Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Mol Cancer Ther ; 14(2): 326-42, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25512618

RESUMO

Most cancer therapies involve a component of treatment that inflicts DNA damage in tumor cells, such as double-strand breaks (DSBs), which are considered the most serious threat to genomic integrity. Complex systems have evolved to repair these lesions, and successful DSB repair is essential for tumor cell survival after exposure to ionizing radiation (IR) and other DNA-damaging agents. As such, inhibition of DNA repair is a potentially efficacious strategy for chemo- and radiosensitization. Homologous recombination (HR) and nonhomologous end-joining (NHEJ) represent the two major pathways by which DSBs are repaired in mammalian cells. Here, we report the design and execution of a high-throughput, cell-based small molecule screen for novel DSB repair inhibitors. We miniaturized our recently developed dual NHEJ and HR reporter system into a 384-well plate-based format and interrogated a diverse library of 20,000 compounds for molecules that selectively modulate NHEJ and HR repair in tumor cells. We identified a collection of novel hits that potently inhibit DSB repair, and we have validated their functional activity in a comprehensive panel of orthogonal secondary assays. A selection of these inhibitors was found to radiosensitize cancer cell lines in vitro, which suggests that they may be useful as novel chemo- and radio sensitizers. Surprisingly, we identified several FDA-approved drugs, including the calcium channel blocker mibefradil dihydrochloride, that demonstrated activity as DSB repair inhibitors and radiosensitizers. These findings suggest the possibility for repurposing them as tumor cell radiosensitizers in the future. Accordingly, we recently initiated a phase I clinical trial testing mibefradil as a glioma radiosensitizer.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Ensaios de Triagem em Larga Escala/métodos , Radiossensibilizantes/farmacologia , Linhagem Celular Tumoral , Proteínas de Fluorescência Verde/metabolismo , Recombinação Homóloga/efeitos dos fármacos , Humanos , Projetos Piloto , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas/farmacologia
3.
Mol Cancer ; 13: 45, 2014 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-24588908

RESUMO

BACKGROUND: Gain of function mutations in B-RAF and N-RAS occur frequently in melanoma, leading to mitogen activating protein kinase (MAPK) pathway activation, and this pathway is the target of drugs in development. Our purpose was to study clinical characteristics of patients with mutations in this pathway and to determine activity of inhibitors of B-RAF and MEK in short term cultures grown from tumors of some of these patients. METHODS: Clinical and pathologic data were collected retrospectively on melanoma patients tested for B-RAF and N-RAS mutations at the Yale Cancer Center and associations with survival were determined. We studied in vitro activity of the pan-RAF inhibitor, RAF265, and the MEK inhibitor, MEK162, in 22 melanoma short term cultures. We further characterized the effect of MEK inhibition on apoptosis and growth of melanoma cultures. RESULTS: In a cohort of 144 metastatic melanoma patients we found that patients with N-RAS mutant melanoma had a worse prognosis. These patients were more likely to have brain metastases at the time of presentation with metastatic disease than their N-RAS-wild-type counterparts. All N-RAS mutant melanoma cultures tested in our study (n = 7) were sensitive to MEK inhibition 162. Exposure to MEK162 reduced ERK1/2 phosphorylation, and induced apoptosis. Clonogenic survival was significantly reduced in sensitive melanoma cell cultures. CONCLUSIONS: The prognosis of patients with melanoma expressing constitutively active N-RAS is poor, consistent with studies performed at other institutions. N-RAS mutant melanoma cultures appear to be particularly sensitive to MEK162, supporting ongoing clinical trials with MEK162 in N-RAS mutated melanoma.


Assuntos
Genes ras/genética , MAP Quinase Quinase Quinases/metabolismo , Melanoma/genética , Melanoma/metabolismo , Idoso , Benzimidazóis/farmacologia , Western Blotting , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Melanoma/mortalidade , Pessoa de Meia-Idade , Mutação , Metástase Neoplásica , Modelos de Riscos Proporcionais , Inibidores de Proteínas Quinases/farmacologia , Células Tumorais Cultivadas
4.
Mol Cell Biol ; 34(6): 1046-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24396066

RESUMO

The simultaneous interaction of poly(A)-binding protein (PABP) with eukaryotic translation initiation factor 4G (eIF4G) and the mRNA 3' poly(A) tail promotes translation initiation. We previously showed that the interaction of PABP-interacting protein 1 (Paip1) with PABP and eukaryotic translation initiation factor 3 (eIF3; via the eIF3g subunit) further stimulates translation. Here, we demonstrate that the interaction of eIF3 with Paip1 is regulated by amino acids through the mTORC1 signaling pathway. The Paip1-eIF3 interaction is impaired by the mTORC1 inhibitors, rapamycin and PP242. We show that ribosomal protein S6 kinases 1 and 2 (S6K1/2) promote the interaction of eIF3 with Paip1. The enhancement of Paip1-eIF3 interaction by amino acids is abrogated by an S6K inhibitor or shRNA against S6K1/2. S6K1 interacts with eIF3f and, in vitro, phosphorylates eIF3. Finally, we show that S6K inhibition leads to a reduction in translation by Paip1. We propose that S6K1/2 phosphorylate eIF3 to stimulate Paip1-eIF3 interaction and consequent translation initiation. Taken together, these data demonstrate that eIF3 is a new translation target of the mTOR/S6K pathway.


Assuntos
Fator de Iniciação 3 em Eucariotos/genética , Fator de Iniciação 3 em Eucariotos/metabolismo , Fatores de Iniciação de Peptídeos/genética , Fatores de Iniciação de Peptídeos/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Aminoácidos/genética , Aminoácidos/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Células HeLa , Humanos , Fosforilação/genética , Ligação Proteica/genética , Domínios e Motivos de Interação entre Proteínas/genética , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/antagonistas & inibidores
6.
Adv Pharmacol ; 65: 1-26, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22959021

RESUMO

Malignant cells arise from particular mutations in genes controlling cell proliferation, invasion, and survival. Older antineoplastic drugs were designed to target vital cellular processes, such as DNA maintenance and repair and cell division. As a result, these drugs can affect all proliferating cells and are associated with unavoidable toxicities. Recent discoveries in cancer research have identified "driver" mutations in some types of cancer, and efforts have been undertaken to develop drugs targeting these oncogenes. In most cases, due to escape mechanisms and adaptive responses, single oncogene targeting is insufficient to induce prolonged responses in solid tumors. Drug combinations are therefore used to enhance the growth inhibitory and cytotoxic effects of the targeted therapies. Depending on the position of additional targets within the signaling network, drug combinations may target either different signaling pathways (parallel targeting) or the same pathway at several fragile nodes (vertical targeting). In this review, we discuss strategies of multitarget inhibition with a focus on vertical signaling pathway targeting.


Assuntos
Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Humanos , Mitógenos/farmacologia , Mitógenos/uso terapêutico , Neoplasias/genética , Oncogenes/genética
7.
Nat Rev Mol Cell Biol ; 12(4): 235-45, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21427765

RESUMO

The translation initiation step in eukaryotes is highly regulated and rate-limiting. During this process, the 40S ribosomal subunit is usually recruited to the 5' terminus of the mRNA. It then migrates towards the initiation codon, where it is joined by the 60S ribosomal subunit to form the 80S initiation complex. Secondary structures in the 5' untranslated region (UTR) can impede binding and movement of the 40S ribosome. The canonical eukaryotic translation initiation factor eIF4A (also known as DDX2), together with its accessory proteins eIF4B and eIF4H, is thought to act as a helicase that unwinds secondary structures in the mRNA 5' UTR. Growing evidence suggests that other helicases are also important for translation initiation and may promote the scanning processivity of the 40S subunit, synergize with eIF4A to 'melt' secondary structures or facilitate translation of a subset of mRNAs.


Assuntos
Códon de Iniciação/genética , Biossíntese de Proteínas/genética , RNA Helicases/metabolismo , RNA Mensageiro/genética , Animais , Fator de Iniciação 4A em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Humanos , Modelos Genéticos , RNA Mensageiro/metabolismo
8.
Cell Cycle ; 9(20): 4106-9, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20948310

RESUMO

Messenger RNA translation or protein synthesis, is a fundamental biological process affecting cell growth, survival and proliferation. Initiation is the rate limiting and hence the most regulated step of translation. In eukaryotes, translation initiation is facilitated by multiple protein factors collectively called eIFs (for eukaryotic translation initiation factors). The complex consisting of the eIF4 group factors including the mRNA cap-binding eIF4E protein, large scaffolding protein eIF4G and RNA helicase eIF4A is assisted by the eIF4B co-factor to unwind local secondary structures and create a ribosome landing pad on mRNA. Recruitment of the ribosome and augmentation in the mRNA scanning process culminates in the positioning of the ribosome over the start codon. Deregulated translational control is believed to play an important role in oncogenic transformation. Indeed, many eIFs are bona fide proto-oncogenes. In many types of human cancers, eIFs are either overexpressed or ectopically activated by Ras-MAPK and PI3K-mTOR signaling cascades, resulting in increased survival and accelerated proliferation. In this review we will analyze the bulk of data describing eIF4B and its role in cell survival and proliferation. Recent studies have shown that eIF4B is phosphorylated and activated by Ras-MAPK and PI3K-mTOR signaling cascades. In addition, eIF4B regulates translation of proliferative and pro-survival mRNAs. Moreover, eIF4B depletion in cancer cells attenuates proliferation, sensitizes them to genotoxic stress-driven apoptosis. Taken together, these findings identify eIF4B as a potential target for development of anti-cancer therapies.


Assuntos
Proliferação de Células , Sobrevivência Celular/fisiologia , Fatores de Iniciação em Eucariotos/metabolismo , Transdução de Sinais/fisiologia , Animais , Fatores de Iniciação em Eucariotos/genética , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas ras/metabolismo
9.
Mol Cell Biol ; 30(6): 1478-85, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20086100

RESUMO

Translation initiation plays an important role in cell growth, proliferation, and survival. The translation initiation factor eIF4B (eukaryotic initiation factor 4B) stimulates the RNA helicase activity of eIF4A in unwinding secondary structures in the 5' untranslated region (5'UTR) of the mRNA in vitro. Here, we studied the effects of eIF4B depletion in cells using RNA interference (RNAi). In agreement with the role of eIF4B in translation initiation, its depletion resulted in inhibition of this step. Selective reduction of translation was observed for mRNAs harboring strong to moderate secondary structures in their 5'UTRs. These mRNAs encode proteins, which function in cell proliferation (Cdc25C, c-myc, and ODC [ornithine decarboxylase]) and survival (Bcl-2 and XIAP [X-linked inhibitor of apoptosis]). Furthermore, eIF4B silencing led to decreased proliferation rates, promoted caspase-dependent apoptosis, and further sensitized cells to camptothecin-induced cell death. These results demonstrate that eIF4B is required for cell proliferation and survival by regulating the translation of proliferative and prosurvival mRNAs.


Assuntos
Fatores de Iniciação em Eucariotos/metabolismo , Regiões 5' não Traduzidas , Apoptose/efeitos dos fármacos , Camptotecina/farmacologia , Caspases/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Células HeLa , Humanos , Biossíntese de Proteínas/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
10.
Proc Natl Acad Sci U S A ; 106(52): 22217-22, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-20018725

RESUMO

Translational control plays an important role in cell growth and tumorigenesis. Cap-dependent translation initiation of mammalian mRNAs with structured 5'UTRs requires the DExH-box protein, DHX29, in vitro. Here we show that DHX29 is important for translation in vivo. Down-regulation of DHX29 leads to impaired translation, resulting in disassembly of polysomes and accumulation of mRNA-free 80S monomers. DHX29 depletion also impedes cancer cell growth in culture and in xenografts. Thus, DHX29 is a bona fide translation initiation factor that potentially can be exploited as a target to inhibit cancer cell growth.


Assuntos
Proliferação de Células , Neoplasias/etiologia , Iniciação Traducional da Cadeia Peptídica/fisiologia , RNA Helicases/metabolismo , Regiões 5' não Traduzidas , Animais , Regulação para Baixo , Células HeLa , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , RNA Helicases/antagonistas & inibidores , RNA Helicases/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Subunidades Ribossômicas Menores de Eucariotos/metabolismo , Transplante Heterólogo
11.
Cancer Cell ; 16(5): 439-46, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19878875

RESUMO

eIF4E, the mRNA 5' cap-binding translation initiation factor, is overexpressed in numerous cancers and is implicated in mechanisms underlying oncogenesis and senescence. 4E-BPs (eIF4E-binding proteins) inhibit eIF4E activity, and thereby act as suppressors of eIF4E-dependent pathways. Here, we show that tumorigenesis is increased in p53 knockout mice that lack 4E-BP1 and 4E-BP2. However, primary fibroblasts lacking 4E-BPs, but expressing p53, undergo premature senescence and resist oncogene-driven transformation. Thus, the p53 status governs 4E-BP-dependent senescence and transformation. Intriguingly, the 4E-BPs engage in senescence via translational control of the p53-stabilizing protein, Gas2. Our data demonstrate a role for 4E-BPs in senescence and tumorigenesis and highlight a p53-mediated mechanism of senescence through a 4E-BP-dependent pathway.


Assuntos
Transformação Celular Neoplásica/genética , Fator de Iniciação 4E em Eucariotos/genética , Proteína Supressora de Tumor p53/genética , Animais , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Senescência Celular/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Camundongos , Camundongos Knockout , Proteína Supressora de Tumor p53/metabolismo
12.
J Biol Chem ; 282(19): 14056-64, 2007 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-17360704

RESUMO

Converging signals from the mammalian target of rapamycin (mTOR) and phosphoinositide 3-kinase (PI3K) pathways are well established to modulate translation initiation. Less is known regarding the molecular basis of protein synthesis regulated by other inputs, such as agonists of the Ras/extracellular signal-regulated kinase (ERK) signaling cascade. Ribosomal protein (rp) S6 is a component of the 40S ribosomal subunit that becomes phosphorylated at several serine residues upon mitogen stimulation, but the exact molecular mechanisms regulating its phosphorylation and the function of phosphorylated rpS6 is poorly understood. Here, we provide evidence that activation of the p90 ribosomal S6 kinases (RSKs) by serum, growth factors, tumor promoting phorbol esters, and oncogenic Ras is required for rpS6 phosphorylation downstream of the Ras/ERK signaling cascade. We demonstrate that while ribosomal S6 kinase 1 (S6K1) phosphorylates rpS6 at all sites, RSK exclusively phosphorylates rpS6 at Ser(235/236) in vitro and in vivo using an mTOR-independent mechanism. Mutation of rpS6 at Ser(235/236) reveals that phosphorylation of these sites promotes its recruitment to the 7-methylguanosine cap complex, suggesting that Ras/ERK signaling regulates assembly of the translation preinitiation complex. These data demonstrate that RSK provides an mTOR-independent pathway linking the Ras/ERK signaling cascade to the translational machinery.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína S6 Ribossômica/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Proteínas ras/metabolismo , Células Cultivadas , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Rim/metabolismo , Luciferases/metabolismo , MAP Quinase Quinase Quinases , Mutação , Fosforilação , Polirribossomos/metabolismo , Biossíntese de Proteínas , RNA Interferente Pequeno/farmacologia , Proteína S6 Ribossômica/antagonistas & inibidores , Proteína S6 Ribossômica/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Ribossomos/metabolismo , Transdução de Sinais , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética
13.
EMBO J ; 25(12): 2781-91, 2006 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-16763566

RESUMO

The eukaryotic translation initiation factor 4B (eIF4B) plays a critical role in recruiting the 40S ribosomal subunit to the mRNA. In response to insulin, eIF4B is phosphorylated on Ser422 by S6K in a rapamycin-sensitive manner. Here we demonstrate that the p90 ribosomal protein S6 kinase (RSK) phosphorylates eIF4B on the same residue. The relative contribution of the RSK and S6K modules to the phosphorylation of eIF4B is growth factor-dependent, and the two phosphorylation events exhibit very different kinetics. The S6K and RSK proteins are members of the AGC protein kinase family, and require PDK1 phosphorylation for activation. Consistent with this requirement, phosphorylation of eIF4B Ser422 is abrogated in PDK1 null embryonic stem cells. Phosphorylation of eIF4B on Ser422 by RSK and S6K is physiologically significant, as it increases the interaction of eIF4B with the eukaryotic translation initiation factor 3.


Assuntos
Fatores de Iniciação em Eucariotos/metabolismo , Sistema de Sinalização das MAP Quinases , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases/metabolismo , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Sequência de Aminoácidos , Animais , Catálise , Fator de Iniciação 3 em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos/química , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação/genética , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Quinases S6 Ribossômicas 90-kDa/química , Proteínas Quinases S6 Ribossômicas 90-kDa/deficiência , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
14.
EMBO J ; 23(8): 1761-9, 2004 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-15071500

RESUMO

The eucaryotic translation initiation factor 4B (eIF4B) stimulates the helicase activity of the DEAD box protein eIF4A to unwind inhibitory secondary structure in the 5' untranslated region of eucaryotic mRNAs. Here, using phosphopeptide mapping and a phosphospecific antiserum, we identify a serum-responsive eIF4B phosphorylation site, Ser422, located in an RNA-binding region required for eIF4A helicase-promoting activity. Ser422 phosphorylation appears to be regulated by the S6Ks: (a) Ser422 phosphorylation is sensitive to pharmacological inhibitors of phosphoinositide-3 kinase and the mammalian target of rapamycin; (b) S6K1/S6K2 specifically phosphorylate Ser422 in vitro; and (c) rapamycin-resistant S6Ks confer rapamycin resistance upon Ser422 phosphorylation in vivo. Substitution of Ser422 with Ala results in a loss of activity in an in vivo translation assay, indicating that phosphorylation of this site plays an important role in eIF4B function. We therefore propose that eIF4B may mediate some of the effects of the S6Ks on translation.


Assuntos
Fatores de Iniciação em Eucariotos/metabolismo , Fosfosserina/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Linhagem Celular , Resistência a Medicamentos , Fatores de Iniciação em Eucariotos/genética , Humanos , Mutação/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Quinases/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR
15.
Endocrinology ; 145(5): 2228-44, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-14736735

RESUMO

The role of ERK, Jun N-terminal kinase (JNK), p38, and c-Src in GnRH-stimulated FSHbeta-subunit promoter activity was examined in the LbetaT-2 gonadotroph cell line. Incubation of the cells with a GnRH agonist resulted in activation of ERK, JNK, p38, and c-Src. The peak of ERK activation was observed at 5 min, whereas that of JNK, p38, and c-Src at 30 min, declining thereafter. ERK activation by GnRH is dependent on protein kinase C (PKC), as evident by activation, inhibition, and depletion of 12-O-tetradecanoylphorbol-13-acetate-sensitive PKC subspecies. Ca(2+) influx, but not Ca(2+) mobilization, is required for ERK activation. GnRH signaling to ERK is partially mediated by dynamin and a protein tyrosine kinase, apparently c-Src. ERK activation by GnRH in LbetaT-2 cells does not involve transactivation of epidermal growth factor receptor or mediation via Gbetagamma or beta-arrestin. Once activated by GnRH, ERK translocates to the nucleus. We examined the role of ERK, JNK, p38, and c-Src in GnRH-stimulated ovine FSHbeta promoter, linked to a luciferase reporter gene (-4741oFSHbeta-LUC). The PKC activator 12-O-tetradecanoylphorbol-13-acetate, but not the Ca(2+) ionophore ionomycin, stimulated FSHbeta-luciferase (LUC) activity. Furthermore, down-regulation of PKC, but not removal of Ca(2+), inhibited the GnRH response. Cotransfection of FSHbeta-LUC and the constitutively active forms of Raf-1 and MEK stimulated FSHbeta-LUC activity, whereas the dominant negatives of Ras, Raf-1, and MEK and the selective MEK inhibitor PD98059, abolished GnRH-induced FSHbeta-LUC activity. The dominant negatives of CDC42 and JNK reduced the GnRH response by 36 and 49%, respectively. Incubation of the cells with the p38 or the c-Src inhibitors SB203580 and PP1 also reduced the GnRH response. Surprisingly, two proximal activator protein-1 sites contribute very little to the GnRH response. Thus, PKC, ERK, JNK, p38, and c-Src, but not Ca(2+), are involved in GnRH induction of the ovine FSHbeta gene.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/genética , Hormônio Liberador de Gonadotropina/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Busserrelina/farmacologia , Proteína Tirosina Quinase CSK , Cálcio/fisiologia , Linhagem Celular , Núcleo Celular/enzimologia , Dinaminas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ionomicina/farmacologia , Ionóforos/farmacologia , MAP Quinase Quinase 4 , Regiões Promotoras Genéticas/genética , Proteína Quinase C/metabolismo , Ovinos , Acetato de Tetradecanoilforbol/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno , Quinases da Família src
16.
Cancer Res ; 62(4): 1093-102, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11861388

RESUMO

Breast cancer progression may be affected by various cellular components expressed by the tumor cells and/or by microenvironmental factors. Many studies report the correlation between breast cancer progression and monocyte infiltration into the tumor site. We have identified recently the CC chemokine regulated on activation, normal T cell expressed and secreted (RANTES), a major monocyte chemoattractant expressed by breast tumor cells, as a potential contributor to breast cancer progression. In the present study, analysis of the regulation of RANTES expression demonstrates that the expression of RANTES in breast tumor cells is elevated significantly and in a synergistic manner by IFN-gamma and tumor necrosis factor-alpha. Identification of the mechanisms by which RANTES may contribute to breast cancer progression included the analysis of the potential ability of RANTES to act in paracrine and indirect mechanisms, as well as directly on the tumor cells, to promote disease progression. Our results suggest that breast tumor cell-derived RANTES may promote breast cancer progression by its partial contribution to monocyte migration into breast tumor sites. Moreover, RANTES promotes the expression of matrix metalloproteinase (MMP) 9 by THP-1 monocytic cells and elevates vascularity in chick chorioallantoic membrane assays. Tumor necrosis factor-alpha, a major monocyte-derived cytokine, was found to promote the expression of MMP9 and MMP2 by MCF-7 and T47D breast adenocarcinoma cells, respectively, and to induce the de novo expression of an additional proteolytic enzyme by T47D cells, presumably MMP9. The possibility that RANTES may act directly on breast tumor cells was supported by detection of the expression of the CCR5 RANTES receptor in biopsy sections of breast cancer patients and by the ability of RANTES to promote the expression of MMP9 by MCF-7 cells. In all, our study suggests that the expression of RANTES by breast tumor cells results not only in monocyte migration to the tumor site but also in protumorigenic activities of RANTES and of proinflammatory cytokines that may facilitate metastasis formation and contribute to disease progression.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Quimiocina CCL5/fisiologia , Animais , Neoplasias da Mama/enzimologia , Carcinoma Ductal de Mama/enzimologia , Quimiocina CCL5/biossíntese , Embrião de Galinha , Citocinas/fisiologia , Progressão da Doença , Humanos , Metaloproteinase 9 da Matriz/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...