Cargando…

Platinum–copper alloy nanoparticles armored with chloride ion transporter to promote electro-driven tumor inhibition

The induction of oxidative species, driven by oscillating electric field (E), has recently emerged as an effective approach for tumor inhibition, so-called electrodynamic therapy (EDT). While it offers a series of advantages attracting considerable attention, the fundamental mechanism and improvemen...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Tong, Han, Gaorong, Li, Xiang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: KeAi Publishing 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8897047/
https://www.ncbi.nlm.nih.gov/pubmed/35310378
http://dx.doi.org/10.1016/j.bioactmat.2021.10.012
_version_ 1784663307181883392
author Chen, Tong
Han, Gaorong
Li, Xiang
author_facet Chen, Tong
Han, Gaorong
Li, Xiang
author_sort Chen, Tong
collection PubMed
description The induction of oxidative species, driven by oscillating electric field (E), has recently emerged as an effective approach for tumor inhibition, so-called electrodynamic therapy (EDT). While it offers a series of advantages attracting considerable attention, the fundamental mechanism and improvement strategies for EDT approach are being endeavored extensively with the aid of new material explorations. An interesting phenomenon observed in early studies is that the on-site concentration of chloride ion is highly favored for the induction of oxidative species and the efficacy of tumor inhibition. Following this discovery ignored previously, here for the first time, fine Pt/Cu alloy nanoparticles (PtCu(3) NPs) are integrated with chloride ion transporter (CIT) for EDT-based combinational therapy. In this system, while PtCu(3) NPs induce oxidative species under an electric field, it also effectively transforms endogenous H(2)O(2) into •OH and consumes intracellular glutathione (GSH). More importantly, with the aid of CIT, PtCu(3)-PEG@CIT NPs promote the intracellular concentration of chloride ion (Cl(−)) by transporting extracellular Cl(−), facilitating the generation of oxidative species considerably. Meanwhile, CIT delivered intracellularly increases lysosomal pH, leading to the disruption of cellular autophagy and weakening the treatment resistance. In consequence, significant tumor inhibition is enabled both in vitro and in vivo, due to the combination of unique characteristics offered by PtCu(3)-PEG@CIT.
format Online
Article
Text
id pubmed-8897047
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher KeAi Publishing
record_format MEDLINE/PubMed
spelling pubmed-88970472022-03-17 Platinum–copper alloy nanoparticles armored with chloride ion transporter to promote electro-driven tumor inhibition Chen, Tong Han, Gaorong Li, Xiang Bioact Mater Article The induction of oxidative species, driven by oscillating electric field (E), has recently emerged as an effective approach for tumor inhibition, so-called electrodynamic therapy (EDT). While it offers a series of advantages attracting considerable attention, the fundamental mechanism and improvement strategies for EDT approach are being endeavored extensively with the aid of new material explorations. An interesting phenomenon observed in early studies is that the on-site concentration of chloride ion is highly favored for the induction of oxidative species and the efficacy of tumor inhibition. Following this discovery ignored previously, here for the first time, fine Pt/Cu alloy nanoparticles (PtCu(3) NPs) are integrated with chloride ion transporter (CIT) for EDT-based combinational therapy. In this system, while PtCu(3) NPs induce oxidative species under an electric field, it also effectively transforms endogenous H(2)O(2) into •OH and consumes intracellular glutathione (GSH). More importantly, with the aid of CIT, PtCu(3)-PEG@CIT NPs promote the intracellular concentration of chloride ion (Cl(−)) by transporting extracellular Cl(−), facilitating the generation of oxidative species considerably. Meanwhile, CIT delivered intracellularly increases lysosomal pH, leading to the disruption of cellular autophagy and weakening the treatment resistance. In consequence, significant tumor inhibition is enabled both in vitro and in vivo, due to the combination of unique characteristics offered by PtCu(3)-PEG@CIT. KeAi Publishing 2021-10-13 /pmc/articles/PMC8897047/ /pubmed/35310378 http://dx.doi.org/10.1016/j.bioactmat.2021.10.012 Text en © 2021 The Authors https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Article
Chen, Tong
Han, Gaorong
Li, Xiang
Platinum–copper alloy nanoparticles armored with chloride ion transporter to promote electro-driven tumor inhibition
title Platinum–copper alloy nanoparticles armored with chloride ion transporter to promote electro-driven tumor inhibition
title_full Platinum–copper alloy nanoparticles armored with chloride ion transporter to promote electro-driven tumor inhibition
title_fullStr Platinum–copper alloy nanoparticles armored with chloride ion transporter to promote electro-driven tumor inhibition
title_full_unstemmed Platinum–copper alloy nanoparticles armored with chloride ion transporter to promote electro-driven tumor inhibition
title_short Platinum–copper alloy nanoparticles armored with chloride ion transporter to promote electro-driven tumor inhibition
title_sort platinum–copper alloy nanoparticles armored with chloride ion transporter to promote electro-driven tumor inhibition
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8897047/
https://www.ncbi.nlm.nih.gov/pubmed/35310378
http://dx.doi.org/10.1016/j.bioactmat.2021.10.012
work_keys_str_mv AT chentong platinumcopperalloynanoparticlesarmoredwithchlorideiontransportertopromoteelectrodriventumorinhibition
AT hangaorong platinumcopperalloynanoparticlesarmoredwithchlorideiontransportertopromoteelectrodriventumorinhibition
AT lixiang platinumcopperalloynanoparticlesarmoredwithchlorideiontransportertopromoteelectrodriventumorinhibition