce LNCaP growth in the absence of androgen. Concurrent transactivation of reporter genes driven by the prostate-specific antigen promoter or a promoter carrying an androgen-responsive element (ARE) indicate that growth stimulation is accompanied by androgen receptor (AR) activation. Furthermore, neurotrophic factor-induced gene activation was also present in PC3 cells transfected with the AR but not in the parental line which lacks the AR. Given that bombesin does not directly bind to the AR and is known to engage a G-protein-coupled receptor, we investigated downstream signaling events that could possibly interact with the AR pathway. We found that three nonreceptor tyrosine kinases, focal adhesion kinase (FAK), Src, and Etk/BMX play important parts in this process. Etk/Bmx activation requires FAK and Src and is critical for neurotrophic factor-induced growth, as LNCaP cells transfected with a dominant-negative Etk/BMX fail to respond to bombesin. Etk's activation requires FAK, Src, but not phosphatidylinositol 3-kinase. Likewise, bombesin-induced AR activation is inhibited by the dominant-negative mutant of either Src or FAK. Thus, in addition to defining a new G-protein pathway, this report makes the following points regarding prostate cancer. (i) Neurotrophic factors can activate the AR, thus circumventing the normal growth inhibition caused by androgen ablation. (ii) Tyrosine kinases are involved in neurotrophic factor-mediated AR activation and, as such, may serve as targets of future therapeutics, to be used in conjunction with current antihormone and antineuropeptide therapies.


Assuntos
Androgênios/metabolismo , Neuropeptídeos/metabolismo , Proteínas Tirosina Quinases/fisiologia , Quinases da Família src/fisiologia , Bombesina/metabolismo , Bombesina/farmacologia , Bromodesoxiuridina/metabolismo , Divisão Celular , Ativação Enzimática , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Genes Dominantes , Genes Reporter , Humanos , Luciferases/metabolismo , Masculino , Modelos Biológicos , Plasmídeos/metabolismo , Testes de Precipitina , Neoplasias da Próstata/metabolismo , Ligação Proteica , Transdução de Sinais , Fatores de Tempo , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas
16.
Oncogene ; 20(45): 6559-69, 2001 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-11641781

RESUMO

In this study, we report the cloning and characterization of a novel human Ste20-related kinase that we designated MST4. The 416 amino acid full-length MST4 contains an amino-terminal kinase domain, which is highly homologous to MST3 and SOK, and a unique carboxy-terminal domain. Northern blot analysis indicated that MST4 is highly expressed in placenta, thymus, and peripheral blood leukocytes. Wild-type but not kinase-dead MST4 can phosphorylate myelin basic protein in an in vitro kinase assay. MST4 specifically activates ERK but not JNK or p38 MAPK in transient transfected cells or in stable cell lines. Overexpression of dominant negative MEK1 or treatment with PD98059 abolishes MST4-induced ERK activity, whereas dominant-negative Ras or c-Raf-1 mutants failed to do so, indicating MST4 activates MEK1/ERK via a Ras/Raf-1 independent pathway. HeLa and Phoenix cell lines overexpressing wild-type, but not kinase-dead, MST4 exhibit increased growth rate and form aggressive soft-agar colonies. These phenotypes can be inhibited by PD98059. These results provide the first evidence that MST4 is biologically active in the activation of MEK/ERK pathway and in mediating cell growth and transformation.


Assuntos
Transformação Celular Neoplásica , Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/etiologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/fisiologia , Sequência de Aminoácidos , Divisão Celular , Clonagem Molecular , Humanos , MAP Quinase Quinase 1 , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Neoplasias/patologia , Filogenia , RNA Mensageiro/biossíntese , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Células Tumorais Cultivadas
17.
Proc Natl Acad Sci U S A ; 98(16): 9203-8, 2001 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-11470922

RESUMO

Marek's disease (MD) is a lymphoproliferative disease of chickens induced by a herpesvirus, the MD virus (MDV). Because MD is a significant economic problem to the poultry industry, there is great interest in enhancing genetic resistance, which is controlled by multiple genes. The influence of the MHC has been clearly demonstrated, and several relevant quantitative trait loci have been mapped; however, no single gene influencing MD resistance has been identified. Transcription of SORF2 is perturbed in the MDV recombinant clone RM1 due to a solo insertion of the reticuloendotheliosis virus long terminal repeat, which may explain the loss of oncogenicity for this strain. Hypothesizing that SORF2-interacting host proteins are involved in MD resistance, we screened a chicken splenic cDNA library by the yeast two-hybrid assay using SORF2 as bait. The chicken growth hormone (GH) structural peptide was identified, and the specific interaction was verified by coimmunoprecipitation. Immunohistochemical staining and indirect immunofluorescence assay indicated that GH and SORF2 can be coexpressed in MDV-infected cells both in vitro and in vivo. Furthermore, polymorphism in the GH gene (GH1) is associated with the number of tissues with tumors in commercial White Leghorn chickens with the MHC B*2/B*15 genotype. We conclude that GH1 may well be a MD resistance gene.


Assuntos
Hormônio do Crescimento/metabolismo , Herpesvirus Galináceo 2/metabolismo , Proteínas Virais/metabolismo , Animais , Galinhas , DNA Complementar , Técnica Indireta de Fluorescência para Anticorpo , Imuno-Histoquímica , Testes de Precipitina , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido
18.
Nat Cell Biol ; 3(5): 439-44, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11331870

RESUMO

Etk/BMX, a member of the Btk family of tyrosine kinases, is highly expressed in cells with great migratory potential, including endothelial cells and metastatic carcinoma cell lines. Here, we present evidence that Etk is involved in integrin signalling and promotes cell migration. The activation of Etk by extracellular matrix proteins is regulated by FAK through an interaction between the PH domain of Etk and the FERM domain of FAK. The lack of Etk activation by extracellular matrix in FAK-null cells could be restored by co-transfection with wild-type FAK. Disrupting the interaction between Etk and FAK diminished the cell migration promoted by either kinase. Furthermore, inhibiting Etk expression in metastatic carcinoma cell lines with an antisense oligonucleotide blocks integrin-mediated migration of these cells. Taken together, our data indicate the essential role of the interaction of the PH domain of Etk and the FERM domain of FAK in integrin signalling.


Assuntos
Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/metabolismo , Animais , Western Blotting , Células COS , Carcinoma/metabolismo , Linhagem Celular , Movimento Celular , Células Cultivadas , Endotélio Vascular/citologia , Ativação Enzimática , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Glutationa Transferase/metabolismo , Humanos , Microscopia de Fluorescência , Oligonucleotídeos/metabolismo , Fosforilação , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
19.
J Virol ; 75(11): 5159-73, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11333897

RESUMO

Chemokines induce chemotaxis, cell migration, and inflammatory responses. We report the identification of an interleukin-8 (IL-8) homolog, termed vIL-8, encoded within the genome of Marek's disease virus (MDV). The 134-amino-acid vIL-8 shares closest homology to mammalian and avian IL-8, molecules representing the prototype CXC chemokine. The gene for vIL-8 consists of three exons which map to the BamHI-L fragment within the repeats flanking the unique long region of the MDV genome. A 0.7-kb transcript encoding vIL-8 was detected in an n-butyrate-treated, MDV-transformed T-lymphoblastoid cell line, MSB-1. This induction is essentially abolished by cycloheximide and herpesvirus DNA polymerase inhibitor phosphonoacetate, indicating that vIL-8 is expressed with true late (gamma2) kinetics. Baculovirus-expressed vIL-8 was found to be secreted into the medium and shown to be functional as a chemoattractant for chicken peripheral blood mononuclear cells but not for heterophils. To characterize the function of vIL-8 with respect to MDV infection in vivo, a recombinant MDV was constructed with a deletion of all three exons and a soluble-modified green fluorescent protein (smGFP) expression cassette inserted at the site of deletion. In two in vivo experiments, the vIL-8 deletion mutant (RB1BvIL-8DeltasmGFP) showed a decreased level of lytic infection in comparison to its parent virus, an equal-passage-level parent virus, and to another recombinant MDV containing the insertion of a GFP expression cassette at the nonessential US2 gene. RB1BvIL-8DeltasmGFP retained oncogenicity, albeit at a greatly reduced level. Nonetheless, we have been able to establish a lymphoblastoid cell line from an RB1BvIL-8DeltasmGFP-induced ovarian lymphoma (MDCC-UA20) and verify the presence of a latent MDV genome lacking vIL-8. Taken together, these data describe the identification and characterization of a chemokine homolog encoded within the MDV genome that is dispensable for transformation but may affect the level of MDV in vivo lytic infection.


Assuntos
Fatores Quimiotáticos/genética , Herpesvirus Galináceo 2/imunologia , Interleucina-8/biossíntese , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Baculoviridae/genética , Sequência de Bases , Linhagem Celular , Linhagem Celular Transformada , Galinhas , Clonagem Molecular , Cicloeximida , Deleção de Genes , Proteínas de Fluorescência Verde , Herpesvirus Galináceo 2/genética , Interleucina-8/genética , Leucócitos Mononucleares/metabolismo , Proteínas Luminescentes/genética , Dados de Sequência Molecular , Mutagênese Insercional , Ácido Fosfonoacéticos , RNA Mensageiro/análise , Proteínas Recombinantes/biossíntese , Alinhamento de Sequência , Proteínas Virais/biossíntese , Proteínas Virais/genética , Replicação Viral
20.
J Biol Chem ; 276(31): 29403-9, 2001 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-11382770

RESUMO

Etk/Bmx, a member of the Tec family of nonreceptor protein-tyrosine kinases, is characterized by an N-terminal pleckstrin homology domain and has been shown to be a downstream effector of phosphatidylinositol 3-kinase. P21-activated kinase 1 (Pak1), another well characterized effector of phosphatidylinositol 3-kinase, has been implicated in the progression of breast cancer cells. In this study, we characterized the role of Etk in mammary development and tumorigenesis and explored the functional interactions between Etk and Pak1. We report that Etk expression is developmentally regulated in the mammary gland. Using transient transfection, coimmunoprecipitation and glutathione S-transferase-pull down assays, we showed that Etk directly associates with Pak1 via its N-terminal pleckstrin homology domain and also phosphorylates Pak1 on tyrosine residues. The expression of wild-type Etk in a non-invasive human breast cancer MCF-7 cells significantly increased proliferation and anchorage-independent growth of epithelial cancer cells. Conversely, expression of kinase-inactive mutant Etk-KQ suppressed the proliferation, anchorage-independent growth, and tumorigenicity of human breast cancer MDA-MB435 cells. These results indicate that Pak1 is a target of Etk and that Etk controls the proliferation as well as the anchorage-independent and tumorigenic growth of mammary epithelial cancer cells.


Assuntos
Neoplasias da Mama/patologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Animais , Apoptose , Neoplasias da Mama/enzimologia , Divisão Celular , Ativação Enzimática , Feminino , Humanos , Cinética , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/enzimologia , Camundongos , Camundongos Nus , Especificidade de Órgãos , Fosforilação , Fosfotirosina/metabolismo , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Transplante Heterólogo , Células Tumorais Cultivadas , Quinases Ativadas por p21
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...