Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Bioorg Med Chem ; 95: 117501, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37864885

RESUMEN

A novel series of 4-(3-1H-indazolyl)amino quinazoline derivatives were developed as PAK4 inhibitors based on a scaffold hopping strategy. Compounds 27e, 27g, 27i and 27j were found to exhibit potent inhibitory activity against PAK4 (IC50 = 10, 13, 11 and 9 nM, respectively). Subsequent cellular assay demonstrated that compound 27e possessed the strongest antiproliferative activity against A549 cells with an IC50 value of 0.61 µM, a little bit better than PF-3758309. Further anticancer mechanistic investigation revealed that compound 27e significantly induced apoptosis of A549 cells in a concentration-dependent manner and blocked the cell cycle at phase G0/G1. A docking model between compound 27e and PAK4 was proposed to elucidate its possible binding modes. As a promising PAK4 inhibitor, compound 27e may serve as a candidate for the development of novel PAK4-targeted anticancer drug.


Asunto(s)
Antineoplásicos , Quinazolinas , Humanos , Quinazolinas/química , Relación Estructura-Actividad , Proliferación Celular , Inhibidores de Proteínas Quinasas/química , Células A549 , Antineoplásicos/química , Ensayos de Selección de Medicamentos Antitumorales , Simulación del Acoplamiento Molecular , Diseño de Fármacos , Línea Celular Tumoral
2.
Fitoterapia ; 163: 105347, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36336137

RESUMEN

Two novel series of 1,2-didehydro-7-hydroxy-3-ox-14-deoxyandrographolide and 1,2-didehydro-8,17-epoxy-3-ox-14-deoxyandrographolide derivatives were designed, synthesized and evaluated for their cytotoxic activity in vitro against two human cancer cell lines HCT-116 (human colon cancer) and MCF-7 (human breast cancer). Most tested compounds, especially those of the first series, displayed better inhibitory activity on both HCT-116 and MCF-7 cells than andrographolide. HCT-116 cells were found to be more sensitive to tested compounds than MCF-7, and compound 13b exhibited the most potent activity against HCT-116, with an IC50 value of 7.32 µM. Further anti-cancer mechanistic investigation demonstrated that compound 13b effectively suppressed the growth of HCT-116 cells by triggering early and late cellular apoptosis in a concentration-dependent manner and arresting cell cycle in S phase.


Asunto(s)
Antineoplásicos , Oxígeno , Humanos , Ensayos de Selección de Medicamentos Antitumorales , Relación Estructura-Actividad , Estructura Molecular , Relación Dosis-Respuesta a Droga , Antineoplásicos/farmacología , Apoptosis , Proliferación Celular , Línea Celular Tumoral , Diseño de Fármacos
3.
Proc Natl Acad Sci U S A ; 117(31): 18470-18476, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32690682

RESUMEN

Lipid membrane fusion is an essential process for a number of critical biological functions. The overall process is thermodynamically favorable but faces multiple kinetic barriers along the way. Inspired by nature's engineered proteins such as SNAP receptor [soluble N-ethylmale-imide-sensitive factor-attachment protein receptor (SNARE)] complexes or viral fusogenic proteins that actively promote the development of membrane proximity, nucleation of a stalk, and triggered expansion of the fusion pore, here we introduce a synthetic fusogen that can modulate membrane fusion and equivalently prime lipid membranes for calcium-triggered fusion. Our fusogen consists of a gold nanoparticle functionalized with an amphiphilic monolayer of alkanethiol ligands that had previously been shown to fuse with lipid bilayers. While previous efforts to develop synthetic fusogens have only replicated the initial steps of the fusion cascade, we use molecular simulations and complementary experimental techniques to demonstrate that these nanoparticles can induce the formation of a lipid stalk and also drive its expansion into a fusion pore upon the addition of excess calcium. These results have important implications in general understanding of stimuli-triggered fusion and the development of synthetic fusogens for biomedical applications.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Oro/química , Membrana Dobles de Lípidos/metabolismo , Nanopartículas del Metal/química , Calcio/química , Membrana Celular/química , Oro/metabolismo , Humanos , Membrana Dobles de Lípidos/química , Fusión de Membrana , Simulación de Dinámica Molecular , Proteínas SNARE/metabolismo , Análisis de Matrices Tisulares
4.
Ann Thorac Surg ; 108(6): 1717-1723, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31039351

RESUMEN

BACKGROUND: Nodal skip metastasis (NSM) is a prognostic factor in certain malignant tumors, but the clinical and prognostic implications of NSM in esophageal squamous cell carcinoma (ESCC) are still unclear. The study aimed to assess its risk factors and prognostic value in thoracic ESCC. METHODS: A retrospective study was conducted in patients with thoracic ESCC who underwent esophagectomy from March 2009 to March 2012 in the Department of Thoracic Surgery, West China Hospital, Sichuan University. The prognostic implications and risk factors of NSM were assessed in our study. RESULTS: The incidence of NSM in the entire cohort was 37.9%. Tumor location (P = .016), pT stage (P = .029), and pN stage (P < .001) were identified to be independent risk factors for NSM. The overall survival (OS) was similar between patients with and without NSM. The OS had no significant difference between pN1 patients with and without NSM, whereas the OS was significantly worse in pN2 patients with NSM than those without NSM (P = .001). The OS was similar between patients with NSM level 1 and NSM level 2, but the OS was significantly better in patients with NSM level 1 than NSM level 2 among patients with lower thoracic ESCC (P = .013). CONCLUSIONS: The effect of NSM on prognosis of thoracic ESCC may be mainly reflected in patients with pN2 stage. The prognostic value of NSM level for thoracic ESCC may be mainly reflected in patients with lower thoracic tumor.


Asunto(s)
Carcinoma de Células Escamosas de Esófago/secundario , Escisión del Ganglio Linfático/métodos , Ganglios Linfáticos/cirugía , Estadificación de Neoplasias , Medición de Riesgo/métodos , Adulto , Anciano , Anciano de 80 o más Años , China/epidemiología , Carcinoma de Células Escamosas de Esófago/epidemiología , Carcinoma de Células Escamosas de Esófago/cirugía , Esofagectomía , Femenino , Humanos , Incidencia , Ganglios Linfáticos/patología , Metástasis Linfática , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Tasa de Supervivencia/tendencias
5.
Biomaterials ; 190-191: 111-120, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30415018

RESUMEN

Although immunotherapy shows great promise for the long-term control of cancer, many tumors still fail to respond to treatment. To improve the outcome, the delivery of immunostimulants to the lymph nodes draining the tumor, where the antitumor immune response is initiated, is key. Efforts to use nanoparticles as carriers for cancer immunotherapy have generally required targeting agents and chemical modification of the drug, and have unfortunately resulted in low delivery and therapeutic efficiency. Here, we report on the efficacy of gold nanoparticles with approximately 5 nm hydrodynamic diameter coated with a mixture of 1-octanethiol and 11-mercaptoundecanesulfonic acid for the delivery of an immunostimulatory TLR7 ligand to tumor-draining lymph nodes. The drug was loaded without modification through nonspecific adsorption into the ligand shell of the nanoparticles, taking advantage of their amphiphilic nature. After loading, nanoparticles retained their stability in solution without significant premature release of the drug, and the drug cargo was immunologically active. Upon subcutaneous injection into tumor-bearing mice, the drug-loaded particles were rapidly transported to the tumor-draining lymph nodes. There, they induced a local immune activation and fostered a cytotoxic T-cell response that was specific for the tumor. Importantly, the particle-delivered TLR7 ligand blocked the growth of large established tumors and significantly prolonged survival compared to the free form of the drug. Thus, we demonstrate for the first time that nanoparticle delivery of a TLR7 immunostimulant to the tumor-draining lymph nodes enhances antitumor immunity and improves the outcome of cancer immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Neoplasias del Colon/terapia , Portadores de Fármacos/química , Oro/química , Nanopartículas del Metal/química , Receptor Toll-Like 7/agonistas , Adyuvantes Inmunológicos/uso terapéutico , Animales , Línea Celular , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Inmunoterapia , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ratones Endogámicos C57BL , Receptor Toll-Like 7/inmunología
6.
J Cell Mol Med ; 23(2): 1375-1385, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30565387

RESUMEN

FAM3B has been suggested to play important roles in the progression of many cancers, such as gastric, oral, colon and prostate cancer. However, little is known about the role of FAM3B in human esophageal squamous cell carcinoma (ESCC). In the present study, we found that FAM3B expression was higher in ESCC tissues than in adjacent normal tissues. Using quantitative real-time polymerase chain reaction, we found similar results in cell lines. FAM3B expression was significantly related to T/TNM stage. Importantly, Kaplan-Meier analysis revealed that a high expression level of FAM3B predicted a poor outcome for ESCC patients. Overexpression of FAM3B inhibits ESCC cell death, increases oesophageal tumour growth in xenografted nude mice, and promotes ESCC cell migration and invasion. Further studies confirmed that FAM3B regulates the AKT-MDM2-p53 pathway and two core epithelial-to-mesenchymal transition process markers, Snail and E-cadherin. Our results provide new insights into the role of FAM3B in the progression of ESCC and suggest that FAM3B may be a promising molecular target and diagnostic marker for ESCC.


Asunto(s)
Citocinas/metabolismo , Transición Epitelial-Mesenquimal , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/secundario , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Movimiento Celular , Proliferación Celular , Citocinas/genética , Progresión de la Enfermedad , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis Linfática , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Pronóstico , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biomater Sci ; 7(1): 113-124, 2018 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-30444251

RESUMEN

We sought to develop a nanoparticle vehicle that could efficiently deliver small molecule drugs to target lymphocyte populations. The synthesized amphiphilic organic ligand-protected gold nanoparticles (amph-NPs) were capable of sequestering large payloads of small molecule drugs within hydrophobic pockets of their ligand shells. These particles exhibit membrane-penetrating activity in mammalian cells, and thus enhanced uptake of a small molecule TGF-ß inhibitor in T cells in cell culture. By conjugating amph-NPs with targeting antibodies or camelid-derived nanobodies, the particles' cell-penetrating properties could be temporarily suppressed, allowing targeted uptake in specific lymphocyte subpopulations. Degradation of the protein targeting moieties following particle endocytosis allowed the NPs to recover their cell-penetrating activity in situ to enter the cytoplasm of T cells. In vivo, targeted amph-NPs showed 40-fold enhanced uptake in CD8+ T cells relative to untargeted particles, and delivery of TGF-ß inhibitor-loaded particles to T cells enhanced their cytokine polyfunctionality in a cancer vaccine model. Thus, this system provides a facile approach to concentrate small molecule compounds in target lymphocyte populations of interest for immunotherapy in cancer and other diseases.


Asunto(s)
Sistemas de Liberación de Medicamentos , Oro/química , Inmunoconjugados/química , Nanopartículas del Metal/química , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Linfocitos T/efectos de los fármacos , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Células Cultivadas , Femenino , Oro/farmacocinética , Inmunoconjugados/farmacocinética , Ratones Endogámicos C57BL , Bibliotecas de Moléculas Pequeñas/farmacología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/análisis
8.
Bioconjug Chem ; 29(4): 1131-1140, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29465986

RESUMEN

The development of synthetic nanomaterials that could embed within, penetrate, or induce fusion between membranes without permanent disruption would have great significance for biomedical applications. Here we describe structure-function relationships of highly water-soluble gold nanoparticles comprised of an ∼1.5-5 nm diameter metal core coated by an amphiphilic organic ligand shell, which exhibit membrane embedding and fusion activity mediated by the surface ligands. Using an environment-sensitive dye anchored within the ligand shell as a sensor of membrane embedding, we demonstrate that particles with core sizes of ∼2-3 nm are capable of embedding within and penetrating fluid bilayers. At the nanoscale, these particles also promote spontaneous fusion of liposomes or spontaneously embed within intact liposomal vesicles. These studies provide nanoparticle design and selection principles that could be used in drug delivery applications, as membrane stains, or for the creation of novel organic/inorganic nanomaterial self-assemblies.


Asunto(s)
Membrana Dobles de Lípidos , Fusión de Membrana , Nanopartículas/química , Permeabilidad , Compuestos de Boro/química , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Liposomas , Tamaño de la Partícula , Electricidad Estática , Relación Estructura-Actividad
10.
Nat Commun ; 8: 14069, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28094297

RESUMEN

Inorganic nanoparticles (NPs) are studied as drug carriers, radiosensitizers and imaging agents, and characterizing nanoparticle biodistribution is essential for evaluating their efficacy and safety. Tracking NPs at the single-cell level with current technologies is complicated by the lack of reliable methods to stably label particles over extended durations in vivo. Here we demonstrate that mass cytometry by time-of-flight provides a label-free approach for inorganic nanoparticle quantitation in cells. Furthermore, mass cytometry can enumerate AuNPs with a lower detection limit of ∼10 AuNPs (3 nm core size) in a single cell with tandem multiparameter cellular phenotyping. Using the cellular distribution insights, we selected an amphiphilic surface ligand-coated AuNP that targeted myeloid dendritic cells in lymph nodes as a peptide antigen carrier, substantially increasing the efficacy of a model vaccine in a B16-OVA melanoma mouse model. This technology provides a powerful new level of insight into nanoparticle fate in vivo.


Asunto(s)
Oro/análisis , Espectrometría de Masas/métodos , Nanopartículas del Metal/análisis , Análisis de la Célula Individual/métodos , Animales , Células Dendríticas/química , Células Dendríticas/metabolismo , Portadores de Fármacos/química , Femenino , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Distribución Tisular , Vacunas/administración & dosificación , Vacunas/química , Vacunas/metabolismo
11.
Nanoscale ; 7(26): 11420-32, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26077112

RESUMEN

Erythrocytes are attractive as potential cell-based drug carriers because of their abundance and long lifespan in vivo. Existing methods for loading drug cargos into erythrocytes include hypotonic treatments, electroporation, and covalent attachment onto the membrane, all of which require ex vivo manipulation. Here, we characterized the properties of amphiphilic gold nanoparticles (amph-AuNPs), comprised of a ∼2.3 nm gold core and an amphiphilic ligand shell, which are able to embed spontaneously within erythrocyte membranes and might provide a means to load drugs into red blood cells (RBCs) directly in vivo. Particle interaction with RBC membranes occurred rapidly at physiological temperature. We further show that amph-AuNP uptake by RBCs was limited by the glycocalyx and was particularly influenced by sialic acids on cell surface proteoglycans. Using a reductionist model membrane system with synthetic lipid vesicles, we confirmed the importance of membrane fluidity and the glycocalyx in regulating amph-AuNP/membrane interactions. These results thus provide evidence for the interaction of amph-AuNPs with erythrocyte membranes and identify key membrane components that govern this interaction, providing a framework for the development of amph-AuNP-carrying erythrocyte 'pharmacytes' in vivo.


Asunto(s)
Membrana Eritrocítica/química , Membrana Eritrocítica/ultraestructura , Glicocálix/química , Glicocálix/ultraestructura , Oro/química , Nanopartículas del Metal/química , Humanos , Nanopartículas del Metal/ultraestructura
12.
ACS Nano ; 8(9): 8992-9002, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25123510

RESUMEN

Amphiphilic gold nanoparticles (amph-NPs), composed of gold cores surrounded by an amphiphilic mixed organic ligand shell, are capable of embedding within and traversing lipid membranes. Here we describe a strategy using crosslink-stabilized lipid nanocapsules (NCs) as carriers to transport such membrane-penetrating particles into tumor cells and promote their transfer to intracellular membranes for enhanced radiotherapy of cancer. We synthesized and characterized interbilayer-crosslinked multilamellar lipid vesicles (ICMVs) carrying amph-NPs embedded in the capsule walls, forming Au-NCs. Confocal and electron microscopies revealed that the intracellular distribution of amph-NPs within melanoma and breast tumor cells following uptake of free particles vs Au-NCs was quite distinct and that amph-NPs initially delivered into endosomes by Au-NCs transferred over a period of hours to intracellular membranes through tumor cells, with greater intracellular spread in melanoma cells than breast carcinoma cells. Clonogenic assays revealed that Au-NCs enhanced radiotherapeutic killing of melanoma cells. Thus, multilamellar lipid capsules may serve as an effective carrier to deliver amphiphilic gold nanoparticles to tumors, where the membrane-penetrating properties of these materials can significantly enhance the efficacy of frontline radiotherapy treatments.


Asunto(s)
Portadores de Fármacos/química , Oro/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Membranas Intracelulares/metabolismo , Lípidos/química , Melanoma Experimental/radioterapia , Nanopartículas del Metal/química , Animales , Transporte Biológico , Línea Celular Tumoral , Citoplasma/metabolismo , Oro/química , Melanoma Experimental/patología , Ratones
13.
Nano Lett ; 13(9): 4060-7, 2013 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-23915118

RESUMEN

Anionic, monolayer-protected gold nanoparticles (AuNPs) have been shown to nondisruptively penetrate cellular membranes. Here, we show that a critical first step in the penetration process is potentially the fusion of such AuNPs with lipid bilayers. Free energy calculations, experiments on unilamellar and multilamellar vesicles, and cell studies all support this hypothesis. Furthermore, we show that fusion is only favorable for AuNPs with core diameters below a critical size that depends on the monolayer composition.


Asunto(s)
Oro/química , Nanopartículas del Metal/química , Membrana Celular/química , Membrana Dobles de Lípidos/química , Tamaño de la Partícula
14.
J Am Chem Soc ; 132(38): 13270-8, 2010 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-20572667

RESUMEN

The water-solvable FePt nanoparticles of 3, 6, and 12 nm in diameter (3 nm-, 6 nm-, and 12 nm-FePt) were synthesized and applied as a dual modality contrast agent for CT/MRI molecular imaging. These nanoparticles present excellent biocompatibility and hemocompatibility in all test concentrations for the imaging contrast. The biodistribution analysis revealed the highest serum concentration and circulation half-life for 12 nm-FePt, followed by 6 nm-FePt then 3 nm-FePt. Thus, the 3 nm-FePt showed higher brain concentrations. Anti-Her2 antibody conjugated FePt nanoparticles demonstrated molecular expression dependent CT/MRI dual imaging contrast effect in MBT2 cell line and its Her2/neu gene knock out counterpart. Selective contrast enhancement of Her2/neu overexpression cancer lesions in both CT and MRI was found in tumor bearing animal after tail vein injection of the nanoparticles. The 12 nm-FePt outperformed 3 nm-FePt in both imaging modalities. These results indicate the potential of FePt nanoparticles to serve as novel multimodal molecular imaging contrast agents in clinical settings.


Asunto(s)
Hierro/química , Imagen por Resonancia Magnética , Nanopartículas del Metal , Platino (Metal)/química , Tomografía Computarizada por Rayos X , Animales , Línea Celular Tumoral , Técnicas de Inactivación de Genes , Genes erbB-2 , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C3H , Microscopía Electrónica de Transmisión , Distribución Tisular , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA