Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Front Immunol ; 12: 607692, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815364

RESUMEN

The transferrin receptor 1 (TfR1), also known as cluster of differentiation 71 (CD71), is a type II transmembrane glycoprotein that binds transferrin (Tf) and performs a critical role in cellular iron uptake through the interaction with iron-bound Tf. Iron is required for multiple cellular processes and is essential for DNA synthesis and, thus, cellular proliferation. Due to its central role in cancer cell pathology, malignant cells often overexpress TfR1 and this increased expression can be associated with poor prognosis in different types of cancer. The elevated levels of TfR1 expression on malignant cells, together with its extracellular accessibility, ability to internalize, and central role in cancer cell pathology make this receptor an attractive target for antibody-mediated therapy. The TfR1 can be targeted by antibodies for cancer therapy in two distinct ways: (1) indirectly through the use of antibodies conjugated to anti-cancer agents that are internalized by receptor-mediated endocytosis or (2) directly through the use of antibodies that disrupt the function of the receptor and/or induce Fc effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), or complement-dependent cytotoxicity (CDC). Although TfR1 has been used extensively as a target for antibody-mediated cancer therapy over the years, interest continues to increase for both targeting the receptor for delivery purposes and for its use as direct anti-cancer agents. This review focuses on the developments in the use of antibodies targeting TfR1 as direct anti-tumor agents.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Receptores de Transferrina/antagonistas & inhibidores , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos CD , Antineoplásicos Inmunológicos/uso terapéutico , Transporte Biológico/efectos de los fármacos , Biomarcadores de Tumor , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica , Humanos , Hierro/metabolismo , Terapia Molecular Dirigida/efectos adversos , Terapia Molecular Dirigida/métodos , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Int J Pharm ; 571: 118754, 2019 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-31604118

RESUMEN

In order to deliver Salvianolic acid B (Sal B) and Baicalin (BA) to the brain tissue to repair neuron damage and improve cerebral ischemia-reperfusion injury (IRI), in our previous study, a nanostructured lipid carrier (NLC) containing BA and Sal B, and modified by the transferrin receptor monoclonal antibody OX26 (OX26-BA/Sal B-NLC) was constructed. The present study is to evaluate its in vitro release behavior, in vitro and in vivo targeting ability, in vitro pharmacodynamics and brain pharmacokinetics. The results showed that the release mechanism of the formulation was in line with the Weibull model release equation. The in-vitro and in-vivo targeting ability study exhibited that OX26 modified formulations was obviously higher than that of non-modified and solution groups. The results of in vitro preliminary study to investigate the protective effect of OX26-BA/Sal B-NLC on oxygen-glucose deprivation/reperfusion injured cells showed that it could decrease the injury. Furthermore, the results of brain microdialysis study showed that the OX26-modified preparation group could significantly increase the content of BA in the brain. In the solution group and the unmodified group, Sal B can only be detected at few time points, while OX26-modified BA/Sal B-NLC could be detected within 4 h. These results indicating that OX26-modified NLC can promote the brain delivery of Sal B and BA combination.


Asunto(s)
Anticuerpos Monoclonales/química , Benzofuranos/administración & dosificación , Portadores de Fármacos/química , Medicamentos Herbarios Chinos/administración & dosificación , Flavonoides/administración & dosificación , Daño por Reperfusión/tratamiento farmacológico , Administración Intravenosa , Animales , Anticuerpos Monoclonales/farmacología , Benzofuranos/farmacocinética , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Modelos Animales de Enfermedad , Portadores de Fármacos/farmacología , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Liberación de Fármacos , Medicamentos Herbarios Chinos/farmacocinética , Flavonoides/farmacocinética , Humanos , Lípidos/química , Masculino , Ratones , Microdiálisis , Nanopartículas/química , Permeabilidad , Receptores de Transferrina/antagonistas & inhibidores , Daño por Reperfusión/patología , Distribución Tisular
3.
Oncol Rep ; 30(3): 1473-82, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23818062

RESUMEN

Retinoblastoma (RB) is an intraocular cancer that affects young children. There is an ongoing effort to find new agents for RB management that are effective, specific and with few side-effects. In the present study, we tested artesunate (ART), a synthetic derivative from the herbal drug artemisinin, used in the clinic for the treatment of malaria. We analyzed ART cytotoxicity in an RB cell line (RB-Y79) and in a retinal epithelial cell line (hTERT-RPE1) by flow cytometric analysis (FCM). We related the effect of ART to the expression of transferrin receptor 1 (TfR-1, also known as CD71) by knocking down CD71 with RNAi and analyzing cell cycle variables by FCM. We found that the cytotoxic action of ART is specific for RB cells in a dose-dependent manner, with low toxicity in normal retina cells. ART is more effective in RB than carboplatin with a markedly strong cytotoxic effect on carboplatin-resistant RB cells. RB had higher CD71 levels at the membrane compared to normal retinal cells. We showed that ART internalization in RB cells is dependent upon the expression of the CD71. In addition, ART blocked the cell cycle progression at the G1 phase, even at low doses, and decreased the proportion of RB cells in the S phase. In conclusion, we showed that ART is a promising drug exhibiting high selective cytotoxicity even against multidrug-resistant RB cells. Thus, we suggest that ART could be used in the treatment of RB.


Asunto(s)
Antígenos CD/metabolismo , Antimaláricos/farmacología , Apoptosis/efectos de los fármacos , Artemisininas/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores de Transferrina/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/patología , Antígenos CD/genética , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Artemisia/química , Artesunato , Western Blotting , Carboplatino/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Transferrina/antagonistas & inhibidores , Receptores de Transferrina/genética , Neoplasias de la Retina/tratamiento farmacológico , Neoplasias de la Retina/metabolismo , Retinoblastoma/tratamiento farmacológico , Retinoblastoma/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
4.
Nature ; 446(7131): 92-6, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17287727

RESUMEN

At least five arenaviruses cause viral haemorrhagic fevers in humans. Lassa virus, an Old World arenavirus, uses the cellular receptor alpha-dystroglycan to infect cells. Machupo, Guanarito, Junin and Sabia viruses are New World haemorrhagic fever viruses that do not use alpha-dystroglycan. Here we show a specific, high-affinity association between transferrin receptor 1 (TfR1) and the entry glycoprotein (GP) of Machupo virus. Expression of human TfR1, but not human transferrin receptor 2, in hamster cell lines markedly enhanced the infection of viruses pseudotyped with the GP of Machupo, Guanarito and Junin viruses, but not with those of Lassa or lymphocytic choriomeningitis viruses. An anti-TfR1 antibody efficiently inhibited the replication of Machupo, Guanarito, Junin and Sabia viruses, but not that of Lassa virus. Iron depletion of culture medium enhanced, and iron supplementation decreased, the efficiency of infection by Junin and Machupo but not Lassa pseudoviruses. These data indicate that TfR1 is a cellular receptor for New World haemorrhagic fever arenaviruses.


Asunto(s)
Antígenos CD/metabolismo , Arenavirus del Nuevo Mundo/metabolismo , Receptores de Transferrina/metabolismo , Receptores Virales/metabolismo , Anticuerpos/inmunología , Anticuerpos/farmacología , Antígenos CD/genética , Antígenos CD/inmunología , Arenavirus del Nuevo Mundo/efectos de los fármacos , Arenavirus del Nuevo Mundo/fisiología , Medios de Cultivo/química , Glicoproteínas/metabolismo , Humanos , Hierro/análisis , Hierro/farmacología , Receptores de Transferrina/antagonistas & inhibidores , Receptores de Transferrina/genética , Receptores de Transferrina/inmunología , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral/efectos de los fármacos
5.
Blood ; 108(8): 2745-54, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16804109

RESUMEN

We have previously reported that an anti-human transferrin receptor IgG3-avidin fusion protein (anti-hTfR IgG3-Av) inhibits the proliferation of an erythroleukemia-cell line. We have now found that anti-hTfR IgG3-Av also inhibits the proliferation of additional human malignant B and plasma cells. Anti-hTfR IgG3-Av induces internalization and rapid degradation of the TfR. These events can be reproduced in cells treated with anti-hTfR IgG3 cross-linked with a secondary Ab, suggesting that they result from increased TfR cross-linking. Confocal microscopy of cells treated with anti-hTfR IgG3-Av shows that the TfR is directed to an intracellular compartment expressing the lysosomal marker LAMP-1. The degradation of TfR is partially blocked by cysteine protease inhibitors. Furthermore, cells treated with anti-hTfR IgG3-Av exhibit mitochondrial depolarization and activation of caspases 9, 8, and 3. The mitochondrial damage and cell death can be prevented by iron supplementation, but cannot be fully blocked by a pan-caspase inhibitor. These results suggest that anti-hTfR IgG3-Av induces lethal iron deprivation, but the resulting cell death does not solely depend on caspase activation. This report provides insights into the mechanism of cell death induced by anti-TfR Abs such as anti-hTfR IgG3-Av, a molecule that may be useful in the treatment of B-cell malignancies such as multiple myeloma.


Asunto(s)
Avidina/farmacología , Neoplasias Hematológicas/terapia , Inmunoglobulina G/farmacología , Receptores de Transferrina/antagonistas & inhibidores , Clorometilcetonas de Aminoácidos/farmacología , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Reactivos de Enlaces Cruzados , Deferoxamina/farmacología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patología , Humanos , Hierro/farmacología , Leucemia de Células Plasmáticas/metabolismo , Leucemia de Células Plasmáticas/patología , Leucemia de Células Plasmáticas/terapia , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Mieloma Múltiple/terapia , Receptores de Transferrina/inmunología , Receptores de Transferrina/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Sideróforos/farmacología
6.
Hepatology ; 23(6): 1512-20, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8675172

RESUMEN

The hepatic uptake of transferrin-bound iron by a nontransferrin receptor (NTR)-mediated process was investigated using the human hepatoma cell line HuH7. Because HuH7 cells also acquire iron from transferrin by a receptor (TR)-mediated process, TR expression was inhibited by transfecting the cells with a plasmid containing human TR complementary DNA in antisense orientation relative to a human cytomegalovirus promoter/enhancer element. Cell clones were obtained that expressed a 50% to 60% reduction in cell surface TR, leading to a corresponding decrease in transferrin and iron uptake compared with wild-type cells. Uptake of transferrin by a second process was nonsaturable and not inhibited by a 100-fold excess of unlabeled transferrin. The amounts of transferrin taken up by the wild-type and antisense cells by this process were similar, showing that it did not involve TR. The proteolytic enzyme Pronase reduced the uptake of transferrin, suggesting that the NTR-mediated process entailed the nonsaturable binding of transferrin to plasma membrane proteins. This process, like the TR-mediated one, involved the internalization and recycling of transferrin, leading to accumulation of iron with time. Iron uptake mediated by NTR process was saturable and displaced by 100-fold excess unlabeled transferrin and reduced by weak bases and metabolic inhibitors. Therefore, the NTR-mediated process entailed transferrin adsorption to membrane-bound proteins, internalization, and release of iron from transferrin by a pH-dependent step followed by the intracellular transport of iron into ferritin and heme by a saturable carrier-mediated mechanism.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Receptores de Transferrina/metabolismo , Transferrina/metabolismo , Transporte Biológico Activo , Carcinoma Hepatocelular/genética , ADN sin Sentido/genética , ADN sin Sentido/farmacología , Expresión Génica , Humanos , Hierro/metabolismo , Cinética , Neoplasias Hepáticas/genética , Receptores de Transferrina/antagonistas & inhibidores , Receptores de Transferrina/genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA