Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Bioorg Chem ; 130: 106199, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36370648

RESUMEN

Due to the diverse H2O2 distribution in organelles, fluorescent probes were usually required to be prepared separately, which limited the convenience and practicability. Herein, we reported a flexible strategy to in-situ construct H2O2 fluorescent probes in different organelles. A tetrazine fused probe TP was developed with rapid click reaction capacity and sensitive H2O2 response. When treated with H2O2, the turn-on fluorescence was effectively quenched by the tetrazine part. Only after click reaction with dienophiles, the fluorescence resumed. In application, cells were firstly treated with triphenylphosphorus tagged norbornene (TPP-NB) to label mitochondria, which was followed by the introduction of probe TP to trigger click reaction. The in-situ constructed probe P1 served as a local H2O2 sensor. In a similar way, probe P2 was in-situ constructed in lysosomes via probe TP and morpholine tagged norbornene (MP-NB). With this on-demand modular assembling and double turn-on features, our strategy to construct fluorescent probes presented high flexibility and anti-interference performance, which was expected to inspired more applications in biological studies.


Asunto(s)
Colorantes Fluorescentes , Peróxido de Hidrógeno , Humanos , Colorantes Fluorescentes/metabolismo , Peróxido de Hidrógeno/metabolismo , Células HeLa , Lisosomas/metabolismo , Mitocondrias , Norbornanos/metabolismo
2.
Ecotoxicol Environ Saf ; 245: 114108, 2022 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-36174319

RESUMEN

CuInS2/ZnS-PEG quantum dots (QDs) are among the most widely used near infrared non-cadmium QDs and are favored because of their non-cadmium content and strong tissue penetration. However, with their increasing use, there is great concern about whether exposure to QDs is potentially risky to the environment and humans. Furthermore, toxicological data related to CuInS2/ZnS-PEG QDs are scarce. In the study, we found that CuInS2/ZnS-PEG QDs (0-100 µg/mL) could internalize into human LAD2 mast cells without affecting their survival rate, nor did it cause degranulation or release of IL-8 and TNF-α. However, CuInS2/ZnS-PEG QDs significantly inhibited Substance P (SP) and LL-37-induced degranulation and chemotaxis of LAD2 cells by inhibiting calcium mobilization. Lower concentrations of CuInS2/ZnS-PEG QDs promoted the release of TNF-α and IL-8 stimulated by SP, but higher concentrations of CuInS2/ZnS-PEG QDs significantly inhibited the release of TNF-α and IL-8. On the other hand, CuInS2/ZnS-PEG QDs promoted LL-37-mediated TNF-α release from LAD2 cells in a dose-dependent manner from 6.25 to 100 µg/mL, while release of IL-8 triggered by LL-37 was dose-dependently inhibited within a dose concentration of 12.5-100 µg/mL. Collectively, our data demonstrated that CuInS2/ZnS-PEG QDs differentially mediated human mast cell activation induced by SP and LL-37.


Asunto(s)
Puntos Cuánticos , Calcio , Trastornos Congénitos de Glicosilación , Cobre , Humanos , Interleucina-8 , Mastocitos , Polietilenglicoles , Puntos Cuánticos/toxicidad , Sustancia P , Sulfuros/farmacología , Factor de Necrosis Tumoral alfa , Compuestos de Zinc/toxicidad
3.
Ecotoxicol Environ Saf ; 211: 111946, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33493718

RESUMEN

Increased applications of quantum dots (QDs) in the biomedical field have aroused attention for their potential toxicological effects. Although numerous studies have been carried out on the toxicity of QDs, their effects on reproductive and development are still unclear. In this study, we systematically evaluated the male reproductive toxicity and developmental toxicity of CdSe/ZnS QDs in BALB/c mice. The male mice were injected intravenously with CdSe/ZnS QDs at the dosage of 2.5 mg/kg BW or 25 mg/kg BW, respectively, and the survival status, biodistribution of QDs in testes, serum sex hormone levels, histopathology, sperm motility and acrosome integrity was measured on Day 1, 7, 14, 28 and 42 after injection. On Day 35 after treatment, male mice were housed with non-exposed female mice, and then offspring number, body weight, organ index and histopathology of major organs, blood routine and biochemical tests of offspring were measured to evaluate the fertility and offspring health. The results showed that CdSe/ZnS QDs could rapidly distribute in the testis, and the fluorescence of QDs could still be detected on Day 42 post-injection. QDs had no adverse effect on the structure of testis and epididymis, but high-dose QDs could induce apoptosis of Leydig cells in testis at an early stage. No significant differences in survival of state, body weight organ index of testis and epididymis, sex hormones levels, sperm quality, sperm acrosome integrity and fertility of male mice were observed in QDs exposed groups. However, the development of offspring was obviously influenced, which was mainly manifested in the slow growth of offspring, changes in organ index of main organs, and the abnormality of liver and kidney function parameters. Our findings revealed that CdSe/ZnS QDs were able to cross the blood-testis barrier (BTB), produce no discernible toxic effects on the male reproductive system, but could affect the healthy growth of future generations to some extent. In view of the broad application prospect of QDs in biomedical fields, our findings might provide insight into the biological safety evaluation of the reproductive health of QDs.


Asunto(s)
Puntos Cuánticos/toxicidad , Acrosoma , Animales , Compuestos de Cadmio/química , Compuestos de Cadmio/toxicidad , Epidídimo , Femenino , Fertilidad , Masculino , Ratones , Ratones Endogámicos BALB C , Puntos Cuánticos/química , Reproducción , Compuestos de Selenio/farmacología , Motilidad Espermática , Espermatozoides , Sulfuros/toxicidad , Testículo , Distribución Tisular , Pruebas de Toxicidad , Compuestos de Zinc/toxicidad
4.
Ecotoxicol Environ Saf ; 207: 111378, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33022524

RESUMEN

The widespread application of cadmium-free CuInS2/ZnS QDs has raised great concern regarding their potential toxicity to humans. To date, toxicological data related to CuInS2/ZnS QDs are scarce. Neurons play extraordinary roles in regulating the activities of organs and systems, and serious consequences occur when neurons are damaged. Currently, the potential toxicity of CuInS2/ZnS QDs on neurons has not been fully elucidated. Here, we investigate the neurotoxicity of PEGylated CuInS2/ZnS (CuInS2/ZnS-PEG) QDs on neuron-like PC12 cells. We found that CuInS2/ZnS-PEG QDs were taken up by PC12 cells, but at a concentration range from 0 to 100 µg/mL, they did not affect the survival rate of the PC12 cells. In addition, we found that CuInS2/ZnS-PEG QDs significantly inhibited neurite outgrowth from and the differentiation of PC12 cells in the presence of NGF, while COOH-modified CuInS2/ZnS QDs or free PEG did not have a similar effect. Further studies showed that CuInS2/ZnS-PEG QDs obviously downregulated the expression of low-affinity NGF receptor (p75NTR) and subsequently negatively regulated the downstream MAPK cascade by dephosphorylating ERK1/2 and AKT. Taken together, these results suggest that CuInS2/ZnS-PEG QDs disturb NGF signal transduction from external stimuli to relevant internal signals, thus affecting normal biological processes such as neurite outgrowth and cell differentiation.


Asunto(s)
Proyección Neuronal/efectos de los fármacos , Puntos Cuánticos/toxicidad , Animales , Cadmio/farmacología , Regulación hacia Abajo/efectos de los fármacos , Humanos , Proteína Quinasa 3 Activada por Mitógenos , Factor de Crecimiento Nervioso , Proteínas del Tejido Nervioso , Células PC12 , Ratas , Receptores de Factor de Crecimiento Nervioso , Transducción de Señal/efectos de los fármacos , Sulfuros , Pruebas de Toxicidad , Compuestos de Zinc
5.
Anticancer Drugs ; 31(10): 989-996, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32694422

RESUMEN

Colorectal cancer (CRC) is the third most common malignancy worldwide. Our previous studies have shown that combinatorial treatment with thioridazine and loratadine may effectively inhibit CRC. However, the translation of these research findings to clinical practice was impaired by issues related to a lack of therapeutic specificity and to immune evasion. Toll-like receptor (TLR) agonists have been used as adjuvants to enhance the effectiveness of cancer vaccines. The aim of this study was to evaluate the therapeutic efficiency of immunotherapy with thioridazine and loratadine in combination with resiqumiod (R848), a small-molecule TLR7 agonist, in suppressing CRC growth in a mouse model. Twenty-four BALB/c mice were randomly assigned to treatment with PBS, R848, thioridazine + loratadine, or thioridazine + loratadine + R848. Cytokine levels were measured with ELISA. Overall survival, as well as tumor volume and tumor weight, was recorded. Cytotoxicity was measured by counting the numbers of CD8 and CD3-positive (CD8CD3) or CD4 and CD3-positive (CD3CD4) T-cells. The immune response induced by cytokines (as interferon-γ, interleukin-6, and tumor necrosis factor-α) was significantly stronger in mice treated with thioridazine + loratadine + R848. Moreover, thioridazine + loratadine + R848 significantly delayed tumor development and prolonged survival, which was associated with enhanced immune response and dendritic cell maturation. This study suggested that thioridazine + loratadine + R848 combinatorial treatment may be effective in overcoming immune evasion by tumor cells, with promising therapeutic potential in CRC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Imidazoles/administración & dosificación , Glicoproteínas de Membrana/agonistas , Linfocitos T Citotóxicos/efectos de los fármacos , Receptor Toll-Like 7/agonistas , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Loratadina/administración & dosificación , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos BALB C , Linfocitos T Citotóxicos/inmunología , Tioridazina/administración & dosificación , Receptor Toll-Like 7/metabolismo
6.
Int J Mol Sci ; 21(19)2020 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-32992627

RESUMEN

InP QDs have shown a great potential as cadmium-free QDs alternatives in biomedical applications. It is essential to understand the biological fate and toxicity of InP QDs. In this study, we investigated the in vivo renal toxicity of InP/ZnS QDs terminated with different functional groups-hydroxyl (hQDs), amino (aQDs) and carboxyl (cQDs). After a single intravenous injection into BALB/c mice, blood biochemistry, QDs distribution, histopathology, inflammatory response, oxidative stress and apoptosis genes were evaluated at different predetermined times. The results showed fluorescent signals from QDs could be detected in kidneys during the observation period. No obvious changes were observed in histopathological detection or biochemistry parameters. Inflammatory response and oxidative stress were found in the renal tissues of mice exposed to the three kinds of QDs. A significant increase of KIM-1 expression was observed in hQDs and aQDs groups, suggesting hQDs and aQDs could cause renal involvement. Apoptosis-related genes (Bax, Caspase 3, 7 and 9) were up-regulated in hQDs and aQDs groups. The above results suggested InP/ZnS QDs with different surface chemical properties would cause different biological behaviors and molecular actions in vivo. The surface chemical properties of QDs should be fully considered in the design of InP/ZnS QDs for biomedical applications.


Asunto(s)
Indio/química , Indio/toxicidad , Riñón/efectos de los fármacos , Fosfinas/química , Fosfinas/toxicidad , Puntos Cuánticos/química , Puntos Cuánticos/toxicidad , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Dióxido de Carbono/química , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Radical Hidroxilo/química , Indio/administración & dosificación , Indio/farmacocinética , Inflamación/inducido químicamente , Inyecciones Intravenosas , Riñón/metabolismo , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos , Fosfinas/administración & dosificación , Fosfinas/farmacocinética , Puntos Cuánticos/administración & dosificación , Sulfuros/administración & dosificación , Sulfuros/química , Sulfuros/farmacocinética , Sulfuros/toxicidad , Propiedades de Superficie , Distribución Tisular , Compuestos de Zinc/administración & dosificación , Compuestos de Zinc/química , Compuestos de Zinc/farmacocinética , Compuestos de Zinc/toxicidad
7.
J Microencapsul ; 36(6): 552-565, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31403342

RESUMEN

Taxane-based chemotherapy-loaded drug delivery systems have great potential for cancer treatment. The docetaxel (DTX)-loaded PAMAM-based poly (γ-benzyl-l-glutamate)-b-d-α-tocopheryl polyethylene glycol 1000 succinate (PAM-PBLG-b-TPGS) nanoparticles and the docetaxel (DTX)-loaded PAMAM-based poly (γ-benzyl-l-glutamate) (PAM-PBLG) nanoparticles were designed using a modified nanoprecipitation method. The particle size, encapsulation efficiency (EE), and in vitro release characteristics of the nanoparticles were tested. The effects of the two nanoparticles on the cellular uptake and cell viability on human cervical cancer cell line Hela and the human breast cancer cell line MCF-7 were compared. Furthermore, their antitumor efficiency was evaluated through in vivo tumour growth experiment in comparison with free DTX. PAM-PBLG-b-TPGS nanoparticles displayed high EE, smaller diameter, and a nice releasing profile. Besides, based on the high EE and 'self-controlled' drug release of the DTX-loaded PAM-PBLG-b-TPGS nanoparticles, they exhibited stronger cytotoxicity (lower survival rate) and higher uptake rate than DTX-loaded PAM-PBLG nanoparticles in Hela cells and MCF-7 cells. Furthermore, compared with DTX-loaded PAM-PBLG nanoparticles and free DTX, DTX-loaded PAM-PBLG-b-TPGS nanoparticles produced a potent anti-tumour effect. Thus, the DTX-loaded PAM-PBLG-b-TPGS nanoparticles provide a novel attractive nanocarrier for the DTX delivery of chemotherapy to human breast cancer cells and human cervical cancer cells.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Docetaxel/administración & dosificación , Portadores de Fármacos/química , Neoplasias del Cuello Uterino/tratamiento farmacológico , Vitamina E/química , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/patología , Dendrímeros/química , Docetaxel/farmacología , Docetaxel/uso terapéutico , Liberación de Fármacos , Femenino , Células HeLa , Humanos , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias del Cuello Uterino/patología
8.
J Nanobiotechnology ; 14: 10, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26846666

RESUMEN

BACKGROUND: The toxicity of CdSe/ZnS quantum dots (QDs) in the environment and biological systems has become a major concern for the nanoparticle community. However, the potential toxicity of QDs on immune cells and its corresponding immune functions remains poorly understood. In this study, we investigated the immunotoxicity of CdSe/ZnS QDs using the in vitro in macrophages and lymphocytes and in vivo in BALB/c mice. RESULTS: Our results indicated that macrophages treated with 1.25 or 2.5 nM QDs exhibited decreased cell viability, increased levels of reactive oxygen species (ROS), elevated apoptotic events, altered phagocytic ability, and decreased release of TNF-α and IL-6 by upon subsequent stimulation with Lipopolysaccharide (LPS). In contrast, lymphocytes exposed to QDs exhibited enhanced cell viability, increased release of TNF-α and IL-6 following exposure with CpG-ODN, and decreased transformation ability treatment in response to LPS. To study the in vivo effects in mice, we showed that QDs injection did not cause significant changes to body weight, hematology, organ histology, and phagocytic function of peritoneal macrophages in QDs-treated mice. In addition, the QDs formulation accumulated in major immune organs for more than 42 days. Lymphocytes from QDs-treated mice showed reduced cell viability, changed subtype proportions, increased TNF-α and IL-6 release, and reduced transformation ability in response to LPS. CONCLUSIONS: Taken together, these results suggested that exposures to CdSe/ZnS QDs could suppress immune-defense against foreign stimuli, which in turn could result in increased susceptibility of hosts to diseases.


Asunto(s)
Compuestos de Cadmio/inmunología , Compuestos de Cadmio/toxicidad , Linfocitos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Puntos Cuánticos/toxicidad , Sulfuros/inmunología , Sulfuros/toxicidad , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Interleucina-6/inmunología , Interleucina-6/metabolismo , Linfocitos/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Nanopartículas/toxicidad , Oligodesoxirribonucleótidos/inmunología , Oligodesoxirribonucleótidos/metabolismo , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
9.
Bioelectromagnetics ; 37(4): 244-55, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27061713

RESUMEN

Magnetic field exposure is an accepted safe and effective modality for nerve injury. However, it is clinically used only as a supplement or salvage therapy at the later stage of treatment. Here, we used a planarian Girardia sinensis decapitated model to investigate beneficial effects of early rotary non-uniform magnetic fields (RMFs) exposure on central nervous regeneration. Our results clearly indicated that magnetic stimulation induced from early RMFs exposure significantly promoted neural regeneration of planarians. This stimulating effect is frequency and intensity dependent. Optimum effects were obtained when decapitated planarians were cultured at 20 °C, starved for 3 days before head-cutting, and treated with 6 Hz 0.02 T RMFs. At early regeneration stage, RMFs exposure eliminated edema around the wound and facilitated subsequent formation of blastema. It also accelerated cell proliferation and recovery of neuron functionality. Early RMFs exposure up-regulated expression of neural regeneration related proteins, EGR4 and Netrin 2, and mature nerve cell marker proteins, NSE and NPY. These results suggest that RMFs therapy produced early and significant benefit in central nervous regeneration, and should be clinically used at the early stage of neural regeneration, with appropriate optimal frequency and intensity.


Asunto(s)
Sistema Nervioso Central/fisiología , Campos Magnéticos , Regeneración Nerviosa , Planarias/fisiología , Rotación , Animales , Biomarcadores/metabolismo , Regulación de la Expresión Génica , Temperatura , Factores de Tiempo
10.
J Transl Med ; 13: 166, 2015 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-25990580

RESUMEN

BACKGROUND: The immune non-recognition is often the underlying cause of failure in tumor immunotherapeutic. This is because most tumor-related antigens are poorly immunogenic, and fail to arouse an efficient immune response against cancers. Here we synthesized a novel TLR7 agonist, and developed a safe and effective immunotherapeutic vaccine by conjugating this TLR7 agonist with the pluripotency antigen OCT4. METHODS: Purified recombinant OCT4 protein was covalently linked with a novel TLR7 agonist to form a TLR7-OCT4 conjugate (T7-OCT4). After conjugation, the in vitro release of IL-12 and IFN-γ was observed in spleen lymphocytes. Mice were immunized with TLR7-OCT4, and the release of IFN-γ, the percentages of CD3+/CD8+ T cells and the OCT4-specific cytotoxicity rates were measured. The immunized mice were challenged with mouse embryonic carcinoma (EC), and the tumor volume and tumor weight were determined. Blood routine examination was performed to evaluate the biosafety of TLR7 agonist and TLR7-OCT4 conjugate in mice. RESULTS: T7-OCT4 conjugate significantly increased the in vitro release of IL-12 and IFN-γ by mouse spleen lymphocytes. In addition, the release of IFN-γ, the percentages of CD3+/CD8+ T cells and the tumor-specific cytotoxicity rates in immunized mice were significantly higher. Importantly, in EC xenografted mice, immunization with T7-OCT4 conjugate decreased the growth of the tumor dramatically up to 90 %, as compared to mice immunized with OCT4 protein or TLR7 agonist alone. Furthermore, blood routine examination demonstrated that no abnormalities of the blood cells and components in the blood fluids were detected by T7-OCT4 and TLR7 agonist injections. CONCLUSIONS: Our results showed that conjugating OCT4 protein to the novel TLR7 agonist produced a vaccine which is effective and safe in preventing tumor growth in mice. Our results suggest that this type of vaccine formulation has great potentiality in preventive vaccines against OCT4 expressing tumors.


Asunto(s)
Carcinoma Embrionario/metabolismo , Glicoproteínas de Membrana/química , Factor 3 de Transcripción de Unión a Octámeros/química , Neoplasias Testiculares/metabolismo , Receptor Toll-Like 7/química , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/citología , Carcinoma Embrionario/prevención & control , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Linfocitos/citología , Masculino , Glicoproteínas de Membrana/agonistas , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Trasplante de Neoplasias , Proteínas Recombinantes/metabolismo , Bazo/metabolismo , Linfocitos T Citotóxicos/citología , Neoplasias Testiculares/prevención & control , Factores de Tiempo , Receptor Toll-Like 7/agonistas
11.
Nanomedicine ; 11(2): 341-50, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25461291

RESUMEN

Along with widespread usage of QDs in electronic and biomedical industries, the likelihood of QDs exposure to the environment and humans is deemed to occur when the QD products are degraded or handled as waste for processing. To date, there are very few toxicological reports available in the literature for non-cadmium QDs in animal models. In this work, we studied the long term in vivo toxicity of InP/ZnS QDs in BALB/c mice. The biodistribution, body weight, hematology, blood biochemistry, and organ histology were determined at a very high dosage (25 mg/kg) of InP/ZnS QDs over 84 days period. Our results manifested that the QDs formulation did not result in observable toxicity in vivo within the evaluation period, thereby suggesting that the InP/ZnS QDs can be utilized as optical probes or nanocarrier for selected in vivo biological applications when an optimized dosage is employed. FROM THE CLINICAL EDITOR: This study investigated the toxicity of quantum dots in BALB/c mice, and concluded that no organotoxicity was detectable despite of using high concentration of InP/ZnS quantum dots with prolonged exposure of 3 months.


Asunto(s)
Indio/toxicidad , Nanopartículas/toxicidad , Fosfinas/toxicidad , Puntos Cuánticos/toxicidad , Sulfato de Zinc/toxicidad , Animales , Humanos , Ratones , Ratones Endogámicos BALB C , Distribución Tisular/efectos de los fármacos
12.
Analyst ; 139(18): 4681-90, 2014 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-25054471

RESUMEN

Biofunctionalized quantum dots (QDs), especially protein-coated QDs, are known to be useful targeted fluorescent labels for cellular and deep-tissue imaging. These nanoparticles can also serve as efficient energy donors in fluorescence resonance energy transfer (FRET) binding assays for the multiplexed sensing of tumor markers. However, current preparation processes for protein-functionalized QDs are laborious and require multiple synthesis steps (e.g. preparing them in high temperature, making them dispersible in water, and functionalizing them with surface ligands) to obtain a high quality and quantity of QD formulations, significantly impeding the progress of employing QDs for clinical diagnostics use such as a QD-based immunohistofluorescence assay. Herein, we demonstrate a one-step synthesis approach for preparing protein-functionalized QDs using a microfluidic (MF) chip setup. Using bovine serum albumin (BSA) molecules as the surface ligand model, we first studied and optimized the MF reaction synthesis parameters (e.g. reaction temperature, and channel width and length) for making protein-functionalized QDs using COMSOL simulation modeling, followed by experimental verification. Moreover, in comparison with the BSA-functionalized QDs synthesized using the conventional bench-top method, BSA-QDs prepared using the MF approach exhibit a significantly higher protein-functionalization efficiency, photostability and colloidal stability. The proposed one-step MF synthesis approach provides a rapid, cost effective, and a small-scale production of nanocrystals platform for developing new QD formulations in applications ranging from cell labeling to biomolecular sensing. Most importantly, this approach will considerably reduce the amount of chemical waste generated during the trial-and-error stage of developing and perfecting the desired physical and optical properties of new QD materials.


Asunto(s)
Técnicas Analíticas Microfluídicas/instrumentación , Nanotecnología/instrumentación , Puntos Cuánticos/química , Albúmina Sérica Bovina/química , Animales , Bovinos , Línea Celular , Diseño de Equipo , Ratones , Imagen Óptica
13.
Small Methods ; : e2301430, 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38191992

RESUMEN

Given the inevitable human exposure owing to its increasing production and utilization, the comprehensive safety evaluation of silica nanoparticles (SiNPs) has sparked concerns. Substantial evidence indicated liver damage by inhaled SiNPs. Notwithstanding, few reports focused on the persistence or reversibility of hepatic injuries, and the intricate molecular mechanisms involved remain limited. Here, rats are intratracheally instilled with SiNPs in two regimens (a 3-month exposure and a subsequent 6-week recovery after terminating SiNPs administration) to assess the hepatic effects. Nontargeted lipidomics revealed alterations in lipid metabolites as a contributor to the hepatic response and recovery effects of SiNPs. In line with the functional analysis of differential lipid metabolites, SiNPs activated oxidative stress, and induced lipid peroxidation and lipid deposition in the liver, as evidenced by the elevated hepatic levels of ROS, MDA, TC, and TG. Of note, these indicators showed great improvements after a 6-week recovery, even returning to the control levels. According to the correlation, ROC curve, and SEM analysis, 11 lipids identified as potential regulatory molecules for ameliorating liver injury by SiNPs. Collectively, the work first revealed the reversibility of SiNP-elicited hepatotoxicity from the perspective of lipidomics and offered valuable laboratory evidence and therapeutic strategy to facilitate nanosafety.

14.
Toxicol Lett ; 391: 100-110, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38040069

RESUMEN

The widespread existence of 2,2',4,4'-tetra-bromodiphenyl ether (BDE-47) in the environment has aroused great concern. BDE-47 induces the occurrence of metabolic dysfunction-associated steatotic liver disease (MASLD), but the mechanism has not been fully elucidated. Here, we further investigate the underlying mechanism using BALB/c mice. After BDE-47 exposure, the livers of mice enlarged, the serum levels of ALT, ALP, TG and TC enhanced, and hepatic steatosis occurred. Transcriptome sequencing identifies 2250 differentially expressed genes (DEGs). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis reveals that down-regulated DEGs are mainly enriched in pathways associated with lipid metabolism, particularly in fatty acid (FA) degradation. And up-regulated DEGs are mainly enriched in pathways related to lipid and FA transport. The expression levels of AhR, Pparγ and Cd36 involved in FA uptake are up-regulated, and those of PPARα and target genes including Cpt1 and Cyp4a1 related to ß and ω-oxidation are inhibited. These results reveal BDE-47 could lead to metabolic dysfunction-associated steatotic liver disease (MASLD) by promoting FA uptake via upregulating Cd36 and hindering oxidative utilization by downregulating PPARα.


Asunto(s)
Hígado Graso , Éteres Difenilos Halogenados , Enfermedades Metabólicas , Enfermedad del Hígado Graso no Alcohólico , Ratones , Animales , Ácidos Grasos/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Ratones Endogámicos BALB C , Hígado Graso/inducido químicamente , Hígado Graso/metabolismo , Hígado/metabolismo , Metabolismo de los Lípidos , Antígenos CD36/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo
15.
Small ; 9(16): 2757-63, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23427041

RESUMEN

The application of small interfering RNA (siRNA)-based RNA interference (RNAi) for cancer gene therapy has attracted great attention. Gene therapy is a promising strategy for cancer treatment because it is relatively non-invasive and has a higher therapeutic specificity than chemotherapy. However, without the use of safe and efficient carriers, siRNAs cannot effectively penetrate the cell membranes and RNAi is impeded. In this work, cationic poly(lactic acid) (CPLA)-based degradable nanocapsules (NCs) are utilized as novel carriers of siRNA for effective gene silencing of pancreatic cancer cells. These CPLA-NCs can readily form nanoplexes with K-Ras siRNA and over 90% transfection efficiency is achieved using the nanoplexes. Cell viability studies show that the nanoparticles are highly biocompatible and non-toxic, indicating that CPLA-NC is a promising potential candidate for gene therapy in a clinical setting.


Asunto(s)
Genes ras/genética , Nanocápsulas/química , Neoplasias Pancreáticas/terapia , Línea Celular Tumoral , Silenciador del Gen/fisiología , Humanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Neoplasias Pancreáticas
16.
Nutrients ; 15(4)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36839265

RESUMEN

Naringin (Nar) is a dihydroflavonoid compound, widely found in citrus fruit and used in Chinese herbal medicine. As a phytochemical, it acts as a dietary supplement that can delay aging and prevent aging-related disease, such as obesity and diabetes. However, its exact mechanism remains unclear. In this study, the high-glucose-induced (HGI) Caenorhabditis elegans model was used to evaluate the anti-aging and anti-obesity effects of Nar. The mean lifespan and fast movement span of HGI worms were extended roughly 24% and 11%, respectively, by Nar treatment. Oil red O staining revealed a significant reduction in fat accumulation and dFP::LGG-labeled worms showed the promotion of autophagy. Additionally, whole transcriptome sequencing and gene set variation analysis suggested that Nar upregulated the lipid biosynthesis and metabolism pathways, as well as the TGF-ß, Wnt and longevity signaling pathways. Protein-protein interaction (PPI) network analysis identified hub genes in these pathways for further analysis. Mutant worms and RNA interference were used to study mechanisms; the suppression of hlh-30, lgg-1, unc-51, pha-4, skn-1 and yap-1 disabled the fat-lowering, lifespan-prolonging, and health-promoting properties of Nar. Collectively, our findings indicate that Nar plays an important role in alleviating HGI-aging and anti-obesity effects by reducing fat accumulation and promoting autophagy.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Animales , Proteínas de Caenorhabditis elegans/metabolismo , Glucosa/metabolismo , Envejecimiento/genética , Longevidad , Autofagia/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas Señalizadoras YAP
17.
Lab Invest ; 92(10): 1503-14, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22890557

RESUMEN

Polo-like kinase 1 (Plk1) is a mitotic serine/threonine kinase and its kinase activity is closely interrelated to cell cycle progression, various types of cancer development and often correlates with poor prognosis. Thus, it is of prime importance to characterize the phenotypes of Plk1 inhibition in cells for drug development and clinical application. Here, we report a novel kinase inhibitory chemical, CBB2001, which specifically inhibited Plk1 kinase activity in vitro with an IC(50) of 0.39 µM. In cervical carcinoma HeLa cells, we found that treatment of CBB2001 caused mitotic cell cycle arrest (EC(50)=0.72 µM) and induction of 'polo' cells (EC(50)=0.32 µM). Interestingly, the cell cycle arrest induced by CBB2001 was associated with accumulation of Plk1 (EC(50)=0.61 µM) and Geminin (EC(50)=0.43 µM) proteins, but distinct from the phenotypes induced by Aurora kinase inhibitors. The inhibitory effects of CBB2001 were phenocopied by RNA interferences of Plk1. We also confirmed the cell cycle inhibitory effects of CBB2001 in other cancer cells. Moreover, CBB2001 inhibited the growth of HeLa cells with an IC(50) of 0.85 µM in MTT assays, which is better than that of reported Plk1 inhibitory chemicals ON01910 (IC(50)=6.46 µM) and LFM-A13 (IC(50)=37.36 µM). CBB2001 also inhibited mouse xenograft tumor growth. Furthermore, CBB2001 inhibited mitotic exit and delayed degradation of APC/C substrates, Geminin, Cyclin B1 and Aurora A. These specific phenotypes may serve as specific features for Plk1 inhibition and for Plk1-based clinic trials.


Asunto(s)
Bencimidazoles/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , División Celular/efectos de los fármacos , Neoplasias/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Tiazoles/farmacología , Amidas/farmacología , Animales , Aurora Quinasa A , Aurora Quinasas , Bencimidazoles/química , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina B1/metabolismo , Femenino , Geminina , Glicina/análogos & derivados , Glicina/farmacología , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias/patología , Nitrilos/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Sulfonas/farmacología , Tiazoles/química , Quinasa Tipo Polo 1
18.
J Hazard Mater ; 422: 126850, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34419847

RESUMEN

As the most abundant congener of polybrominated diphenyl ethers (PBDEs) detected in environment and human biotic samples, 2, 2', 4, 4'-tetrabromodiphenyl ether (BDE-47) has been found to accumulate in brain and induce neurotoxicity, however, the detailed mechanism has not been clearly elucidated. To investigate the neurotoxicity of BDE-47, undifferentiated PC-12 cells were exposed to different doses of BDE-47, and BDE-47 dissolved in corn oil was orally administered to mice for 8 consecutive weeks. Our data showed that BDE-47 obviously changed cell morphology, altered cell viability, promoted cell apoptosis, and induced reactive oxygen species (ROS) production. BDE-47 promoted the differentiation of PC-12 cells by enhancing the expression of TrkA receptor and the phosphorylation levels of ERK and Akt. Moreover, BDE-47-induced differentiation of PC-12 cells was suppressed by inhibitors of corresponding pathways (MAPK/ERK and PI3K/Akt). H&E staining of brain showed neurons in DG and CA1 areas of hippocampus decreased after BDE-47 exposure. Transcriptome sequencing of brain tissue suggested that multiple signaling pathways related to neuron death and nerve function were significantly regulated. In conclusion, these results provided new evidence for revealing the neurotoxicity of BDE-47, and offered important experimental basis for environmental controlling and post-exposure health risk assessment of BDE-47.


Asunto(s)
Éteres Difenilos Halogenados , Fosfatidilinositol 3-Quinasas , Animales , Diferenciación Celular , Éteres Difenilos Halogenados/toxicidad , Hipocampo , Ratones , Ratones Endogámicos BALB C , Neuronas
19.
Neurotoxicology ; 88: 134-143, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34785253

RESUMEN

As a newly developed cadmium-free quantum dot (QD), CuInS2/ZnS has great application potential in many fields, but its biological safety has not been fully understood. In this study, the in vitro toxicity of CuInS2/ZnS QDs on U87 human glioma cell line was explored. The cells were treated with different concentrations of QDs (12.5, 25, 50 and 100 µg/mL), and the uptake of QDs by the U87 cells was detected by fluorescence imaging and flow cytometry. The cell viability was observed by MTT assay, and the gene expression profile was analyzed by transcriptome sequencing. These results showed that QDs could enter the cells and mainly located in the cytoplasm. The uptake rate was over 90 % when the concentration of QDs reached 25 µg/mL. The cell viability (50 and 100 µg/mL) increased at 24 h (P < 0.05), but no significant difference after 48 h and 72 h treatment. The results of differential transcription showed that coding RNA accounted for the largest proportion (62.15 %), followed by long non-coding RNA (18.65 %). Total 220 genes were up-regulated and 1515 genes were down-regulated, and significantly altered gene functions included nucleosome, chromosome-DNA binding, and chromosome assembly. In conclusion, CuInS2/ZnS QDs could enter U87 cells, did not reduce the cell viability, but would obviously alter the gene expression profile. These findings provide valuable information for a proper understanding of the toxicity risk of CuInS2/ZnS QD and promote the rational utilization of QDs in the future.


Asunto(s)
Neuroglía/efectos de los fármacos , Puntos Cuánticos/toxicidad , Transcriptoma/efectos de los fármacos , Línea Celular , Cobre , Relación Dosis-Respuesta a Droga , Humanos , Indio , Microscopía Fluorescente , Neuroglía/metabolismo , Puntos Cuánticos/metabolismo , Sulfuros , Compuestos de Zinc
20.
Chemosphere ; 308(Pt 3): 136562, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36152834

RESUMEN

The health risks associated with 2,2',4,4'-tetra-bromodiphenyl ether (BDE-47) have become an increasing concern due to its widespread presence in the environment and biological samples. To date, the potential toxicity of BDE-47 to immune system remains unclear. In this study, we aimed to study the immunotoxicity of BDE-47 using spleen-derived lymphocytes in vitro and BALB/c mice in vivo. In vitro results showed that lymphocytes exposed to 12.5-100 µM BDE-47 exhibited unchanged cell viability but decreased release of IL-6 and TNF-α when responding to lipopolysaccharide (LPS). The expression levels of p-p65, p-IκBα, TrkA and p-Akt involved in NF-κB pathway were obviously decreased, and NF-κB activator PMA could recover the BDE-47-induced inhibitory effect on IL-6 and TNF-α release by lymphocytes in response to LPS. In vivo data showed that BDE-47 orally administered to mice (1 mg/kg, 10 mg/kg, 100 mg/kg per day, 30 days) did not significantly affect body weight, organ index and histomorphology of spleen. However, ELISA assay showed that serum IL-6 and TNF-α levels from BDE-47-treated mice after intraperitoneal injection of LPS were significantly reduced, and high-throughput mouse cytokines screening found 13 more cytokines down-regulated in the serum. Transcriptomic sequencing of spleens identified 488 differential expressed genes (DEGs). GO enrichment analysis of these DEGs suggested that the GO term of response to LPS (GO: 0032,496) was significantly involved. KEGG enrichment analysis showed that the down-regulated DEGs significantly enriched in multiple immune-related signaling pathways including the NF-κB signaling pathway (mmu04064). Overall, these data suggested that BDE-47 could negatively regulate NF-κB signaling pathways to inhibit the immune response of lymphocytes to LPS, suggesting that exposures to BDE-47 may disturb the immune balance and increase the body's susceptibility to infectious diseases.


Asunto(s)
Lipopolisacáridos , FN-kappa B , Animales , Citocinas/metabolismo , Éteres Difenilos Halogenados/toxicidad , Inmunidad , Interleucina-6 , Lipopolisacáridos/toxicidad , Linfocitos/metabolismo , Ratones , Ratones Endogámicos BALB C , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt , Factor de Necrosis Tumoral alfa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA