Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Colloid Interface Sci ; 628(Pt B): 419-434, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-35998465

RESUMO

As an emerging technology, nanocatalytic medicine attracts much attention, especially the ones according to the enzymatic reaction by using excess H2O2 in the tumor. Among various candidates, single-atom catalyst (SAC) revealed unique and outstanding redox reaction performance, since the active sites consisting of single metal atoms may achieve the maximum utilization of metal atoms and emerge obviously amplified reaction rate. Here we developed an M-Nx (M = Mn, Zn) center-based SAC with a hollow structure by calcination of Mn2+-doped zeolitic imidazolate frameworks (ZIF-8), and PEGylation was applied to improve the hydrophilicity. According to the enzymatic reaction, the M-Nx (M = Mn, Zn) centers have an inherent peroxidase-like activity to catalyze over-expressed H2O2 in the weak acidic tumor microenvironment and generate a large amount of toxic reactive oxygen species (ROS) like hydroxyl radicals for therapy. To keep efficient therapeutic output, we integrated the hollow SAC with Au which could expend the glucose in tumor and supply H2O2 as the substrate of peroxidase-like activity. Better yet, Au may boost the photothermal effect of SAC and offer another non-invasive photothermal therapy (PTT) to promote the effect of tumor removal. This platform provided a new idea for the construction of more efficient peroxidase-like activity in tumor therapy.


Assuntos
Neoplasias , Terapia Fototérmica , Humanos , Espécies Reativas de Oxigênio , Peróxido de Hidrogênio/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Glucose , Peroxidases , Linhagem Celular Tumoral , Microambiente Tumoral
2.
J Colloid Interface Sci ; 623: 787-798, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35636288

RESUMO

Although nanocatalytic therapy has become an emerging strategy for tumor treatment, the therapeutic effects of reactive oxygen species (ROS)-mediated treatment are still seriously limited by the inherent flaws of the enzymatic activities and the specific physicochemical properties of the tumor microenvironment (TME). Herein, we report an ultrasmall bimetallic oxide nanozyme (CuFe2O4@PEG, CFOs) for programmable multienzyme-like activities-primed combined therapy. Under the acidic condition, abundant highly toxic ROS can be generated through the peroxidase activity of CFOs with overexpressed hydrogen peroxide (H2O2) in the tumor. High metal ion utilization of bimetallic oxide nanozymes is related to the size effect and topological structure. Furthermore, glutathione peroxidase activity-initiated depletion of GSH disrupts the intracellular antioxidant defense system and further amplifies the oxidative stress in turn. Subsequently, oxygen generation originating from the catalase activity of CFOs relieves tumor hypoxia and achieves exceptional TME-customized therapeutic effects. Notably, the high photothermal effect (η = 41.12%) of CFOs in the second near-infrared biological windows leads to the combinational inhibition of tumor growth. In summary, this report provides a paradigm for the rational design of TME-responsive and ROS-mediated nanocatalytic treatment, which is promising for achieving superior therapeutic efficiency.


Assuntos
Peróxido de Hidrogênio , Neoplasias , Antioxidantes , Linhagem Celular Tumoral , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Estresse Oxidativo , Óxidos/farmacologia , Espécies Reativas de Oxigênio , Microambiente Tumoral
3.
ACS Nano ; 16(3): 3647-3663, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35266697

RESUMO

The nanocatalytic activity of nanozymes provides a vision for tumor treatment. However, the glutathione (GSH)-related antioxidant defense system (ADS) formed on the basis of excessive GSH in the tumor microenvironment limits its catalytic activity. Here, dendritic mesoporous silica nanoparticles (DMSNs) were employed as nanocarrier; ultrasmall Fe3O4 nanoparticles, Mn2+ ions, and glutaminase inhibitor Telaglenastat (CB-839) were subsequently integrated into large mesopores of DMSNs, forming DMSN/Fe3O4-Mn@CB-839 (DFMC) nanomedicine. This nanomedicine exhibits peroxidase mimicking activities under acidic conditions, which catalyzes the decomposition of hydrogen peroxide (H2O2) into hydroxyl radical (•OH). This also promotes the formation of lipid peroxides, which is required for ferroptosis. Furthermore, this nanomedicine can effectively deplete the existing GSH, thereby enhancing reactive oxygen species (ROS)-mediated tumor catalytic therapy. Moreover, the introduced CB-839 blocks the endogenous synthesis of GSH, further enhancing GSH depletion performance, which reduces the excretion of oxaliplatin (GSH-related resistance) from tumor cells, thereby restoring the chemical sensitivity of oxaliplatin. The dual GSH depletion property significantly weakens the GSH-related ADS and restores the chemical sensitivity of oxaliplatin, leading to the high DFMC-induced apoptosis and ferroptosis of tumor cells. Our developed nanomedicine based on integrated nanotechnology and clinical drug may aid the development of tumor treatment.


Assuntos
Nanomedicina , Peroxidase , Apoptose , Linhagem Celular Tumoral , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio , Oxaliplatina/farmacologia , Peroxidases , Dióxido de Silício/química
4.
ACS Appl Mater Interfaces ; 12(51): 56886-56897, 2020 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-33290033

RESUMO

Chemodynamic therapy (CDT) based on the Fenton reaction is a promising strategy for nonlight cancer treatment. However, the traditional Fenton reaction is only efficient in strongly acidic conditions (pH = 2-4), resulting in the limited curative effect in a weakly acidic tumor microenvironment (TME). Herein, we first developed a simple in situ growth method to confine FeOCl nanosheets into hollow dendritic mesoporous organosilicon (H-DMOS) nanoparticles to obtain FeOCl@H-DMOS nanospheres. Ascorbic acid (AA) was then absorbed on the nanosystem as a H2O2 prodrug and, meanwhile, was used for the regeneration of Fentons reagent for Fe2+. Finally, poly(ethylene glycol) (PEG) was coated on FeOCl@H-DMOS-AA to enhance the permeability and retention (EPR) effect in tumor tissue. The as-fabricated FeOCl@H-DMOS-AA/PEG can generate a large amount of highly toxic hydroxyl radicals (•OH) by catalyzing H2O2 even in neutral pH conditions with the help of AA. As a result, the effect of CDT has been markedly enhanced by the increased amount of H2O2 and the efficient Fenton reaction in mild acidic TME, which can remove almost all of the tumors in mice. In addition, FeOCl also endows the nanosystem with T2-weighted MR imaging capability (r2 = 34.08 mM-1 s-1), thus realizing the imaging-guided cancer therapy. All in all, our study may contribute a new direction and may have a bright future for enhanced CDT with a neutral pH range.


Assuntos
Antineoplásicos/uso terapêutico , Meios de Contraste/uso terapêutico , Compostos de Ferro/uso terapêutico , Nanopartículas/química , Neoplasias/tratamento farmacológico , Compostos de Organossilício/química , Animais , Antineoplásicos/química , Antineoplásicos/toxicidade , Ácido Ascórbico/química , Ácido Ascórbico/uso terapêutico , Ácido Ascórbico/toxicidade , Meios de Contraste/química , Meios de Contraste/toxicidade , Feminino , Células HeLa , Humanos , Peróxido de Hidrogênio/metabolismo , Radical Hidroxila/metabolismo , Compostos de Ferro/química , Compostos de Ferro/toxicidade , Imageamento por Ressonância Magnética , Camundongos , Nanopartículas/toxicidade , Neoplasias/diagnóstico por imagem , Compostos de Organossilício/toxicidade , Polietilenoglicóis/química , Polietilenoglicóis/toxicidade , Porosidade , Pró-Fármacos/química , Pró-Fármacos/uso terapêutico , Pró-Fármacos/toxicidade , Nanomedicina Teranóstica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...