Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Oncol Rep ; 47(1)2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34738625

RESUMO

The tumor blood vessel endothelium forms a barrier that must be crossed by circulating immune cells in order for them to reach and kill cancer cells. Epidermal growth factor­like domain 7 (Egfl7) represses this immune infiltration by lowering the expression levels of leukocyte adhesion receptors on the surface of endothelial cells. However, the protein domains involved in these properties are not completely understood. Egfl7 is structurally composed of the predicted EMI­, EGF­ and C­terminal domains. The present study aimed to investigate the roles of these different domains in tumor development by designing retroviruses coding for deletion mutants and then infecting 4T1 breast cancer cell populations, which consequently overexpressed the variants. By performing in vitro soft­agar assays, it was found that Egfl7 and its deletion variants did not affect cell proliferation or anchorage­independent growth. When 4T1 cells expressing either the wild­type Egfl7 protein or Egfl7 domain variants were implanted in mice, Egfl7 expression markedly promoted tumor development and deletion of the EGF repeats decreased the tumor growth rate. By contrast, deleting any other domain displayed no significant effect on tumor development. The overexpression of Egfl7 also decreased T cell and natural killer cell infiltration in tumors, as determined by immunofluorescence staining of tumor sections, whereas deletion of the EGF repeats inhibited this effect. Reverse transcription­quantitative PCR analysis of the mechanisms involved revealed that deleting the EGF repeats partially restored the expression levels of vascular cell adhesion molecule 1 and E­selectin, which were suppressed by overexpression of Egfl7 in endothelial cells in vitro. This resulted in a higher number of lymphocytes bound to HUVEC expressing Egfl7­ΔEGF compared with HUVEC expressing wild­type Egfl7, as assessed by fluorescent­THP­1 adhesion assays onto endothelial cells. Overall, the present study demonstrated that the EGF repeats may participate in the protumoral and anti­inflammatory effects of Egfl7.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Família de Proteínas EGF/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Sistema Imunitário/metabolismo , Evasão Tumoral/genética , Animais , Proteínas de Ligação ao Cálcio/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Regulação para Baixo , Família de Proteínas EGF/genética , Feminino , Camundongos , Camundongos Endogâmicos BALB C
2.
Oncol Rep ; 20(5): 1283-7, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18949434

RESUMO

Bortezomib and other proteasome inhibitors have demonstrated an interesting antitumor activity against glioma cell lines. The present study aimed to evaluate the cytotoxic potential of bortezomib in vivo on two human malignant glioma xenografts using doses relevant to clinical practice. The TCG3 and U87 malignant glioma xenografts were heterotopically implanted onto nude mice. Bortezomib effects were evaluated using the three different doses of 0.25, 0.45 and 0.90 mg/kg. Proteasome chymotrypsin-like activity was measured by a fluorimetric method. Analysis of the cell cycle distribution was performed after propidium iodide staining. The apoptotic rate and proliferative index were determined by an immunohistochemical detection of cleaved caspase-3 and Ki-67, respectively. Our data showed that bortezomib induced a dose-dependent inhibition of proteasome chymotrypsin-like activity in the two glioma models. Maximal inhibition was achieved 24 h after drug injection and was approximately 30% of basal proteasome activity. However, this effect did not induce any increase in the apoptotic rate and did not modify cell cycle distribution. At the maximal dose tested (0.90 mg/kg), bortezomib did not show any growth delay as compared to untreated tumors, in either of the xenograft models. In conclusion, our study is the first to demonstrate that bortezomib, at a clinically relevant dose, did not have any effect on the apoptosis and proliferation of malignant gliomas in vivo. These results contrast with the promising preclinical data obtained in vitro with this drug and emphasize the importance of performing preclinical studies on animal models, in conditions close to clinical settings.


Assuntos
Antineoplásicos/uso terapêutico , Ácidos Borônicos/uso terapêutico , Glioma/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Pirazinas/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Bortezomib , Feminino , Humanos , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cancer Res ; 71(23): 7176-86, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22037871

RESUMO

Downregulating the leukocyte adhesion molecules expressed by endothelial cells that line tumor blood vessels can limit the entry of immune effector cells into the tumor mass, thereby contributing to tumoral immune escape. Egfl7 (also known as VE-statin) is a secreted protein specifically expressed by endothelial cells in normal tissues and by cancer cells in various human tumors. High levels of Egfl7 correlate with higher tumor grade and poorer prognosis. Here we show that expression of Egfl7 in breast and lung carcinoma cells accelerates tumor growth and metastasis in immunocompetent mice but not in immunodeficient mice. Tumors expressing Egfl7 were infiltrated relatively poorly by immune cells and were characterized by reduced levels of immunostimulatory cytokines [IFN-γ, interleukin-12 (IL-12)] and fewer endothelial adhesion molecules [intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1)]. In vitro studies revealed that Egfl7 inhibited the expression of leukocyte adhesion molecules by endothelial cells, preventing lymphocyte adhesion. In contrast, Egfl7 did not exert any effects on immune cell activation. Human breast cancer lesions expressing high levels of Egfl7 also expressed less ICAM-1 and VCAM-1 in their blood vessels, also indicating an inverse correlation between expression levels of Egfl7 and IFN-γ. Thus, Egfl7 expression in tumors promotes tumor progression by reducing the expression of endothelial molecules that mediate immune cell infiltration. Our findings highlight a novel mechanism through which tumors escape immune control.


Assuntos
Células Endoteliais/imunologia , Fatores de Crescimento Endotelial/imunologia , Proteínas/imunologia , Evasão Tumoral/imunologia , Animais , Proteínas de Ligação ao Cálcio , Adesão Celular/genética , Adesão Celular/imunologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Cultivadas , Progressão da Doença , Família de Proteínas EGF , Células Endoteliais/patologia , Fatores de Crescimento Endotelial/genética , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-12/metabolismo , Células Jurkat , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Metástase Neoplásica/genética , Metástase Neoplásica/imunologia , Metástase Neoplásica/patologia , Proteínas/genética , Evasão Tumoral/genética , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA