Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Elife ; 122024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38747577

RESUMEN

Certain bacteria demonstrate the ability to target and colonize the tumor microenvironment, a characteristic that positions them as innovative carriers for delivering various therapeutic agents in cancer therapy. Nevertheless, our understanding of how bacteria adapt their physiological condition to the tumor microenvironment remains elusive. In this work, we employed liquid chromatography-tandem mass spectrometry to examine the proteome of E. coli colonized in murine tumors. Compared to E. coli cultivated in the rich medium, we found that E. coli colonized in tumors notably upregulated the processes related to ferric ions, including the enterobactin biosynthesis and iron homeostasis. This finding indicated that the tumor is an iron-deficient environment to E. coli. We also found that the colonization of E. coli in the tumor led to an increased expression of lipocalin 2 (LCN2), a host protein that can sequester the enterobactin. We therefore engineered E. coli in order to evade the nutritional immunity provided by LCN2. By introducing the IroA cluster, the E. coli synthesizes the glycosylated enterobactin, which creates steric hindrance to avoid the LCN2 sequestration. The IroA-E. coli showed enhanced resistance to LCN2 and significantly improved the anti-tumor activity in mice. Moreover, the mice cured by the IroA-E. coli treatment became resistant to the tumor re-challenge, indicating the establishment of immunological memory. Overall, our study underscores the crucial role of bacteria's ability to acquire ferric ions within the tumor microenvironment for effective cancer therapy.


Asunto(s)
Escherichia coli , Hierro , Lipocalina 2 , Animales , Escherichia coli/genética , Escherichia coli/metabolismo , Lipocalina 2/metabolismo , Lipocalina 2/genética , Ratones , Hierro/metabolismo , Neoplasias/terapia , Neoplasias/inmunología , Enterobactina/metabolismo , Microambiente Tumoral , Línea Celular Tumoral
2.
ACS Nano ; 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38718220

RESUMEN

Mesoporous silica nanoparticles (MSNs) represent a promising avenue for targeted brain tumor therapy. However, the blood-brain barrier (BBB) often presents a formidable obstacle to efficient drug delivery. This study introduces a ligand-free PEGylated MSN variant (RMSN25-PEG-TA) with a 25 nm size and a slight positive charge, which exhibits superior BBB penetration. Utilizing two-photon imaging, RMSN25-PEG-TA particles remained in circulation for over 24 h, indicating significant traversal beyond the cerebrovascular realm. Importantly, DOX@RMSN25-PEG-TA, our MSN loaded with doxorubicin (DOX), harnessed the enhanced permeability and retention (EPR) effect to achieve a 6-fold increase in brain accumulation compared to free DOX. In vivo evaluations confirmed the potent inhibition of orthotopic glioma growth by DOX@RMSN25-PEG-TA, extending survival rates in spontaneous brain tumor models by over 28% and offering an improved biosafety profile. Advanced LC-MS/MS investigations unveiled a distinctive protein corona surrounding RMSN25-PEG-TA, suggesting proteins such as apolipoprotein E and albumin could play pivotal roles in enabling its BBB penetration. Our results underscore the potential of ligand-free MSNs in treating brain tumors, which supports the development of future drug-nanoparticle design paradigms.

3.
ACS Appl Mater Interfaces ; 16(17): 21722-21735, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38629735

RESUMEN

While temozolomide (TMZ) has been a cornerstone in the treatment of newly diagnosed glioblastoma (GBM), a significant challenge has been the emergence of resistance to TMZ, which compromises its clinical benefits. Additionally, the nonspecificity of TMZ can lead to detrimental side effects. Although TMZ is capable of penetrating the blood-brain barrier (BBB), our research addresses the need for targeted therapy to circumvent resistance mechanisms and reduce off-target effects. This study introduces the use of PEGylated mesoporous silica nanoparticles (MSN) with octyl group modifications (C8-MSN) as a nanocarrier system for the delivery of docetaxel (DTX), providing a novel approach for treating TMZ-resistant GBM. Our findings reveal that C8-MSN is biocompatible in vitro, and DTX@C8-MSN shows no hemolytic activity at therapeutic concentrations, maintaining efficacy against GBM cells. Crucially, in vivo imaging demonstrates preferential accumulation of C8-MSN within the tumor region, suggesting enhanced permeability across the blood-brain tumor barrier (BBTB). When administered to orthotopic glioma mouse models, DTX@C8-MSN notably prolongs survival by over 50%, significantly reduces tumor volume, and decreases side effects compared to free DTX, indicating a targeted and effective approach to treatment. The apoptotic pathways activated by DTX@C8-MSN, evidenced by the increased levels of cleaved caspase-3 and PARP, point to a potent therapeutic mechanism. Collectively, the results advocate DTX@C8-MSN as a promising candidate for targeted therapy in TMZ-resistant GBM, optimizing drug delivery and bioavailability to overcome current therapeutic limitations.


Asunto(s)
Barrera Hematoencefálica , Docetaxel , Resistencia a Antineoplásicos , Glioblastoma , Nanopartículas , Dióxido de Silicio , Temozolomida , Temozolomida/química , Temozolomida/farmacología , Temozolomida/uso terapéutico , Temozolomida/farmacocinética , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/metabolismo , Docetaxel/química , Docetaxel/farmacología , Docetaxel/farmacocinética , Docetaxel/uso terapéutico , Dióxido de Silicio/química , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Animales , Nanopartículas/química , Humanos , Ratones , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Porosidad , Portadores de Fármacos/química , Ratones Desnudos , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos
4.
EMBO Mol Med ; 16(2): 416-428, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38225455

RESUMEN

The tumor microenvironment (TME) presents differential selective pressure (DSP) that favors the growth of cancer cells, and monovalent therapy is often inadequate in reversing the cancer cell dominance in the TME. In this work, we introduce bacteria as a foreign species to the TME and explore combinatorial treatment strategies to alter DSP for tumor eradication. We show that cancer-selective chemotherapeutic agents and fasting can provide a strong selection pressure against tumor growth in the presence of bacteria. Moreover, we show that an immunogenic drug (oxaliplatin), but not a non-immunogenic one (5-FU), synergizes with the bacteria to activate both the innate and adaptive immunity in the TME, resulting in complete tumor remission and a sustained anti-tumor immunological memory in mice. The combination of oxaliplatin and bacteria greatly enhances the co-stimulatory and antigen-presenting molecules on antigen-presenting cells, which in turn bridge the cytotoxic T cells for cancer-cell killing. Our findings indicate that rational combination of bacterial therapy and immunogenic chemotherapy can promote anticancer immunity against the immunosuppressive TME.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Ratones , Oxaliplatino/uso terapéutico , Microambiente Tumoral , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Linfocitos T Citotóxicos , Inmunoterapia/métodos , Línea Celular Tumoral
5.
ChemSusChem ; 15(8): e202200188, 2022 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-35243793

RESUMEN

Using renewable energy to convert CO2 into liquid products, as a sustainable way to produce fuels and chemicals, has attracted intense attention. Herein, a novel heterostructured photocathode composed of Si wafer, TiO2 layer, and Sn metal particles has been successfully fabricated by combining of a facile hydrothermal and electrodeposition method. The obtained Sn/TiO2 /Si photocathode shows enhanced light absorption performance by the surface plasmon resonance effect of Sn metal. Especially, the Sn/TiO2 /Si photocathode together with rich oxygen vacancy defects jointly promote photoelectrochemical CO2 reduction, harvesting a high faradaic efficiency of HCOOH and a desirable average current density (-4.72 mA cm-2 ) at -1.0 V vs. reversible hydrogen electrode. Significantly, the photocathode Sn/TiO2 /Si also shows good stability due to the design of protecting layer TiO2 . This study provides a facile strategy of constructing an efficient photocathode to improve the light absorption performance and the electron transfer efficiency, exhibiting great potential in the CO2 reduction.

6.
Pharmaceutics ; 13(7)2021 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-34371758

RESUMEN

The efficient and safe delivery of therapeutic drugs, proteins, and nucleic acids are essential for meaningful therapeutic benefits. The field of nanomedicine shows promising implications in the development of therapeutics by delivering diagnostic and therapeutic compounds. Nanomedicine development has led to significant advances in the design and engineering of nanocarrier systems with supra-molecular structures. Smart mesoporous silica nanoparticles (MSNs), with excellent biocompatibility, tunable physicochemical properties, and site-specific functionalization, offer efficient and high loading capacity as well as robust and targeted delivery of a variety of payloads in a controlled fashion. Such unique nanocarriers should have great potential for challenging biomedical applications, such as tissue engineering, bioimaging techniques, stem cell research, and cancer therapies. However, in vivo applications of these nanocarriers should be further validated before clinical translation. To this end, this review begins with a brief introduction of MSNs properties, targeted drug delivery, and controlled release with a particular emphasis on their most recent diagnostic and therapeutic applications.

7.
Adv Biol (Weinh) ; 5(1): e2000200, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33724730

RESUMEN

The effect of the intracellular pH of macrophages after taking up biodegradable polymer nanoparticles (NPs) on immunomodulating functions has not been explored so far. Previous studies have demonstrated that biodegradable polyurethane (PU) NPs exhibit immunosuppressive activity. Yet, the intracellular mechanism is not clearly understood. In this study, a uniquely designed pH nanosensor is employed for tracking the intracellular pH value of macrophages to reveal the intracellular journey of PU NPs and to clarify the intracellular pH effect on the corresponding inflammatory response. First, fluorescent mesoporous silica nanoparticles (FRMSNs) is used to detect the pH change in macrophages after endo/phagocytosis of PU NPs. Second, PU is coated on the external surface of FRMSNs to examine the intracellular trafficking process of PU in the macrophages. The results show that the majority of PU-coated FRMSNs remain to stay at the cytosol-early endosome/phagosome regions. The intracellular pH value and other supporting results show that the immune response of PU NPs may be correlated to their internalization journey. The retardation in the degradation process of the PU NPs may intervene with the lysosome activity and repress the immunostimulatory effect, which contributes to the low immune response of PU NPs.


Asunto(s)
Nanopartículas , Poliuretanos , Humanos , Concentración de Iones de Hidrógeno , Inflamación , Macrófagos , Fagosomas
8.
Microsc Microanal ; 27(2): 420-424, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33487212

RESUMEN

Lysosomes are integration hubs for several signaling pathways, such as autophagy and endocytosis, and also crucial stores of ions, including Zn2+. Lysosomal dysfunction caused by changes in their morphology by fusion and fission processes can result in several pathological disorders. However, the role of Zn2+ in modulating the morphology of lysosomes is unclear. The resolution of conventional epifluorescence microscopy restricts accurate observation of morphological changes of subcellular fluorescence punctum. In this study, we used a modified epifluorescence microscopy to identify the center of a punctum from a series of z-stack images and calculate the morphological changes. We stained primary cultured rat embryonic cortical neurons with FluoZin3, a Zn2+-sensitive fluorescent dye, and Lysotracker, a lysosome-specific marker, to visualize the distribution of Zn2+-enriched vesicles and lysosomes, respectively. Our results revealed that treating neurons with N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine, a cell-permeable Zn2+ chelator, shrank Zn2+-enriched vesicles and lysosomes by up to 25% in an hour. Pretreating the neurons with YM201636, a blocker of lysosome fission, could suppress this shrinkage. These results demonstrate the usefulness of the modified epifluorescence microscopy for investigating the homeostasis of intracellular organelles and related disorders.


Asunto(s)
Lisosomas , Neuronas , Animales , Autofagia , Células Cultivadas , Ratas , Zinc
9.
J Colloid Interface Sci ; 584: 647-659, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33198979

RESUMEN

HYPOTHESIS: Multistage silicate self-organization into light-weight, high-strength, hierarchically patterned diatom frustules carries hints for innovative silica-based nanomaterials. With sodium silicate in a biomimetic sol-gel system templated by a tri-surfactant system of hexadecyltrimethylammonium bromide, sodium dodecylsulfate, and poly(oxyethylene-b-oxypropylene-b-oxyethylene) (P123), mesoporous silica nanochannel plates with perpendicular channel orientation are synthesized. The formation process, analogous to that of diatom frustules, is postulated to be directed by an oriented self-assembly of the block copolymer micelles shelled with charged catanionic surfactants upon silication. EXPERIMENTS: The postulated formation process for the oriented silica nanochannel plates was investigated using time-resolved small-angle X-ray and neutron scattering (SAXS/SANS) and freeze fracture replication transmission electron microscopy (FFR-TEM). FINDINGS: With fine-tuned molar ratios of the anionic, cationic, and nonionic surfactants, the catanionic combination and the nonionic copolymer form charged, prolate ternary micelles in aqueous solutions, which further develop into prototype monolayered micellar plates. The prolate shape and maximized surfactant adsorption of the complex micelles, revealed from combined SAXS/SANS analysis, are of critical importance in the subsequent micellar self-assembly upon silicate deposition. Time-resolved SAXS and FFR-TEM indicate that the silicate complex micelles coalesce laterally into the prototype micellar nanoplates, which further fuse with one another into large sheets of monolayered silicate micelles of in-plane lamellar packing. Upon silica polymerization, the in-plane lamellar packing of the micelles further transforms to 2D hexagonal packing of vertically oriented silicate channels. The unveiled structural features and their evolution not only elucidate the previously unresolved self-assembly process of through-thickness silica nanochannels but also open a new line of research mimicking free-standing frustules of diatoms.

10.
Biomedicines ; 9(1)2020 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-33374319

RESUMEN

Mesoporous silica nanoparticles (MSNs) have emerged as a prominent nanomedicine platform, especially for tumor-related nanocarrier systems. However, there is increasing concern about the ability of nanoparticles (NPs) to penetrate solid tumors, resulting in compromised antitumor efficacy. Because the physicochemical properties of NPs play a significant role in their penetration and accumulation in solid tumors, it is essential to systematically study their relationship in a model system. Here, we report a multihierarchical assessment of the accumulation and penetration of fluorescence-labeled MSNs with nine different physicochemical properties in tumor spheroids using two-photon microscopy. Our results indicated that individual physicochemical parameters separately could not define the MSNs' ability to accumulate in a deeper tumor region; their features are entangled. We observed that the MSNs' stability determined their success in reaching the hypoxia region. Moreover, the change in the MSNs' penetration behavior postprotein crowning was associated with both the original properties of NPs and proteins on their surfaces.

11.
ACS Appl Mater Interfaces ; 12(51): 56741-56752, 2020 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-33305564

RESUMEN

Reversing the immunosuppressive tumor microenvironment (TME) is a strategic initiative to sensitize cancer immunotherapy. Emerging evidence shows that cyclic diguanylate monophosphate (c-di-GMP or cdG) can induce the stimulator of interferon genes (STING) pathway activation of antigen-presenting cells (APCs) and upregulate expression of type I interferons (IFNs) to enhance tumor immunogenicity. In vitro anionic cdG revealed fast plasma clearance, poor membrane permeability, and inadequate cytosolic bioavailability. Therefore, we explored a comprehensive "in situ vaccination" strategy on the basis of nanomedicine to trigger robust antitumor immunity. Rhodamine B isothiocyanate (RITC) fluorescent mesoporous silica nanoparticles (MSN) synthesized and modified with poly(ethylene glycol) (PEG) and an ammonium-based cationic molecule (TA) were loaded with negatively charged cdG via electrostatic interactions to form cdG@RMSN-PEG-TA. Treatment of RAW 264.7 cells with cdG@RMSN-PEG-TA markedly stimulated the secretion of IL-6, IL-1ß, and IFN-ß along with phospho-STING (Ser365) protein expression. In vivo cdG@RMSN-PEG-TA enhanced infiltration of leukocytes, including CD11c+ dendritic cells, F4/80+ macrophages, CD4+ T cells, and CD8+ T cells within the tumor microenvironment (TME), resulting in dramatic tumor growth inhibition in 4T1 breast tumor-bearing Balb/c mice. Our findings suggest that a nanobased platform can overcome the obstacles bare cdG can face in the TME. Our approach of an in situ vaccination using a STING agonist provides an attractive immunotherapy-based strategy for treating breast cancer.


Asunto(s)
Neoplasias de la Mama/terapia , GMP Cíclico/análogos & derivados , Activación de Linfocitos/efectos de los fármacos , Proteínas de la Membrana/agonistas , Nanopartículas/química , Dióxido de Silicio/química , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , GMP Cíclico/farmacología , Femenino , Colorantes Fluorescentes/química , Inmunoterapia/métodos , Ratones , Ratones Endogámicos BALB C , Porosidad , Células RAW 264.7 , Rodaminas/química , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/inmunología
12.
Front Chem ; 8: 734, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33005606

RESUMEN

We investigate dynamics of water (H2O) and methanol (CH3OH and CH3OD) inside mesoporous silica materials with pore diameters of 4.0, 2.5, and 1.5 nm using low-field (LF) nuclear magnetic resonance (NMR) relaxometry. Experiments were conducted to test the effects of pore size, pore volume, type of fluid, fluid/solid ratio, and temperature on fluid dynamics. Longitudinal relaxation times (T1) and transverse relaxation times (T2) were obtained for the above systems. We observe an increasing deviation in confined fluid behavior compared to that of bulk fluid with decreasing fluid-to-solid ratio. Our results show that the surface area-to-volume ratio is a critical parameter compared to pore diameter in the relaxation dynamics of confined water. An increase in temperature for the range between 25 and 50°C studied did not influence T2 times of confined water significantly. However, when the temperature was increased, T1 times of water confined in both silica-2.5 nm and silica-1.5 nm increased, while those of water in silica-4.0 nm did not change. Reductions in both T1 and T2 values as a function of fluid-to-solid ratio were independent of confined fluid species studied here. The parameter T1/T2 indicates that H2O interacts more strongly with the pore walls of silica-4.0 nm than CH3OH and CH3OD.

13.
ACS Appl Mater Interfaces ; 12(38): 42472-42484, 2020 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-32657564

RESUMEN

Nanoparticle (NP)-based targeted drug delivery is intended to transport therapeutically active molecules to specific cells and particular intracellular compartments. However, there is limited knowledge regarding the complete route of NPs in this targeting scenario. In this study, simultaneously performing motion and dynamic pH sensing using single-particle tracking (SPT) leads to an alternative method of gaining insights into the mesoporous silica nanoparticle's (MSN) journey in targeting lysosome. Two different pH-sensitive dyes and a reference dye are incorporated into mesoporous silica nanoparticles (MSNs) via co-condensation to broaden the measurable pH range (pH 4-7.5) of the nanoprobe. The phosphonate, amine, and lysosomal sorting peptides (YQRLGC) are conjugated onto the MSN's surface to study intracellular nano-biointeractions of two oppositely charged and lysosome-targetable MSNs. The brightness and stability of these MSNs allow their movement and dynamic pH evolution during their journey to be simultaneously monitored in real time. Importantly, a multidimensional analysis of MSN's movement and local pH has revealed new model intracellular dynamic states and distributions of MSNs, previously inaccessible when using single parameters alone. A key result is that YQRLGC-conjugated MSNs took an alternative route to target lysosomes apart from the traditional one, which sped up to 4 h and enhanced their targeting efficiency (up to 32%). The findings enrich our understanding of the intracellular journey of MSNs. This study offers complementary information on correlating the surface design with the full pathway of nanoparticles to achieve targeted delivery of therapeutic payload.


Asunto(s)
Lisosomas/química , Nanopartículas/química , Dióxido de Silicio/química , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Tamaño de la Partícula , Porosidad , Propiedades de Superficie , Células Tumorales Cultivadas
14.
Biomaterials ; 246: 119997, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32247937

RESUMEN

Transcription factor complex NF-κB (p65/p50) is localized to the cytoplasm by its inhibitor IκBα. Upon activation, the Rel proteins p65/p50 are released from IκBα and transported through nuclear pore to affect many gene expressions. While inhibitions of up or down stream signal pathways are often ineffective due to crosstalks and compensations, direct blocking of the Rel proteins p65/p50 has long been proposed as a potential target for cancer therapy. In this work, a nanoparticle/antibody complex targeting NF-κB is employed to catch the Rel protein p65 in perinuclear region and thus blocking the translocation near the nuclear pore gate. TAT peptide conjugated on mesoporous silica nanoparticles (MSN) help non-endocytosis cell-membrane transducing and converge toward perinuclear region, where the p65 specific antibody performed the targeting and catching against active NF-κB p65 effectively. The size of the p65 bound nanoparticle becomes too big to enter nucleus. Simultaneous treatment of mice with the hybrid MSN and doxorubicin conferred a significant therapeutic effect against 4T1 tumor-bearing mice. The new approach of anti-body therapy targeting on transcription factor with "nucleus focusing" and "size exclusion blocking" effects of the antibody-conjugated nanoparticle is general and may be applicable to modulating other transcription factors.


Asunto(s)
FN-kappa B , Nanopartículas , Transporte Activo de Núcleo Celular , Animales , Ratones , FN-kappa B/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/metabolismo
15.
ACS Appl Mater Interfaces ; 12(6): 7459-7465, 2020 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-31961650

RESUMEN

We report on our use of a thin-layered vertical mesoporous silica thin film (MSTF) with tunable pore size overlaid on an anodic aluminum oxide (AAO) membrane for advancing water purification. The features of ultrathin thickness (about 20 nm), a uniform vertical pore orientation, low tortuosity, high porosity, and a hydrophilic surface endow the MSTF membranes with ultrahigh water permeability compared with that of state-of-the-art membranes. The modified E-MSTF membrane with a small pore diameter of 2.1 ± 0.1 nm demonstrates superior nanofiltration performance for dye molecules with a cutoff of 520 Da and ultrahigh water permeability of 310 ± 8 L m-2 h-1 bar-1. Furthermore, the precise molecular sieving of dye/salt mixtures was realized with outstanding salt permeation (97.5% NaCl, 96.0% Na2SO4) and a high retention of dye (99.0%). The water permeance and selectivity of the modified E-MSTF membrane are higher than that of reported membranes with similar dye rejections. This work opens up new avenues for constructing tailor-made membranes with tunable pore size and remarkable separation performance.

16.
Sci Rep ; 9(1): 11457, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31391525

RESUMEN

Transgenic genome integration using non-viral vehicles is a promising approach for gene therapy. Previous studies reported that asparagine is a key regulator of cancer cell amino acid homeostasis, anabolic metabolism and cell proliferation. The depletion of asparagine would inhibit the growth of many cancer cells. In this study, we develop a nanoparticle delivery system to permanently integrate the asparaginase gene into the genome of human lung adenocarcinoma cells. The asparaginase plasmid and the Sleeping Beauty plasmid were co-transfected using amine-functionalized mesoporous nanoparticles into the human lung adenocarcinoma cells. The intracellular asparaginase expression led to the cell cytotoxicity for PC9 and A549 cells. In addition, the combination of the chemotherapy and the asparaginase gene therapy additively enhanced the cell cytotoxicity of PC9 and A549 cells to 69% and 63%, respectively. Finally, we showed that the stable cell clones were successfully made by puromycin selection. The doxycycline-induced expression of asparaginase caused almost complete cell death of PC9 and A549 asparaginase-integrated stable cells. This work demonstrates that silica-based nanoparticles have great potential in gene delivery for therapeutic purposes.


Asunto(s)
Antineoplásicos/farmacología , Asparaginasa/genética , Terapia Genética/métodos , Neoplasias/terapia , Transposasas/genética , Células A549 , Antineoplásicos/uso terapéutico , Asparaginasa/metabolismo , Asparagina/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Cisplatino/farmacología , Cisplatino/uso terapéutico , Terapia Combinada , Elementos Transponibles de ADN/genética , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Portadores de Fármacos/química , Técnicas de Transferencia de Gen , Humanos , Nanopartículas/química , Neoplasias/metabolismo , Plásmidos/administración & dosificación , Plásmidos/genética , Dióxido de Silicio/química , Transfección , Transposasas/administración & dosificación
17.
ACS Appl Mater Interfaces ; 11(22): 19808-19818, 2019 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-31066542

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most prevalent and deadly malignancies characterized by high rate of recurrence. Tumor recurrence is often attributed to the presence of a subpopulation of cells with stem cell properties, referred to as cancer stem cells (CSCs). Traditionally, cancer therapies target the entire bulk of tumor cells; however, they are poorly effective against CSCs, characterized by higher drug resistance. Therefore, approaches targeting CSCs may be required in addition to conventional chemotherapy to prevent tumor recurrence. In this study, we investigated an approach to target HCC by combining the conventional chemotherapeutic drug, cisplatin, to target the bulk of tumor cells, and differentiation therapy by delivering the gene encoding HNF4α, an important regulator of hepatocyte differentiation, to target CSCs. We used the Huh7 cell line as an in vitro model of HCC, which is characterized by a high proportion of CD133-expressing CSCs. By using flow cytometry, we separated CD133+ and CD133- Huh7 cell subpopulations and have shown that the former has highly pronounced in vivo tumorigenic capacity in contrast to the latter, which could not generate tumors in vivo. For the dual delivery of HNF4α-encoding plasmid and cisplatin, we used polyethyleneimine-modified mesoporous silica nanoparticles (PMSNs) as the nanocarriers. Here, we show that the treatment of CD133-expressing Huh7 cells with HNF4α-loaded PMSNs can suppress their proliferation rate, decrease the proportion of CSCs, downregulate stemness-associated genes, and increase the expression of mature hepatocyte-associated genes. At the same time, the treatment of Huh7 with PMSNs loaded with both HNF4α-encoding plasmid and cisplatin could block them in the S-phase of the cell cycle and cause apoptosis. In addition, dually loaded PMSNs were the most efficient formulation in suppressing tumor growth in vivo. To summarize, in this study, we tested the nanoparticle-based delivery system as both chemotherapy and gene-based therapy agents, which has great potential for development of effective treatment of HCC.


Asunto(s)
Antígeno AC133/metabolismo , Carcinoma Hepatocelular/metabolismo , Cisplatino/administración & dosificación , Cisplatino/farmacología , Factor Nuclear 4 del Hepatocito/metabolismo , Nanopartículas/química , Dióxido de Silicio/química , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Animales , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Citometría de Flujo , Factor Nuclear 4 del Hepatocito/genética , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Ratones SCID , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
ACS Appl Mater Interfaces ; 11(17): 15322-15331, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30986029

RESUMEN

Reactive oxygen species (ROS)-induced oxidative stress leads to neuron damage and is involved in the pathogenesis of chronic inflammation in neurodegenerative diseases (NDs), such as Alzheimer's, Parkinson's, and amyotrophic lateral sclerosis. Researchers, therefore, are looking for antiinflammatory drugs and gene therapy approaches to slow down or even prevent neurological disorders. Combining therapeutics has shown a synergistic effect in the treatment of human diseases. Many nanocarriers could be designed for the simultaneous codelivery of drugs with genes to fight diseases. However, only a few researches have been performed in NDs. In this study, we developed a mesoporous silica nanoparticle (MSN)-based approach for neurodegenerative therapy. This MSN-based platform involved multiple designs in the targeted codelivery of (1) curcumin, a natural antioxidant product, to protect ROS-induced cell damage and (2) plasmid RhoG-DsRed, which is associated with the formation of lamellipodia and filopodia for promoting neurite outgrowth. At the same time, TAT peptide was introduced to the plasmid RhoG-DsRed via electrostatic interaction to elevate the efficiency of nonendocytic pathways and the nuclear plasmid delivery of RhoG-DsRed in cells for enhanced gene expression. Besides, such a plasmid RhoG-DsRed/TAT complex could work as a noncovalent gatekeeper. The release of curcumin inside the channel of the MSN could be triggered when the complex was dissociated from the MSN surface. Taken together, this MSN-based platform combining genetic and pharmacological manipulations of an actin cytoskeleton as well as oxidative stress provides an attractive way for ND therapy.


Asunto(s)
Curcumina/farmacología , Portadores de Fármacos/química , Nanopartículas/química , Proyección Neuronal/efectos de los fármacos , Plásmidos/metabolismo , Dióxido de Silicio/química , Citoesqueleto de Actina/efectos de los fármacos , Animales , Línea Celular Tumoral , Curcumina/química , GTP Fosfohidrolasas/genética , Ratones , Estrés Oxidativo , Tamaño de la Partícula , Fragmentos de Péptidos/química , Plásmidos/química , Porosidad , Especies Reactivas de Oxígeno/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/química
19.
ACS Appl Mater Interfaces ; 11(5): 4790-4798, 2019 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-30624037

RESUMEN

Mesoporous silica nanoparticles (MSNs) hold great potential as a versatile platform for biomedical applications, especially drug delivery. However, evidence shows that MSNs even when PEGylated are rapidly cleared from the bloodstream by the monocyte phagocytic system. Erythrocytes, also called red blood cells (RBCs), can serve as biocompatible carriers of various bioactive substances, including drugs, enzymes, and peptides. In this work, we synthesize a series of fluorescent PEGylated MSNs with different synthetic diameters ranging from 10 to 200 nm and investigate the size effect on their encapsulation in human RBCs (hRBCs) by a hypotonic dialysis-based method. According to fluorescence images and flow cytometry analyses, we demonstrated that a hydrodynamic diameter below 30 nm is critical for efficient MSN encapsulation. Confocal microscopy and scanning electron microscopy images further confirmed that PEGylated MSNs were successfully embedded inside RBC. PEGylation serves an important role not only for stabilizing MSNs in biological milieu but also for reducing significant hemolysis caused by bare MSNs and thus for successful encapsulation. In addition to PEGylation, we further introduce positively charged functional groups onto the MSNs to show that nanoparticle-encapsulated hRBCs could serve as depots for delivering biological molecules through electrostatic attraction or chemical conjugation with MSNs. Also, we verify the existence of CD47 membrane protein, a marker of self, on the nanoparticle-encapsulated hRBCs and assess its ability of circulation in the blood, which could act as a circulation reservoir for delivering pharmacological substances through an osmosis-based method with MSNs.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Eritrocitos/metabolismo , Nanopartículas/química , Dióxido de Silicio/química , Animales , Antígeno CD47/sangre , Antígeno CD47/metabolismo , Eritrocitos/química , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Hemólisis/efectos de los fármacos , Humanos , Ratones , Ratones SCID , Microscopía Confocal , Nanopartículas/toxicidad , Polietilenglicoles/química , Dióxido de Silicio/farmacocinética
20.
Enzymes ; 43: 123-153, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30244805

RESUMEN

The unique features of Mesoporous Silica Nanoparticles (MSNs) provide a suitable platform to carry fluorescence dyes for various bioimaging applications. Several strategies have been developed to conjugate a variety of dyes either in the pores or on the surfaces of MSNs to form the fluorescence MSNs (FMSNs). In this chapter, we will discuss recent research progress and future development of FMSNs for living system imaging. We will first describe different strategies for the fabrications of FMSNs. Then, we will discuss the recent developments of cellular and intracellular imaging including self-probe for the interactions of FMSNs with the cells, receptor and organelle labeling, sensing and tracking of biological system, and monitoring the drug delivery and release processes. Moreover, we will include the applications of FMSNs as contrast agents for in vivo imaging. Finally, we will conclude and highlight the challenges and opportunities for MSNs in medical applications.


Asunto(s)
Sistemas de Liberación de Medicamentos , Colorantes Fluorescentes/administración & dosificación , Imagen Molecular/métodos , Nanopartículas/administración & dosificación , Dióxido de Silicio/administración & dosificación , Medios de Contraste/administración & dosificación , Medios de Contraste/química , Colorantes Fluorescentes/análisis , Humanos , Nanopartículas/química , Porosidad , Dióxido de Silicio/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA