Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
FASEB J ; 27(11): 4561-71, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23925653

RESUMEN

This study was designed to evaluate and characterize the molecular basis of antitumor activity of naturally occurring resveratrol (RES; 3,5,4'-trihydroxy-trans-stilbene) derivatives. The compounds were isolated from plants in previous studies and characterized spectroscopically. The antitumor activities of 31 RES derivatives, including dimers, trimers, and tetramers of RES, were evaluated using cell-based assays and validated on a murine model. Several trimeric and a tetrameric stilbenoids induced tumor cell apoptosis or growth arrest of several tumor cell lines with IC50 values (2.8-19.7 µM), significantly lower than that of RES (IC50>70 µM). Using pauciflorol B (PauB) as an example, we showed that the compound induced apoptosis p53 dependently, inducing p53 accumulation and p53-modulated gene expression in cells with wild-type p53, but not in those with nonfunctional p53. Reexpression of p53 in p53-null cells rescued cell death response. In parallel, the MAPK/p38 was activated and critical for PauB-induced killing. Interestingly, activation of p38 in p53 deficient cells was sufficient to drive cells into senescence via the p16-pRb pathway. Finally, PauB dose-dependently inhibited tumor growth on nude mice. Naturally occurring trimeric and tetrameric stilbenoids are potent antitumor agents. Those compounds exert antitumor effect through p53-dependent induction of apoptosis or senescence.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Estilbenos/farmacología , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/uso terapéutico , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Genes p16 , Células HeLa , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Polímeros/química , Resveratrol , Estilbenos/química , Estilbenos/uso terapéutico , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
Chin Med J (Engl) ; 135(15): 1821-1828, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35903953

RESUMEN

BACKGROUND: Previous evidence suggests inflammation may be a double-edged sword with cancer-promoting and cancer suppressing function. In this study, we explore the impact of local and systemic inflammation on cancer growth. METHODS: Female BALB/C mice were subcutaneously implanted with foreign body (plastic plates) to build up a local inflammation and intraperitoneally injected with PolyIC or lipopolysaccharides (LPS) to build up a systemic inflammation, followed by subcutaneous injection of 5  × 10 5 colon cancer cells. Immunohistochemistry and enzyme linked immunosorbent assay were utilized to detect the Ki67 and interleukin (IL) 6, IL-1ß, and monocyte chemoattractant protein-1 expression in the tumor tissues and serum, respectively. The distributions of immune cells and expression of toll-like receptors (TLRs) were evaluated by flow cytometry (FCM) and quantitative real time-polymerase chain reaction. RESULTS: The results showed that local inflammation induced by foreign body implantation suppressed tumor growth with decreased tumor weight ( P   =  0.001), volume ( P   =  0.004) and Ki67 index ( P   <  0.001). Compared with the control group, myeloid-derived suppressive cells sharply decreased ( P   =  0.040), while CD4 + T cells slightly increased in the tumor tissues of the group of foreign body-induced local inflammation ( P   =  0.035). Moreover, the number of M1 macrophages ( P   =  0.040) and expression of TLRs, especially TLR3 ( P  < 0.001) and TLR4 ( P  < 0.001), were significantly up-regulated in the foreign body group. Contrarily, tumor growth was significantly promoted in LPS or PolyIC-induced systemic inflammation ( P   =  0.009 and 0.006). FCM results showed M1 type macrophages ( P   =  0.017 and 0.006) and CD8 + T cells ( P   =  0.031 and 0.023) were decreased, while M2 type macrophages ( P  = 0.002 and 0.007) were significantly increased in tumor microenvironment of LPS or PolyIC-induced systemic inflammation group. In addition, the decreased expression of TLRs was detected in LPS or PolyIC group. CONCLUSIONS: The foreign body-induced local inflammation inhibited tumor growth, while LPS or PolyIC- induced systemic inflammation promoted tumor growth. The results suggested that the different outcomes of tumor growth might be attributed to the infiltration of anti-tumor or pro-tumor immune cells, especially M1 or M2 type macrophages into tumor microenvironment.


Asunto(s)
Cuerpos Extraños , Neoplasias , Animales , Quimiocina CCL2/metabolismo , Citocinas/metabolismo , Femenino , Inflamación/metabolismo , Interleucina-6/metabolismo , Antígeno Ki-67/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Plásticos/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Microambiente Tumoral
3.
Int J Nanomedicine ; 16: 5675-5692, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34456564

RESUMEN

PURPOSE: Since immune cells in the tumor microenvironment (TME) can affect the development and progression of tumors, strategies modulating immune cells are considered to have an important therapeutic effect. As a TLR7/8 agonist, R848 effectively activates the innate immune cells to exert an anti-tumor effect. Mn2+ has been reported to strongly promote the maturation of antigen-presenting cells (APCs), thereby enhancing the cytotoxicity of CD8+ T cells. Thus, we tried to investigate whether chitosan-poly(acrylic acid) nanoparticles (CS-PAA NPs) loaded with R848 and MnCl2 (R-M@CS-PAA NPs) could exert an anti-tumor effect by regulating the function of immune cells. METHODS: R-M@CS-PAA NPs were prepared, and their basic characteristics, anti-tumor effect, and potential mechanisms were explored both in vitro and in vivo. RESULTS: R-M@CS-PAA NPs easily released MnCl2 and R848 at low pH. In B16F10 mouse melanoma model, R-M@CS-PAA NPs exerted the most significant anti-melanoma effect compared with the control group and CS-PAA NPs loaded with R848 or MnCl2 alone. FITC-labeled R-M@CS-PAA NPs were displayed to be accumulated at the tumor site. R-M@CS-PAA NPs significantly increased the infiltration of M1 macrophages and CD8+ T cells but reduced the number of suppressive immune cells in the TME. Moreover, in vitro experiments showed that R-M@CS-PAA NPs polarized macrophages into the M1 phenotype to inhibit the proliferation of B16F10 cells. R-M@CS-PAA NPs also enhanced the killing function of CD8+ T cells to B16F10 cells. Of note, R-M@CS-PAA NPs not only promoted the maturation of APCs such as dendritic cells and macrophages by STING and NF-кB pathways, but also enhanced the ability of dendritic cells to present ovalbumin to OT-I CD8+ T cells to enhance the cytotoxicity of OT-I CD8+ T cells to ovalbumin-expressing B16F10 cells. CONCLUSION: These data indicate that the administration of R-M@CS-PAA NPs is an effective therapeutic strategy against melanoma.


Asunto(s)
Quitosano , Melanoma , Nanopartículas , Resinas Acrílicas , Animales , Linfocitos T CD8-positivos , Células Dendríticas , Macrófagos , Ratones , Microambiente Tumoral
4.
Mol Cancer Ther ; 14(2): 491-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25670331

RESUMEN

Lung cancer is the deadliest type of cancer for both men and women. In this study, we evaluate the in vitro and in vivo efficacy of a biotherapeutic agent composed of a lysosomal protein (Saposin C, SapC) and a phospholipid (dioleoylphosphatidylserine, DOPS), which can be assembled into nanovesicles (SapC-DOPS) with selective antitumor activity. SapC-DOPS targets phosphatidylserine, an anionic phospholipid preferentially exposed in the surface of cancer cells and tumor-associated vasculature. Because binding of SapC to phosphatidylserine is favored at acidic pHs, and the latter characterizes the milieu of many solid tumors, we tested the effect of pH on the binding capacity of SapC-DOPS to lung tumor cells. Results showed that SapC-DOPS binding to cancer cells was more pronounced at low pH. Viability assays on a panel of human lung tumor cells showed that SapC-DOPS cytotoxicity was positively correlated with cell surface phosphatidylserine levels, whereas mitochondrial membrane potential measurements were consistent with apoptosis-related cell death. Using a fluorescence tracking method in live mice, we show that SapC-DOPS specifically targets human lung cancer xenografts, and that systemic therapy with SapC-DOPS induces tumor apoptosis and significantly inhibits tumor growth. These results suggest that SapC-DOPS nanovesicles are a promising treatment option for lung cancer.


Asunto(s)
Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida , Nanoestructuras/química , Fosfatidilserinas/química , Saposinas/uso terapéutico , Liposomas Unilamelares/química , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones Desnudos , Saposinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Adv Mater ; 24(9): 1232-7, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22298490

RESUMEN

A poly (acryl acid) (PAA) post-treatment method is performed to modify the surface charge of ZnO nanospheres, hematite nanocubes, and Ag nanoprisms from highly positive to very negative by adjusting the PAA concentration, to and greatly modify their photoluminescence, cytotoxicity, magnetism, and surface plasmon resonance. This method provides a general way to tune the nanoparticle properties for broad physicochemical and biological applications.


Asunto(s)
Resinas Acrílicas/química , Compuestos Férricos/química , Nanopartículas/química , Plata/química , Óxido de Zinc/química , Línea Celular , Citotoxinas/química , Citotoxinas/toxicidad , Compuestos Férricos/toxicidad , Humanos , Luminiscencia , Nanopartículas/toxicidad , Nanopartículas/ultraestructura , Plata/toxicidad , Electricidad Estática , Resonancia por Plasmón de Superficie , Propiedades de Superficie , Óxido de Zinc/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA