Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Langmuir ; 37(4): 1353-1364, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33482065

RESUMEN

Stimuli-responsive emulsifiers have emerged as a class of smart agents that can permit regulated stabilization and destabilization of emulsions, which is essential for food, cosmetic, pharmaceutical, and petroleum industries. Here, we report the synthesis of novel "smart" hydroxyapatite (HaP) magnetic nanoparticles and their corresponding stimuli-responsive Pickering emulsions and explore their movement under confined spaces using a microfluidic platform. Pickering emulsions prepared with our magnetic stearic acid-functionalized Fe2O3@HaP nanoparticles exhibited pronounced pH-responsive behavior. We observed that the diameter of emulsion droplets decreases with an increase in pH. Swift demulsification was achieved by lowering the pH, whereas the reformation of emulsions was achieved by increasing the pH; this emulsification-demulsification cycling was successful for at least ten cycles. We used a microfluidic platform to test the stability of the emulsions under flowing conditions and their response to a magnetic field. We observed that the emulsion stability was diminished and droplet coalescence was enhanced by the application of the magnetic field. The smart nanoparticles we developed and their HaP-based emulsions present promising materials for pharmaceutical and petroleum industries, where responsive emulsions with controlled stabilities are required.

2.
Int J Mol Sci ; 20(11)2019 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-31142002

RESUMEN

Mesenchymal stem cells (MSCs) are emerging as a potential therapeutic intervention for brain injury due to their neuroprotective effects and safe profile. However, the homing ability of MSCs to injury sites still needs to be improved. Fibroblast Growth Factor 21 (FGF21) was recently reported to enhance cells migration in different cells type. In this study, we investigated whether MSCs that overexpressing FGF21 (MSC-FGF21) could exhibit enhanced homing efficacy in brain injury. We used novel Molday IONEverGreen™ (MIEG) as cell labeling probe that enables a non-invasive, high-sensitive and real-time MRI tracking. Using a mouse model of traumatic brain injury (TBI), MIEG labeled MSCs were transplanted into the contralateral lateral ventricle followed by real-time MRI tracking. FGF21 retained MSC abilities of proliferation and morphology. MSC-FGF21 showed significantly greater migration in transwell assay compared to control MSC. MIEG labeling showed no effects on MSCs' viability, proliferation and differentiation. Magnetic resonance imaging (MRI) revealed that FGF21 significantly enhances the homing of MSC toward injury site. Histological analysis further confirmed the MRI findings. Taken together, these results show that FGF21 overexpression and MIEG labeling of MSC enhances their homing abilities and enables non-invasive real time tracking of the transplanted cells, provides a promising approach for MSC based therapy and tracking in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Movimiento Celular , Factores de Crecimiento de Fibroblastos/genética , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Animales , Células Cultivadas , Factores de Crecimiento de Fibroblastos/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL
3.
Nanomedicine ; 14(3): 1019-1031, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29391212

RESUMEN

Non-small-cell lung cancer (NSCLC) is the most common type of lung cancer. Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors are commonly used as the first-line treatment for advanced NSCLC; however, the efficacy of drug delivery remains unknown. Hence, we successfully developed erlotinib-conjugated iron oxide nanoparticles (FeDC-E NPs) as theranostic probe that can potentially provide a new avenue for monitoring drug delivering through noninvasive magnetic resonance imaging. MRI ΔR2* relaxivity measurements offer an opportunity to quantitatively evaluate the uptake of FeDC-E NPs at cellular and tumoral levels. Additionally, NF-κB reporter gene system provides NF-κB activation status monitoring to validate the therapeutic efficiency of FeDC-E NPs. FeDC-E NPs not only inhibit the tumor growth and NF-κB-modulated antiapoptotic mechanism but also trigger extrinsic and intrinsic apoptotic pathways. Taken together, dual functional FeDC-E NPs offer diagnostic and therapeutic benefits against lung cancers, indicating that our presented probe could be applied in clinical.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Sistemas de Liberación de Medicamentos , Clorhidrato de Erlotinib/farmacología , Neoplasias Pulmonares/patología , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita/administración & dosificación , FN-kappa B/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Clorhidrato de Erlotinib/administración & dosificación , Clorhidrato de Erlotinib/química , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Procesamiento de Imagen Asistido por Computador , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Nanopartículas de Magnetita/química , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/genética , Fantasmas de Imagen , Células Tumorales Cultivadas
4.
J Nanobiotechnology ; 15(1): 86, 2017 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-29166921

RESUMEN

BACKGROUND: Targeted superparamagnetic iron oxide (SPIO) nanoparticles have emerged as a promising biomarker detection tool for molecular magnetic resonance (MR) image diagnosis. To identify patients who could benefit from Epidermal growth factor receptor (EGFR)-targeted therapies, we introduce lipid-encapsulated SPIO nanoparticles and hypothesized that anti-EGFR antibody cetuximab conjugated of such nanoparticles can be used to identify EGFR-positive glioblastomas in non-invasive T2 MR image assays. The newly introduced lipid-coated SPIOs, which imitate biological cell surface and thus inherited innate nonfouling property, were utilized to reduce nonspecific binding to off-targeted cells and prevent agglomeration that commonly occurs in nanoparticles. RESULTS: The synthesized targeted EGFR-antibody-conjugated SPIO (EGFR-SPIO) nanoparticles were characterized using dynamic light scattering, zeta potential assays, gel electrophoresis mobility shift assays, transmission electron microscopy (TEM) images, and cell line affinity assays, and the results showed that the conjugation was successful. The targeting efficiency of the synthesized EGFR-SPIO nanoparticles was confirmed through Prussian blue staining and TEM images by using glioblastoma cell lines with high or low EGFR expression levels. The EGFR-SPIO nanoparticles preferentially targeted U-251 cells, which have high EGFR expression, and were internalized by cells in a prolonged incubation condition. Moreover, the T2 MR relaxation time of EGFR-SPIO nanoparticles could be used for successfully identifying glioblastoma cells with elevated EGFR expression in vitro and distinguishing U-251 cells from U-87MG cells, which have low EFGR expression. CONCLUSION: These findings reveal that the lipid-encapsulated EGFR-SPIO nanoparticles can specifically target cells with elevated EGFR expression in the three tested human glioblastoma cell lines. The results of this study can be used for noninvasive molecular MR image diagnosis in the future.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Biomarcadores de Tumor/metabolismo , Cetuximab/farmacología , Receptores ErbB/metabolismo , Inmunoconjugados/farmacocinética , Nanopartículas de Magnetita/administración & dosificación , Neuroglía/efectos de los fármacos , Antineoplásicos Inmunológicos/química , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Cetuximab/química , Composición de Medicamentos , Receptores ErbB/genética , Expresión Génica , Humanos , Inmunoconjugados/química , Ligandos , Lípidos/química , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita/química , Neuroglía/metabolismo , Neuroglía/patología , Unión Proteica
5.
Neurotherapeutics ; 19(2): 616-634, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35267171

RESUMEN

Temozolomide (TMZ) monotherapy is known to be insufficient for resistant/relapsed glioblastoma (GBM), thus seeking a sensitization agent for TMZ is necessary. It was found that regorafenib may improve the overall survival of relapsed GBM patients. We aimed to discover whether regorafenib can enhance the anti-GBM effects of TMZ, and elucidate underlying mechanism. Our analysis of The Cancer Genome Atlas database revealed that the increased expression of CXCR4 is linked to poor survival of GBM patients. Additionally, TMZ treatment may trigger CXCR4/CXCL12 axis of GBM. We used two GBM cell lines, two primary GBM cells, and animal model to identify underlying mechanism and treatment efficacy of regorafenib combined with TMZ by cytotoxicity, apoptosis, reporter gene and invasion/migration assays, chemokine array, Western blotting, MRI, microarray, and immunohistochemistry. We observed that the chemokine CXCL-12 and its receptor CXCR4 regulate the resistance to TMZ, whereas the inhibition of CXCL-12/CXCR4 signaling sensitizes GBM cells to TMZ. The TMZ-induced CXCL-12/CXCR4 signaling, phosphor-extracellular signal-regulated kinases 1 and 2 (ERK1/2) and nuclear factor kappa light chain enhancer of activated B cells (NF-κB), and NF-κB-related proteins can effectively diminish when combining with regorafenib. Regorafenib significantly enhanced the TMZ-induced extrinsic/intrinsic apoptotic pathways, and facilitated the suppression of invasion and migration potential in GBM. Orthotopic tumor experiments demonstrated tumor size reduction and prolonged survival in combination group even with half-dose of TMZ. Our findings provide promising evidence that regorafenib may sensitize GBM to TMZ treatment through inhibition of the CXCL12/CXCR4/ERK/NF-κB signaling.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Compuestos de Fenilurea , Piridinas , Animales , Apoptosis , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Quimiocina CXCL12/farmacología , Quimiocina CXCL12/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , FN-kappa B/metabolismo , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Receptores CXCR4/uso terapéutico , Temozolomida/farmacología , Temozolomida/uso terapéutico
6.
Nanomaterials (Basel) ; 10(6)2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32516988

RESUMEN

Considerable health and climate benefits arising from the use of low-sulfur fuels has propelled the research on desulfurization of fossil fuels. Ideal fuels are urgently needed and are expected to be ultra-low in sulfur (10-15 ppm), with no greater than 50 ppm sulfur content. Although several sulfur removal techniques are available in refineries and petrochemical units, their high operational costs, complex operational needs, low efficiencies, and higher environmental risks render them unviable and challenging to implement. In recent years, mesoporous silica-based materials have emerged as promising desulfurizing agents, owing to their high porosity, high surface area, and easier functionalization compared to conventional materials. In this review, we report on recent progress in the synthesis and chemistry of new functionalized mesoporous silica materials aiming to lower the sulfur content of fuels. Additionally, we discuss the role of special active sites in these sorbent materials and investigate the formulations capable of encapsulating and trapping the sulfur-based molecules, which are challenging to remove due to their complexity, for example the species present in JP-8 jet fuels.

7.
Int J Nanomedicine ; 15: 97-114, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32021167

RESUMEN

BACKGROUND: Regenerative medicine field is still lagging due to the lack of adequate knowledge regarding the homing of therapeutic cells towards disease sites, tracking of cells during treatment, and monitoring the biodistribution and fate of cells. Such necessities require labeling of cells with imaging agents that do not alter their biological characteristics, and development of suitable non-invasive imaging modalities. PURPOSE: We aimed to develop, characterize, and standardize a facile labeling strategy for engineered mesenchymal stem cells without altering their viability, secretion of FGF21 protein (neuroprotective), and differentiation capabilities for non-invasive longitudinal MRI monitoring in live mice brains with high sensitivity. METHODS: We compared the labeling efficiency of different commercial iron oxide nanoparticles towards our stem cells and determined the optimum labeling conditions using prussian blue staining, confocal microscopy, transmission electron microscopy, and flow cytometry. To investigate any change in biological characteristics of labeled cells, we tested their viability by WST-1 assay, expression of FGF21 by Western blot, and adipogenic and osteogenic differentiation capabilities. MRI contrast-enhancing properties of labeled cells were investigated in vitro using cell-agarose phantoms and in mice brains transplanted with the therapeutic stem cells. RESULTS: We determined the nanoparticles that showed best labeling efficiency and least extracellular aggregation. We further optimized their labeling conditions (nanoparticles concentration and media supplementation) to achieve high cellular uptake and minimal extracellular aggregation of nanoparticles. Cell viability, expression of FGF21 protein, and differentiation capabilities were not impeded by nanoparticles labeling. Low number of labeled cells produced strong MRI signal decay in phantoms and in live mice brains which were visible for 4 weeks post transplantation. CONCLUSION: We established a standardized magnetic nanoparticle labeling platform for stem cells that were monitored longitudinally with high sensitivity in mice brains using MRI for regenerative medicine applications.


Asunto(s)
Encéfalo/diagnóstico por imagen , Factores de Crecimiento de Fibroblastos/metabolismo , Imagen por Resonancia Magnética/métodos , Nanopartículas de Magnetita , Células Madre Mesenquimatosas/química , Células Madre Mesenquimatosas/fisiología , Adipogénesis , Animales , Diferenciación Celular , Medios de Contraste , Compuestos Férricos/química , Factores de Crecimiento de Fibroblastos/genética , Citometría de Flujo , Ingeniería Genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Osteogénesis , Distribución Tisular
8.
Oncotarget ; 9(15): 11922-11937, 2018 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-29552282

RESUMEN

The small-molecule naphtha [2,3-f]quinoxaline-7,12-dione (NSC745887) can effectively inhibit the proliferation of various cancers by trapping DNA-topoisomerase cleavage. The aim of this study was to elucidate cellular responses of NSC745887 in human glioblastoma multiforme (GBM, U118MG and U87MG cells) and investigate the underlying molecular mechanisms. NSC745887 reduced the cell survival rate and increased the sub-G1 population in dose- and time-dependent manners in GBM cells. Moreover, NSC745887 increased expression of γH2AX and caused DNA fragmentation leading to DNA damage. Furthermore, Annexin V/propidium iodide and Br-dTP staining showed the apoptotic effect of NSC745887 in GBM cells. DNA repair proteins of ataxia-telangiectasia mutated (ATM), ATM and Rad3-related, and decoy receptor 3 also decreased with NSC745887 treatment. In addition, NSC745887 caused apoptosis by the caspase-8/9-caspase-3-poly(ADP-ribose) polymerase cascade. An in vivo study indicated that NSC745887 suppressed the [18F]-FDG-specific uptake value in brain tumors. Histological staining also indicated a decrease in Ki-67 and increases in γH2AX and cleaved caspase-3 in the brain tumor area. These data provide preclinical evidence for NSC745887 as a potential new small molecule drug for managing glioblastomas.

9.
PLoS One ; 11(4): e0154278, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27100886

RESUMEN

The novel compounds NSC745885 and NSC757963 developed at our laboratory were tested against a panel of 60 cancer cell lines at the National Cancer Institute, USA, and a panel of 39 cancer cell lines at the Japanese Foundation of Cancer Research. Both compounds demonstrated selective unique multi-log differential patterns of activity, with GI50 values in the sub-micro molar range against cancer cells rather than normal cardiac cells. NSC757963 showed high selectivity towards the leukemia subpanel. Activities of both compounds strongly correlated to expression of NFKB1 and CSNK2B genes, implying that they may inhibit the NF-κB pathway. Immunocytochemical microscopy of OVCAR-3 cells showed clear cytosolic accumulation of the NF-κB p65 subunit following treatment. Western blotting showed dose dependent inhibition of the nuclear expression of the NF-κB p65 subunit with subsequent accumulation in the cytosol following treatment. Docking experiments showed binding of both compounds to the NF-κB activator IKKß subunit preventing its translocation to the nucleus. Collectively, these results confirm the ability of our compounds to inhibit the constitutively active NF-κB pathway of OVCAR-3 cells. Furthermore, COMPARE analysis indicated that the activity of NSC757963 is similar to the antituberculosis agent rifamycin SV, this was confirmed by testing the antimycobacterial activity of NSC757963 against Mycobacterium tuberculosis, results revealed potent activity suitable for use in clinical practice. Molecular properties and Lipinski's parameters predicted acceptable bioavailability properties with no indication of mutagenicity, tumorigenicity, irritability and reproductive effects. Oral absorption experiments using the human Caco-2 model showed high intestinal absorption of NSC745885 by passive transport mechanism with no intestinal efflux or active transport mechanisms. The unique molecular characterization as well as the illustrated anticancer spectra of activity and bioavailability properties warrant further development of our compounds and present a foundation brick in the pre-clinical investigations to implement such compounds in clinical practice.


Asunto(s)
Antineoplásicos/farmacología , Antituberculosos/farmacología , Regulación Neoplásica de la Expresión Génica , Tiadiazoles/farmacología , Factor de Transcripción ReIA/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antituberculosos/síntesis química , Disponibilidad Biológica , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Perfilación de la Expresión Génica , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Absorción Intestinal/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Modelos Biológicos , Simulación del Acoplamiento Molecular , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/crecimiento & desarrollo , Transducción de Señal , Tiadiazoles/síntesis química , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
10.
Sci Rep ; 6: 36650, 2016 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-27833124

RESUMEN

We designed and synthesized novel theranostic nanoparticles that showed the considerable potential for clinical use in targeted therapy, and non-invasive real-time monitoring of tumors by MRI. Our nanoparticles were ultra-small with superparamagnetic iron oxide cores, conjugated to erlotinib (FeDC-E NPs). Such smart targeted nanoparticles have the preference to release the drug intracellularly rather than into the bloodstream, and specifically recognize and kill cancer cells that overexpress EGFR while being non-toxic to EGFR-negative cells. MRI, transmission electron microscopy and Prussian blue staining results indicated that cellular uptake and intracellular accumulation of FeDC-E NPs in the EGFR overexpressing cells was significantly higher than those of the non-erlotinib-conjugated nanoparticles. FeDC-E NPs inhibited the EGFR-ERK-NF-κB signaling pathways, and subsequently suppressed the migration and invasion capabilities of the highly invasive and migrative CL1-5-F4 cancer cells. In vivo tumor xenograft experiments using BALB/c nude mice showed that FeDC-E NPs could effectively inhibit the growth of tumors. T2-weighted MRI images of the mice showed significant decrease in the normalized signal within the tumor post-treatment with FeDC-E NPs compared to the non-targeted control iron oxide nanoparticles. This is the first study to use erlotinib as a small-molecule targeting agent for nanoparticles.


Asunto(s)
Medios de Contraste/farmacología , Sistemas de Liberación de Medicamentos/métodos , Clorhidrato de Erlotinib/farmacología , Imagen por Resonancia Magnética , Nanopartículas de Magnetita/uso terapéutico , Neoplasias Experimentales/diagnóstico por imagen , Animales , Medios de Contraste/química , Clorhidrato de Erlotinib/química , Humanos , Células Jurkat , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/ultraestructura , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA