Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Nanobiotechnology ; 21(1): 9, 2023 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-36609374

RESUMEN

As one of the most significant imaging modalities currently available, magnetic resonance imaging (MRI) has been extensively utilized for clinically accurate cancer diagnosis. However, low signal-to-noise ratio (SNR) and low specificity for tumors continue to pose significant challenges. Inspired by the distance-dependent magnetic resonance tuning (MRET) phenomenon, the tumor microenvironment (TME)-activated off-on T1-T2 dual-mode MRI nanoswitch is presented in the current study to realize the sensitive early diagnosis of tumors. The tumor-specific nanoswitch is designed and manufactured on the basis of PDGFB-conjugating ferroferric oxide coated by Mn-doped silica (PDGFB-FMS), which can be degraded under the high-concentration GSH and low pH in TME to activate the T1-T2 dual-mode MRI signals. The tumor-specific off-on dual-mode MRI nanoswitch can significantly improve the SNR and is used successfully for the accurate diagnosis of early-stage tumors, particularly for orthotopic prostate cancer. In addition, the systemic delivery of the nanoswitch did not cause blood or tissue damage, and it can be excreted out of the body in a timely manner, demonstrating excellent biosafety. Overall, the strategy is a significant step in the direction of designing off-on dual-mode MRI nanoprobes to improve imaging accuracy, which opens up new avenues for the development of new MRI probes.


Asunto(s)
Medios de Contraste , Neoplasias , Masculino , Humanos , Proteínas Proto-Oncogénicas c-sis , Neoplasias/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Dióxido de Silicio , Microambiente Tumoral
2.
J Nanobiotechnology ; 21(1): 59, 2023 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-36810074

RESUMEN

Chemodynamic therapy of cancer is limited by insufficient endogenous H2O2 generation and acidity in the tumor microenvironment (TME). Herein, we developed a biodegradable theranostic platform (pLMOFePt-TGO) involving composite of dendritic organosilica and FePt alloy, loaded with tamoxifen (TAM) and glucose oxidase (GOx), and encapsulated by platelet-derived growth factor-B (PDGFB)-labeled liposomes, that effectively uses the synergy among chemotherapy, enhanced chemodynamic therapy (CDT), and anti-angiogenesis. The increased concentration of glutathione (GSH) present in the cancer cells induces the disintegration of pLMOFePt-TGO, releasing FePt, GOx, and TAM. The synergistic action of GOx and TAM significantly enhanced the acidity and H2O2 level in the TME by aerobiotic glucose consumption and hypoxic glycolysis pathways, respectively. The combined effect of GSH depletion, acidity enhancement, and H2O2 supplementation dramatically promotes the Fenton-catalytic behavior of FePt alloys, which, in combination with tumor starvation caused by GOx and TAM-mediated chemotherapy, significantly increases the anticancer efficacy of this treatment. In addition, T2-shortening caused by FePt alloys released in TME significantly enhances contrast in the MRI signal of tumor, enabling a more accurate diagnosis. Results of in vitro and in vivo experiments suggest that pLMOFePt-TGO can effectively suppress tumor growth and angiogenesis, thus providing an exciting potential strategy for developing satisfactory tumor theranostics.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Línea Celular Tumoral , Microambiente Tumoral , Peróxido de Hidrógeno/metabolismo , Neoplasias/tratamiento farmacológico , Apoptosis , Glucosa Oxidasa/metabolismo
3.
J Nanobiotechnology ; 20(1): 264, 2022 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672821

RESUMEN

The application of chemodynamic therapy (CDT) for cancer is a serious challenge owing to the low efficiency of the Fenton catalyst and insufficient H2O2 expression in cells. Herein, we fabricated a PDGFB targeting, biodegradable FePt alloy assembly for magnetic resonance imaging (MRI)-guided chemotherapy and starving-enhanced chemodynamic therapy for cancer using PDGFB targeting, pH-sensitive liposome-coated FePt alloys, and GOx (pLFePt-GOx). We found that the Fenton-catalytic activity of FePt alloys was far stronger than that of traditional ultrasmall iron oxide nanoparticle (UION). Upon entry into cancer cells, pLFePt-GOx nanoliposomes degraded into many tiny FePt alloys and released GOx owing to the weakly acidic nature of the tumor microenvironment (TME). The released GOx-mediated glucose consumption not only caused a starvation status but also increased the level of cellular H2O2 and acidity, promoting Fenton reaction by FePt alloys and resulting in an increase in reactive oxygen species (ROS) accumulation in cells, which ultimately realized starving-enhanced chemodynamic process for killing tumor cells. The anticancer mechanism of pLFePt-GOx involved ROS-mediated apoptosis and ferroptosis, and glucose depletion-mediated starvation death. In the in vivo assay, the systemic delivery of pLFePt-GOx showed excellent antitumor activity with low biological toxicity and significantly enhanced T2-weighted magnetic resonance imaging (MRI) signal of the tumor, indicating that pLFePt-GOx can serve as a highly efficient theranostic tool for cancer. This work thus describes an effective, novel multi-modal cancer theranostic system.


Asunto(s)
Nanopartículas , Neoplasias , Aleaciones , Línea Celular Tumoral , Glucosa , Humanos , Peróxido de Hidrógeno/metabolismo , Imagen por Resonancia Magnética , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas Proto-Oncogénicas c-sis/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral
4.
Acta Biomater ; 177: 400-413, 2024 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-38336268

RESUMEN

Herein, we developed a doxorubicin (Dox)-loaded and 4T1 cancer cell membrane-modified hydrogenated manganese oxide nanoparticles (mHMnO-Dox) to elicit systemic antitumor immune responses. The results revealed that mHMnO-Dox actively recognized tumor cells and then effectively delivered Dox into the cells. Upon entering tumor cells, the mHMnO-Dox underwent rapid degradation and abundant release of Mn2+ and chemotherapeutic drugs. The released Mn2+ not only catalysed a Fenton-type reaction to produce excessive reactive oxygen species (ROS) but also activated the cGAS-STING pathway to boost dendritic cell (DC) maturation. This process increased cytotoxic T lymphocyte infiltration as well as natural killer cell recruitment into the tumor site. In addition, the released Dox could contribute to a chemotherapeutic effect, while activating DC cells and subsequently intensifying immune responses through immunogenic cell death (ICD) of tumor cells. Consequently, the mHMnO-Dox suppressed the primary and distal tumor growth and inhibited tumor relapse and metastasis, as well as prolonged the lifespan of tumor-bearing mice. Thus, the mHMnO-Dox multimodally activated DC cells to demonstrate synergistic antitumor activity, which was mediated via the activation of the cGAS-STING signalling pathway to regulate tumor microenvironment, ICD-mediated immunotherapy and ROS-mediated CDT. These findings suggest the therapeutic potential of mHMnO-Dox in cancer immunotherapy. STATEMENT OF SIGNIFICANCE: A cancer cell membrane-camouflaged hydrogenated mesoporous manganese oxide (mHMnO) has been developed as a cGAS-STING agonist and ICD inducer. The mHMnO effectively induced abundance of ROS production in cancer cells, which caused cancer cell death and then promoted DC maturation via tumour-associated antigen presentation. Meanwhile, the mHMnO significantly activated cGAS-STING pathway to facilitate DC maturation and cytotoxic T lymphocyte infiltration as well as natural killer cell recruitment, which further enhanced tumour immune response. In addition, the combination of the mHMnO and Dox could synergistically promote tumour ICD and then multimodally induce DC maturation, achieving an enhanced CIT. Overall, this study provides a potential strategy to design novel immunologic adjuvant for enhanced CIT.


Asunto(s)
Inmunoterapia , Compuestos de Manganeso , Neoplasias , Óxidos , Animales , Ratones , Especies Reactivas de Oxígeno , Doxorrubicina , Neoplasias/tratamiento farmacológico , Células Dendríticas , Microambiente Tumoral
5.
Carbohydr Polym ; 322: 121309, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37839850

RESUMEN

Water pollution caused by dyes has become a focal point of attention. Among them, the heterogeneous Fenton reaction has emerged as an effective solution to this problem. In this study, we designed a ferric alginate gel (PAGM) tri-modified with poly(vinyl alcohol), graphene oxide, and MoS2 as a heterogeneous Fenton catalyst for organic dye degradation. PAGM addresses the drawbacks of alginate gel, such as poor mechanical properties and gel chain dissolution, thereby significantly extending the catalyst's lifespan. The removal rate of rhodamine B by PAGM reached 95.5 % within 15 min, which was 5.9 times higher than that of unmodified ferric alginate gel. Furthermore, due to the π-π interactions, PAGM exhibits unique adsorption properties for pollutants containing benzene rings. Additionally, PAGM can be regenerated multiple times through a simple soaking procedure without any performance degradation. Finally, the reaction column constructed with PAGM maintained an 83.5 % removal rate even after 319 h of continuous wastewater treatment. This work introduces a novel concept for the study of alginate-based gel catalysts in heterogeneous Fenton reactions.

6.
ACS Biomater Sci Eng ; 9(2): 809-820, 2023 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-36622161

RESUMEN

Recently, starvation-inducing nutrient deprivation has been regarded as a promising strategy for tumor suppression. As a first-line lipid-lowering drug, atorvastatin (ATV) significantly reduces caloric intake, suggesting its potential in starvation therapy for suppressing tumors. Accordingly, we developed a novel starvation therapy agent (HA-Se-ATV) in this study to suppress tumor growth by using hyaluronic acid (HA)-conjugated chitosan polymer-coated nano-selenium (Se) for loading ATV. HA-Se-ATV targets cancer cells, following which it effectively accumulates in the tumor tissue. The HA-Se-ATV nanoplatform was then activated by inducing a weakly acidic tumor microenvironment and subsequently releasing ATV. ATV and Se synergistically downregulate the levels of cellular adenosine triphosphate while inhibiting the expression of thioredoxin reductase 1. Consequently, the starvation-stress reaction of cancer cells is significantly elevated, leading to cancer cell death. Furthermore, the in vivo results indicate that HA-Se-ATV effectively suppresses tumor growth with a low level of toxicity, demonstrating its great potential for clinical translation.


Asunto(s)
Neoplasias , Selenio , Humanos , Atorvastatina/farmacología , Atorvastatina/uso terapéutico , Selenio/farmacología , Neoplasias/tratamiento farmacológico , Concentración de Iones de Hidrógeno , Microambiente Tumoral
7.
Nanoscale ; 14(32): 11600-11611, 2022 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-35861683

RESUMEN

A self-preservation Pt(IV) nanoplatform, amorphous ferric oxide-coating selenium core-shell nanoparticles (iAIO@NSe-Pt), was developed for H2O2 depletion-mediated tumor anti-angiogenesis, apoptosis, and ferroptosis. Upon entry into the blood, the ferric oxide shell effectively blocked the contact Pt(IV) prodrug with reduced molecules, then avoided the inactivation of the Pt(IV) prodrug and increased its accumulation in the tumor. After entering cancer cells, iAIO@NSe-Pt caused a series of cascade reactions: (1) AIO on the surface of iAIO@NSe-Pt quickly dissolved, released an abundance of Fe(II) because of the weakly acidic tumor microenvironment, and then catalyzed cellular H2O2 into highly toxic ˙OH, resulting in cellular H2O2 deficiency and cell ferroptosis. (2) The platinum(IV) prodrugs were exposed and quickly reduced to highly toxic Pt(II) by depleting GSH. This process inactivated GPX4, promoted ROS accumulation, and further accelerated ferroptosis. In addition, the generated Pt(II) quickly inhibited DNA replication, achieving effective apoptotic cell death. Meanwhile, Pt(II) inactivated SOD1, which blocked the synthesis of cellular H2O2 and accelerated ROS (superoxide anion radical) accumulation. (3) The deficiency of cellular H2O2 significantly inhibited the expression of vascular endothelial growth factor-A (VEGF-A), blocking tumor angiogenesis and then improving the anticancer effect. (4) After such a cascade reaction, the exposed NSe successively disrupted mitochondrial respiration and inhibited cancer angiogenesis, further inducing cancer cell death. Collectively, our functional and mechanical investigation suggested that iAIO@NSe-Pt exhibits excellent tumor targeting, biocompatibility and anti-tumor efficiency in vitro and in vivo, and provides a novel example of a self-preservation Pt(IV) nanoplatform for H2O2 depletion-mediated tumor anti-angiogenesis, apoptosis, and ferroptosis, showing great promise for future clinical use.


Asunto(s)
Ferroptosis , Nanopartículas , Neoplasias , Profármacos , Selenio , Apoptosis , Línea Celular Tumoral , Compuestos Férricos , Humanos , Peróxido de Hidrógeno/uso terapéutico , Neoplasias/tratamiento farmacológico , Profármacos/farmacología , Profármacos/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular
8.
ACS Appl Mater Interfaces ; 14(25): 28514-28526, 2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-35698257

RESUMEN

Nanoparticles are widely used in biological research and cancer therapy. In hepatocellular carcinoma, several nanoplatforms have been synthesized and studied to improve the drug efficacy; however, these nanoplatforms are still insufficient to eradicate tumors. Herein, we have synthesized a novel vanadium (V)-iron-oxide (ION) nanoparticle (VIO) that combines chemodynamic, photothermal, and diagnostic capacities to enhance the tumor suppression effect in one agent with multiple functions. In the in vitro models, hepatocellular carcinoma cells are significantly inhibited by VIO-based nanoagents. The mechanistic study validates that VIO increases reactive oxygen species (ROS), which led to apoptosis and ferroptosis resulting in cell death. To our surprise, VIO targets not only tumor cells but also endothelial cells. In addition to inducing cell death, VIO also blocks tube formation and cell migration in human umbilical vein endothelial cell (HUVEC) and C166 models, indicating an antiangiogenic potential. In mouse tumor models, VIO retards tumor growth and induces apoptosis in tumor tissues. Furthermore, a significant blood vessel regression is seen in VIO-treated groups accompanied with larger necrotic areas. More interestingly, the activation of photothermal therapy completely eradicates tumor tissues. Taken together, this VIO nanoplatform could be a powerful anticancer candidate for nanodrug development.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hierro/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Ratones , Óxidos/farmacología , Vanadio
9.
Experimental Neurobiology ; : 97-111, 2009.
Artículo en Inglés | WPRIM | ID: wpr-202567

RESUMEN

The aim of this study is to verify the feasibility of control of one-dimensional (1-D) rotating machine using neural activities of Prefrontal cortex (PFC) in a BCI system. In this study, adult male Sprague-Dawley rats received bilateral implantation of recording micro-electrodes in PFC area. The spontaneous activities of a pair of PFC neurons of water-deprived rats were encoded and converted through a triple-step threshold comparator algorithm to three commands for one-dimensional movement control of a robotic wheel for accessing water. Averaged activities of two PFC neurons were quantized in every 200 ms to four ranges of activities around the mean firing rates (+/-0.5 SD) and were converted to four values. After comparison of the values of two chosen neuron units, direction and speed of rotation were decided. Rats were trained to complete one-dimensional control task to obtain water reward. The results indicated the percentage of stop event increased alone with more training. Different brain activity significantly influenced total water-drinking duration and non-water-drinking duration. Events generated from neuronal activity differed according to variant experimental sessions. Correlation between two signal units impacted controlling performance. Overall, the results of this study suggest that rats were able to manipulate the 1-D BCI system by differentially modulating PFC single neuron activities according to different circumstances.


Asunto(s)
Adulto , Animales , Humanos , Masculino , Ratas , Encéfalo , Interfaces Cerebro-Computador , Incendios , Neuronas , Corteza Prefrontal , Ratas Sprague-Dawley , Recompensa , Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA