Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Clin Exp Pharmacol Physiol ; 46(10): 920-927, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31278773

RESUMO

Recombinant human endostatin (MES), showing potent inhibition on angiogenesis and tumour growth, has great potential as a therapeutic agent for tumours. The aim of this study was to evaluate the biophysical and biological characterization of PEGylated recombinant human endostatin (M2 ES). Recombinant human endostatin was mono-PEGylated by conjugation with methoxy polyethylene glycol aldehyde (mPEG-ALD), and the modification site was identified by digested peptide mapping and matrix assisted laser desorption/ionization-time of flight-mass spectrometry (MALDI-TOF-MS). The purity was assessed by SDS-PAGE, high-performance liquid chromatography (HPLC), and capillary zone electrophoresis. The physicochemical property was analyzed through fluorescence spectroscopy, and circular dichroism. The bioactivity and anti-tumour efficacy of M2 ES were evaluated using an in vitro endothelial cell migration model and a null-mouse xenograft model of a prostatic cancer, respectively. M2 ES molecules contain a single 20 kDa mPEG-ALD molecule conjugated at the N-terminal portion of MES. The purity of M2 ES was greater than 98%. The physicochemical analysis demonstrated that PEGylation does not change the secondary and tertiary structure of MES. Notably, M2 ES retards endothelial cell migration and tumour growth when compared to control group. These biophysical and biological characterization study data contribute to the initiation of the ongoing clinical study.


Assuntos
Polietilenoglicóis/química , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Sequência de Aminoácidos , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Humanos , Camundongos
2.
BMC Cancer ; 13: 479, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24128285

RESUMO

BACKGROUND: Endostatin (ES) inhibits endothelial cell proliferation, migration, invasion, and tube formation. It also shows antiangiogenesis and antitumor activities in several animal models. Endostatin specifically targets tumor vasculature to block tumor growth. Lidamycin (LDM), which consists of an active enediyne chromophore (AE) and a non-covalently bound apo-protein (LDP), is a member of chromoprotein family of antitumor antibiotics with extremely potent cytotoxicity to cancer cells. Therefore, we reasoned that endostatin-lidamycin (ES-LDM) fusion proteins upon energizing with enediyne chromophore may obtain the combined capability targeting tumor vasculature and tumor cell by respective ES and LDM moiety. METHODS: In this study, we designed and obtained two new endostatin-based fusion proteins, endostatin-LDP (ES-LDP) and LDP-endostatin (LDP-ES). In vitro, the antiangiogenic effect of fusion proteins was determined by the wound healing assay and tube formation assay and the cytotoxicity of their enediyne-energized analogs was evaluated by CCK-8 assay. Tissue microarray was used to analyze the binding affinity of LDP, ES or ES-LDP with specimens of human lung tissue and lung tumor. The in vivo efficacy of the fusion proteins was evaluated with human lung carcinoma PG-BE1 xenograft and the experimental metastasis model of 4T1-luc breast cancer. RESULTS: ES-LDP and LDP-ES disrupted the formation of endothelial tube structures and inhibited endothelial cell migration. Evidently, ES-LDP accumulated in the tumor and suppressed tumor growth and metastasis. ES-LDP and ES show higher binding capability than LDP to lung carcinoma; in addition, ES-LDP and ES share similar binding capability. Furthermore, the enediyne-energized fusion protein ES-LDP-AE demonstrated significant efficacy against lung carcinoma xenograft in athymic mice. CONCLUSIONS: The ES-based fusion protein therapy provides some fundamental information for further drug development. Targeting both tumor vasculature and tumor cells by endostatin-based fusion proteins and their enediyne-energized analogs probably provides a promising modality in cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Endostatinas/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Endostatinas/farmacocinética , Endostatinas/uso terapêutico , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Feminino , Humanos , Pulmão/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Análise Serial de Tecidos , Distribuição Tecidual , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Biol Chem ; 285(51): 40039-49, 2010 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-20937816

RESUMO

Heat shock protein 90α (Hsp90α) is a ubiquitously expressed molecular chaperone that is essential for eukaryotic homeostasis. Hsp90α can also be secreted extracellularly, where it has been shown to be involved in tumor metastasis. Extracellular Hsp90α interacts with and promotes the proteolytic activity of matrix metalloproteinase-2 (MMP-2). However, the regulatory mechanism of Hsp90α on MMP-2 activity is still unknown. Here we show that Hsp90α stabilizes MMP-2 and protects it from degradation in tumor cells. Further investigation reveals that this stabilization effect is isoform-specific, ATP-independent, and mediated by the interaction between the Hsp90α middle domain and the MMP-2 C-terminal hemopexin domain. Moreover, this mechanism also applies to endothelial cells that secrete more Hsp90α in their proliferating status. Furthermore, endothelial cell transmigration, Matrigel plug, and tumor angiogenesis assays demonstrate that extracellular Hsp90α promotes angiogenesis in an MMP-2-dependent manner. In sum, this study provides new insights into the molecular mechanism of how Hsp90α regulates its extracellular client proteins and also reveals for the first time the function of extracellular Hsp90α in promoting tumor angiogenesis.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neovascularização Patológica/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Células Endoteliais , Estabilidade Enzimática/genética , Proteínas de Choque Térmico HSP90/genética , Humanos , Metaloproteinase 2 da Matriz/genética , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patologia , Neovascularização Patológica/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína
4.
Biochemistry ; 48(49): 11655-63, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19877579

RESUMO

Endostatin is a potent angiogenesis inhibitor with heparin-dependent activities. Nucleolin, a novel functional receptor of endostatin, mediates both the internalization to endothelial cells and the antiangiogenic activity of endostatin. To define the exact role of the heparin binding motif in mediating the interaction between endostatin and its receptor nucleolin, up to six arginine residues (R155, R158, R184, R270, R193, and R194) located in the heparin binding motif of endostatin were substituted by alanine to make double, quadruple, or hexad point mutations, respectively. Contributions of the heparin binding motif to both the interaction with nucleolin and the biological activities of endostatin were investigated from in vitro to in vivo. Here we show that Arg to Ala point mutagenesis of the heparin binding motif does not interrupt the folding of endostatin but significantly impairs the interaction between endostatin and nucleolin. Double and quadruple mutants showed significantly decreased internalization to endothelial cells and antitumor activities, while the hexad Arg to Ala mutant completely lost its interaction with nucleolin and biological functions. Taken together, the present study demonstrates that the arginine clusters in the heparin binding motif of endostatin significantly contribute to its interaction with receptor nucleolin and mediate the antiangiogenic and antitumor activities of endostatin.


Assuntos
Endostatinas/metabolismo , Heparina/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Alanina/genética , Motivos de Aminoácidos/genética , Substituição de Aminoácidos/genética , Inibidores da Angiogênese/antagonistas & inibidores , Inibidores da Angiogênese/metabolismo , Animais , Antineoplásicos/antagonistas & inibidores , Antineoplásicos/metabolismo , Arginina/genética , Arginina/fisiologia , Linhagem Celular Tumoral , Endostatinas/genética , Endostatinas/fisiologia , Heparina/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Sítio-Dirigida , Fosfoproteínas/antagonistas & inibidores , Mutação Puntual , Ligação Proteica/genética , Conformação Proteica , Proteínas de Ligação a RNA/antagonistas & inibidores , Nucleolina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA