Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cancer Res ; 5(4): 331-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17426248

RESUMO

Cancer is a highly heterogeneous disease in terms of the genetic profile and the response to therapeutics. An early identification of a genomic marker in drug discovery may help select patients that would respond to treatment in clinical trials. Here we suggest coupling compound screening with comparative genomic hybridization analysis of the model systems for early discovery of genomic biomarkers. A Bcl-2 antagonist, ABT-737, has recently been discovered and shown to induce regression of solid tumors, but its activity is limited to a fraction of small-cell lung carcinoma (SCLC) models tested. We used comparative genomic hybridization on high-density single-nucleotide polymorphism genotyping arrays to carry out a genome-wide analysis of 23 SCLC cell lines sensitive and resistant to ABT-737. The screen revealed a number of novel recurrent gene copy number abnormalities, which were also found in an independent data set of 19 SCLC tumors and confirmed by real-time quantitative PCR. A previously unknown amplification was identified on 18q and associated with the sensitivity of SCLC cell lines to ABT-737 and another Bcl-2 antagonist. The region of gain contains Bcl-2 and NOXA, two apoptosis-related genes. Expression microarray profiling showed that the genes residing in the amplified region of 18q are also overexpressed in the sensitive lines relative to the resistant lines. Fluorescence in situ hybridization analysis of tumors revealed that Bcl-2 gain is a frequent event in SCLC. Our findings suggest that 18q21-23 copy number will be a clinically relevant predictor for sensitivity of SCLC to Bcl-2 family inhibitors. The 18q21-23 genomic marker may have a broader application in cancer because Bcl-2 is associated with apoptosis evasion and chemoresistance.


Assuntos
Carcinoma de Células Pequenas/genética , Aberrações Cromossômicas , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Hibridização de Ácido Nucleico/métodos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Linhagem Celular Tumoral , Cromossomos Humanos Par 18 , Análise por Conglomerados , Genes Neoplásicos , Marcadores Genéticos , Genoma , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-bcl-2/genética
2.
Mol Cancer Ther ; 6(1): 253-61, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17237284

RESUMO

Patients with prostate cancer develop osteoblastic metastases when tumor cells arrive in the bone and stimulate osteoblasts by secreting growth-promoting factors. Endothelin 1 (ET-1) is believed to be a key factor in promoting osteoblastic metastasis. Selective blockade of the ET(A) receptor is an established strategy in the development of cancer therapeutics. However, the molecular mechanisms whereby prostate cancer promotes abnormal bone growth are not fully understood. In this study, we have applied genomic approaches to elucidate the molecular mechanism of stimulation of osteoblasts by ET-1. To examine the ET-1 axis, we generated genomic signatures for osteoblasts treated with ET-1, in the presence and absence of a selective ET(A) antagonist (ABT-627). The ET-1 signature was comprised of several motifs, such as osteoblastic differentiation, invasion, and suppression of apoptosis. The signature also pointed at possible activation of the calcineurin/NFAT pathway. We showed that ET-1 activates calcineurin and causes nuclear translocation of NFATc1, implicating the pathway in the ET-1-mediated stimulation of osteoblasts. We also showed that ET-1 inhibits apoptosis in osteoblasts, implying that the suppression of apoptosis may be an important factor in the promotion of osteoblastic growth by ET-1.


Assuntos
Calcineurina/metabolismo , Endotelinas/farmacologia , Genoma , Fatores de Transcrição NFATC/metabolismo , Osteoblastos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Anexina A5/metabolismo , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Endotelinas/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Genoma/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Osteoblastos/citologia , Transporte Proteico/efeitos dos fármacos
3.
Cancer Res ; 65(11): 4663-72, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15930284

RESUMO

Kringle 5 (K5) of human plasminogen has been shown to inhibit angiogenesis by inducing the apoptosis of proliferating endothelial cells. Peptide regions around the lysine-binding pocket of K5 largely mediate these effects, particularly the peptide PRKLYDY, which we show to compete with K5 for the binding to endothelial cells. The cell surface binding site for K5 that mediates these effects has not been defined previously. Here, we report that glucose-regulated protein 78, exposed on cell surfaces of proliferating endothelial cells as well as on stressed tumor cells, plays a key role in the antiangiogenic and antitumor activity of K5. We also report that recombinant K5-induced apoptosis of stressed HT1080 fibrosarcoma cells involves enhanced activity of caspase-7, consistent with the disruption of glucose-regulated protein 78-procaspase-7 complexes. These results establish recombinant K5 as an inhibitor of a stress response pathway, which leads to both endothelial and tumor cell apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Fragmentos de Peptídeos/farmacologia , Plasminogênio/farmacologia , Sequência de Aminoácidos , Inibidores da Angiogênese/metabolismo , Inibidores da Angiogênese/farmacologia , Apoptose/fisiologia , Sítios de Ligação , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Chaperona BiP do Retículo Endoplasmático , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/metabolismo , Fibrossarcoma/patologia , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/antagonistas & inibidores , Fragmentos de Peptídeos/metabolismo , Plasminogênio/antagonistas & inibidores , Plasminogênio/metabolismo , Ligação Proteica , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia
4.
Cancer Res ; 63(22): 7861-9, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14633714

RESUMO

Methionine aminopeptidase (MetAP)-2 has been suggested as a novel target for cancer therapy because the anticancer agent TNP-470 irreversibly inactivates the catalytic activity of this enzyme. However, the importance of MetAP2 in cell growth and tumor progression was uncertain because previous data were based on the chemically reactive TNP-470. Here we show that a rationally designed reversible MetAP2 inhibitor, A-357300, suppresses tumor growth preclinically without the toxicities observed with TNP-470. We have synthesized this bestatin-type MetAP2 inhibitor with the aid of crystal structures of the enzyme-inhibitor complexes and parallel synthesis. A-357300 induces cytostasis by cell cycle arrest at the G(1) phase selectively in endothelial cells and in a subset of tumor cells, but not in most primary cells of nonendothelial type. A-357300 inhibits angiogenesis both in vitro and in vivo and shows potent antitumor efficacy in carcinoma, sarcoma, and neuroblastoma murine models. These data affirm that MetAP2 plays a pivotal role in cell growth and establish that reversible MetAP2 inhibitors are promising novel cancer therapeutic agents.


Assuntos
Aminopeptidases/antagonistas & inibidores , Antineoplásicos/farmacologia , Clorobenzenos/farmacologia , Metaloendopeptidases/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/toxicidade , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Clorobenzenos/química , Clorobenzenos/toxicidade , Córnea/irrigação sanguínea , Cicloexanos , Desenho de Fármacos , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Feminino , Fibrossarcoma/tratamento farmacológico , Fibrossarcoma/enzimologia , Humanos , Camundongos , Camundongos SCID , Modelos Moleculares , Neovascularização Fisiológica/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/enzimologia , O-(Cloroacetilcarbamoil)fumagilol , Inibidores de Proteases/química , Inibidores de Proteases/toxicidade , Sesquiterpenos/química , Sesquiterpenos/farmacologia , Sesquiterpenos/toxicidade , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Mol Cancer Ther ; 9(11): 2943-50, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20829195

RESUMO

Evasion of apoptosis is a known feature of cancer cells. One mechanism of deregulating the apoptotic pathway is through overexpression of antiapoptotic BCL2 family members. ABT-263 (navitoclax) is a first-in-class BCL2 family inhibitor that restores the ability of cancer cells to undergo apoptosis. However, many cancer cells are resistant to ABT-263 due to high levels of a BCL2 family member, MCL1, which is not targeted by the drug. MCL1 expression is regulated transcriptionally, translationally, and through proteasome-mediated degradation. Recently, MCL1 expression was shown to be affected by microRNAs (miRNA). To identify miRNAs that modulate the sensitivity of cancer cells to ABT-263, we screened a library of 810 human miRNA mimics in HCT-116 cells in the presence of ABT-263. The screen revealed 19 miRNAs that sensitize HCT-116 cells to ABT-263. Fifteen of these miRNAs were also shown to sensitize CHL1 melanoma cells to the same agent. We further evaluated 12 of the strongest sensitizers in these cell lines. We found that these sensitizers induced apoptosis only in the presence of ABT-263. In addition, whereas all 12 of these miRNAs reduced MCL1 protein expression, only 10 of them targeted MCL1 through direct binding to the 3'-untranslated region of the gene, raising the possibility that other resistance regulators of MCL1 expression may be identified using our method. Finally, because sensitizing miRNA expression is lower in tumors compared with normal tissues, our data can facilitate the design of miRNA replacement therapies to increase sensitivity to BCL2 antagonists.


Assuntos
Compostos de Anilina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , MicroRNAs/análise , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Sulfonamidas/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Ensaios de Triagem em Larga Escala , Humanos , MicroRNAs/genética , Modelos Biológicos , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Interferente Pequeno/farmacologia
6.
Eur J Cancer ; 46(5): 966-73, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20061137

RESUMO

The blood protein plasminogen is proteolytically cleaved to produce angiostatin and kringle 5 (K5), both of which are known angiogenesis inhibitors. A common structural element between K5, angiostatin and other endogenous angiogenesis inhibitors is the presence of the kringle protein-interacting domain. Another kringle domain-containing protein, hepatocyte growth factor (HGF), promotes angiogenesis by binding to and stimulating the tyrosine kinase receptor Met. HGF binding to Met is dependent on the kringle domains of HGF. Because both K5 and HGF contain kringle motifs and because these proteins have opposite effects on angiogenesis, we hypothesised that K5 can antagonise HGF-mediated signalling in a Met-dependent manner. We determined that K5 binding to H1299 cells is competed by HGF suggesting that these two proteins bind to the same protein. Purified K5 immunoprecipitates with Met and this interaction is abolished by increasing doses of HGF. Using proliferation, phosphorylation of Met and Akt as markers of HGF activity, we determined that K5 inhibits HGF-mediated signalling. Taken together, these data support a model by which K5 binds to Met and functions as a competitive antagonist of HGF signalling and presents a novel mechanism of action of K5.


Assuntos
Inibidores da Angiogênese/farmacologia , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fragmentos de Peptídeos/farmacologia , Plasminogênio/farmacologia , Animais , Humanos , Pichia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Coelhos , Proteínas Recombinantes/farmacologia
7.
J Proteome Res ; 7(11): 4807-20, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18828628

RESUMO

The catalytic activity of methionine aminopeptidase-2 (MetAP2) has been pharmacologically linked to cell growth, angiogenesis, and tumor progression, making this an attractive target for cancer therapy. An assay for monitoring specific protein changes in response to MetAP2 inhibition, allowing pharmacokinetic (PK)/pharmacodynamic (PD) models to be established, could dramatically improve clinical decision-making. Candidate MetAP2-specific protein substrates were discovered from undigested cell culture-derived proteomes by MALDI-/SELDI-MS profiling and a biochemical method using (35)S-Met labeled protein lysates. Substrates were identified either as intact proteins by FT-ICR-MS or applying in-gel protease digestions followed by LC-MS/MS. The combination of these approaches led to the discovery of novel MetAP2-specific substrates including thioredoxin-1 (Trx-1), SH3 binding glutamic acid rich-like protein (SH3BGRL), and eukaryotic elongation factor-2 (eEF2). These studies also confirmed glyceraldehye 3-phosphate dehydrogenase (GAPDH) and cyclophillin A (CypA) as MetAP2 substrates. Additional data in support of these proteins as MetAP2-specific substrates were provided by in vitro MetAP1/MetAP2 enzyme assays with the corresponding N-terminal derived peptides and 1D/2D Western analyses of cellular and tissue lysates. FT-ICR-MS characterization of all intact species of the 18 kDa substrate, CypA, enabled a SELDI-MS cell-based assay to be developed for correlating N-terminal processing and inhibition of proliferation. The MetAP2-specific protein substrates discovered in this study have diverse properties that should facilitate the development of reagents for testing in preclinical and clinical environments.


Assuntos
Aminopeptidases/antagonistas & inibidores , Biomarcadores Tumorais/metabolismo , Metaloendopeptidases/antagonistas & inibidores , Inibidores de Proteases/farmacologia , Animais , Biomarcadores Tumorais/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Células K562 , Leucemia Eritroblástica Aguda/patologia , Camundongos , Peso Molecular , Inibidores de Proteases/classificação , Proteômica/métodos , Fatores de Tempo
8.
Anticancer Drugs ; 14(1): 13-9, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12544254

RESUMO

Many conventional chemotherapeutics, such as the microtubule-stabilizing anticancer drug paclitaxel (Taxol), have been shown to have anti-angiogenic activity and clinical application of a continuous low dose of these agents has been suggested for cancer therapy. In this study, we show that paclitaxel selectively inhibits the proliferation of human endothelial cells (ECs) at ultra low concentrations (0.1-100 pM), with an IC50 = 0.1 pM, while it inhibits non-endothelial type human cells at 10(4) - to 10(5) -fold higher concentrations, with IC50 = 1-10 nM. The selectivity of paclitaxel inhibition of cell proliferation is also species specific, as mouse ECs are not sensitive to paclitaxel at ultra low concentrations. They are inhibited by higher concentrations of paclitaxel with IC50 = 1-10 nM. Inhibition of human ECs by paclitaxel at ultra low concentrations does not affect the cellular microtubule structure, and the treated cells do not show G2/M cell cycle arrest and apoptosis, suggesting a novel but as yet unidentified mechanism of action. In an in vitro angiogenesis assay, paclitaxel at ultra low concentrations blocks human ECs from forming sprouts and tubes in the three-dimensional fibrin matrix. In summary, paclitaxel selectively inhibits human EC proliferation and in vitro angiogenesis at low picomolar concentrations. The data support a clinical application of continuous ultra-low-dose paclitaxel to treat cancer.


Assuntos
Microtúbulos/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Paclitaxel/administração & dosagem , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular , Relação Dose-Resposta a Droga , Inibidores do Crescimento/administração & dosagem , Humanos , Camundongos , Microtúbulos/fisiologia , Neovascularização Fisiológica/fisiologia
9.
Biochemistry ; 42(17): 5035-42, 2003 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-12718546

RESUMO

The identity of the physiological metal cofactor for human methionine aminopeptidase-2 (MetAP2) has not been established. To examine this question, we first investigated the effect of eight divalent metal ions, including Ca(2+), Co(2+), Cu(2+), Fe(2+), Mg(2+), Mn(2+), Ni(2+), and Zn(2+), on recombinant human methionine aminopeptidase apoenzymes in releasing N-terminal methionine from three peptide substrates: MAS, MGAQFSKT, and (3)H-MASK(biotin)G. The activity of MetAP2 on either MAS or MGAQFSKT was enhanced 15-25-fold by Co(2+) or Mn(2+) metal ions in a broad concentration range (1-1000 microM). In the presence of reduced glutathione to mimic the cellular environment, Co(2+) and Mn(2+) were also the best stimulators (approximately 30-fold) for MetAP2 enzyme activity. To determine which metal ion is physiologically relevant, we then tested inhibition of intracellular MetAP2 with synthetic inhibitors selective for MetAP2 with different metal cofactors. A-310840 below 10 microM did not inhibit the activity of MetAP2-Mn(2+) but was very potent against MetAP2 with other metal ions including Co(2+), Fe(2+), Ni(2+), and Zn(2+) in the in vitro enzyme assays. In contrast, A-311263 inhibited MetAP2 with Mn(2+), as well as Co(2+), Fe(2+), Ni(2+), and Zn(2+). In cell culture assays, A-310840 did not inhibit intracellular MetAP2 enzyme activity and did not inhibit cell proliferation despite its ability to permeate and accumulate in cytosol, while A-311263 inhibited both intracellular MetAP2 and proliferation in a similar concentration range, indicating cellular MetAP2 is functioning as a manganese enzyme but not as a cobalt, zinc, iron, or nickel enzyme. We conclude that MetAP2 is a manganese enzyme and that therapeutic MetAP2 inhibitors should inhibit MetAP2-Mn(2+).


Assuntos
Aminopeptidases/metabolismo , Manganês/fisiologia , Metaloendopeptidases/metabolismo , Sequência de Aminoácidos , Aminopeptidases/química , Aminopeptidases/genética , Apoenzimas/efeitos dos fármacos , Apoenzimas/metabolismo , Sequência de Bases , Cátions Bivalentes/farmacologia , Primers do DNA , Humanos , Cinética , Metaloendopeptidases/química , Metaloendopeptidases/genética , Dados de Sequência Molecular , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA