Autophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeutics

Abstract Ultrasound-triggered sonodynamic therapy (SDT) represents an emerging therapeutic modality for cancer treatment based on its specific feature of noninvasiveness, high tissue-penetrating depth and desirable therapeutic efficacy, but the SDT-induced pro-survival cancer-cell autophagy would si...

Full description

Bibliographic Details
Main Authors: Liqiang Zhou, Minfeng Huo, Xiaoqin Qian, Li Ding, Luodan Yu, Wei Feng, Xinwu Cui, Yu Chen
Format: Article
Language:English
Published: BMC 2021-04-01
Series:Journal of Nanobiotechnology
Subjects:
Online Access:https://doi.org/10.1186/s12951-021-00855-y
id doaj-58cc01a2c90e4b9e8dfe0087902e7126
record_format Article
spelling doaj-58cc01a2c90e4b9e8dfe0087902e71262021-04-25T11:08:16ZengBMCJournal of Nanobiotechnology1477-31552021-04-0119111510.1186/s12951-021-00855-yAutophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeuticsLiqiang Zhou0Minfeng Huo1Xiaoqin Qian2Li Ding3Luodan Yu4Wei Feng5Xinwu Cui6Yu Chen7Sino-German Tongji-Caritas Research Center of Ultrasound in Medicine, Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyState Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of SciencesDepartment of Ultrasound, Affiliated People’s Hospital of Jiangsu UniversityState Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of SciencesSchool of Life Sciences, Shanghai UniversitySchool of Life Sciences, Shanghai UniversitySino-German Tongji-Caritas Research Center of Ultrasound in Medicine, Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologySchool of Life Sciences, Shanghai UniversityAbstract Ultrasound-triggered sonodynamic therapy (SDT) represents an emerging therapeutic modality for cancer treatment based on its specific feature of noninvasiveness, high tissue-penetrating depth and desirable therapeutic efficacy, but the SDT-induced pro-survival cancer-cell autophagy would significantly lower the SDT efficacy for cancer treatment. Here we propose an “all-in-one” combined tumor-therapeutic strategy by integrating nanosonosensitizers-augmented noninvasive SDT with autophagy inhibition based on the rationally constructed nanoliposomes that co-encapsulates clinically approved sonosensitizers protoporphyrin IX (PpIX) and early-phase autophagy-blocking agent 3-methyladenine (3-MA). It has been systematically demonstrated that nanosonosensitizers-augmented SDT induced cytoprotective pro-survival autophagy through activation of MAPK signaling pathway and inhibition of AMPK signaling pathway, and this could be efficaciously inhibited by 3-MA in early-phase autophagy, which significantly decreased the cell resistance to intracellular oxidative stress and complied a remarkable synergistic effect on SDT medicated cancer-cell apoptosis both in vitro at cellular level and in vivo on tumor-bearing animal model. Therefore, our results provide a proof-of-concept combinatorial tumor therapeutics based on nanosonosensitizers for the treatment of ROS-resistant cancer by autophagy inhibition-augmented SDT.https://doi.org/10.1186/s12951-021-00855-yAutophagySonodynamic therapyAutophagy inhibitionNanoliposomesTumor therapy
collection DOAJ
language English
format Article
sources DOAJ
author Liqiang Zhou
Minfeng Huo
Xiaoqin Qian
Li Ding
Luodan Yu
Wei Feng
Xinwu Cui
Yu Chen
spellingShingle Liqiang Zhou
Minfeng Huo
Xiaoqin Qian
Li Ding
Luodan Yu
Wei Feng
Xinwu Cui
Yu Chen
Autophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeutics
Journal of Nanobiotechnology
Autophagy
Sonodynamic therapy
Autophagy inhibition
Nanoliposomes
Tumor therapy
author_facet Liqiang Zhou
Minfeng Huo
Xiaoqin Qian
Li Ding
Luodan Yu
Wei Feng
Xinwu Cui
Yu Chen
author_sort Liqiang Zhou
title Autophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeutics
title_short Autophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeutics
title_full Autophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeutics
title_fullStr Autophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeutics
title_full_unstemmed Autophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeutics
title_sort autophagy blockade synergistically enhances nanosonosensitizer-enabled sonodynamic cancer nanotherapeutics
publisher BMC
series Journal of Nanobiotechnology
issn 1477-3155
publishDate 2021-04-01
description Abstract Ultrasound-triggered sonodynamic therapy (SDT) represents an emerging therapeutic modality for cancer treatment based on its specific feature of noninvasiveness, high tissue-penetrating depth and desirable therapeutic efficacy, but the SDT-induced pro-survival cancer-cell autophagy would significantly lower the SDT efficacy for cancer treatment. Here we propose an “all-in-one” combined tumor-therapeutic strategy by integrating nanosonosensitizers-augmented noninvasive SDT with autophagy inhibition based on the rationally constructed nanoliposomes that co-encapsulates clinically approved sonosensitizers protoporphyrin IX (PpIX) and early-phase autophagy-blocking agent 3-methyladenine (3-MA). It has been systematically demonstrated that nanosonosensitizers-augmented SDT induced cytoprotective pro-survival autophagy through activation of MAPK signaling pathway and inhibition of AMPK signaling pathway, and this could be efficaciously inhibited by 3-MA in early-phase autophagy, which significantly decreased the cell resistance to intracellular oxidative stress and complied a remarkable synergistic effect on SDT medicated cancer-cell apoptosis both in vitro at cellular level and in vivo on tumor-bearing animal model. Therefore, our results provide a proof-of-concept combinatorial tumor therapeutics based on nanosonosensitizers for the treatment of ROS-resistant cancer by autophagy inhibition-augmented SDT.
topic Autophagy
Sonodynamic therapy
Autophagy inhibition
Nanoliposomes
Tumor therapy
url https://doi.org/10.1186/s12951-021-00855-y
work_keys_str_mv AT liqiangzhou autophagyblockadesynergisticallyenhancesnanosonosensitizerenabledsonodynamiccancernanotherapeutics
AT minfenghuo autophagyblockadesynergisticallyenhancesnanosonosensitizerenabledsonodynamiccancernanotherapeutics
AT xiaoqinqian autophagyblockadesynergisticallyenhancesnanosonosensitizerenabledsonodynamiccancernanotherapeutics
AT liding autophagyblockadesynergisticallyenhancesnanosonosensitizerenabledsonodynamiccancernanotherapeutics
AT luodanyu autophagyblockadesynergisticallyenhancesnanosonosensitizerenabledsonodynamiccancernanotherapeutics
AT weifeng autophagyblockadesynergisticallyenhancesnanosonosensitizerenabledsonodynamiccancernanotherapeutics
AT xinwucui autophagyblockadesynergisticallyenhancesnanosonosensitizerenabledsonodynamiccancernanotherapeutics
AT yuchen autophagyblockadesynergisticallyenhancesnanosonosensitizerenabledsonodynamiccancernanotherapeutics
_version_ 1721509997803732992