Cargando…

Metal-organic framework-encapsulated dihydroartemisinin nanoparticles induces apoptotic cell death in ovarian cancer by blocking ROMO1-mediated ROS production

Dihydroartemisinin (DHA), a natural product derived from the herbal medicine Artemisia annua, is recently used as a novel anti-cancer agent. However, some intrinsic disadvantages limit its potential for clinical management of cancer patients, such as poor water solubility and low bioavailability. No...

Descripción completa

Detalles Bibliográficos
Autores principales: Yan, Yuanliang, Yang, Xiaoxin, Han, Ning, Liu, Yuanhong, Liang, Qiuju, Li, Liu-Gen, Hu, Jun, Li, Tong-Fei, Xu, Zhijie
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10308639/
https://www.ncbi.nlm.nih.gov/pubmed/37386404
http://dx.doi.org/10.1186/s12951-023-01959-3
_version_ 1785066287263645696
author Yan, Yuanliang
Yang, Xiaoxin
Han, Ning
Liu, Yuanhong
Liang, Qiuju
Li, Liu-Gen
Hu, Jun
Li, Tong-Fei
Xu, Zhijie
author_facet Yan, Yuanliang
Yang, Xiaoxin
Han, Ning
Liu, Yuanhong
Liang, Qiuju
Li, Liu-Gen
Hu, Jun
Li, Tong-Fei
Xu, Zhijie
author_sort Yan, Yuanliang
collection PubMed
description Dihydroartemisinin (DHA), a natural product derived from the herbal medicine Artemisia annua, is recently used as a novel anti-cancer agent. However, some intrinsic disadvantages limit its potential for clinical management of cancer patients, such as poor water solubility and low bioavailability. Nowadays, the nanoscale drug delivery system emerges as a hopeful platform for improve the anti-cancer treatment. Accordingly, a metal-organic framework (MOF) based on zeolitic imidazolate framework-8 was designed and synthesized to carry DHA in the core (ZIF-DHA). Contrast with free DHA, these prepared ZIF-DHA nanoparticles (NPs) displayed preferable anti-tumor therapeutic activity in several ovarian cancer cells accompanied with suppressed production of cellular reactive oxygen species (ROS) and induced apoptotic cell death. 4D-FastDIA-based mass spectrometry technology indicated that down-regulated reactive oxygen species modulator 1 (ROMO1) might be regarded as potential therapeutic targets for ZIF-DHA NPs. Overexpression of ROMO1 in ovarian cancer cells significantly reversed the cellular ROS-generation induced by ZIF-DHA, as well as the pro-apoptosis effects. Taken together, our study elucidated and highlighted the potential of zeolitic imidazolate framework-8-based MOF to improve the activity of DHA to treat ovarian cancer. Our findings suggested that these prepared ZIF-DHA NPs could be an attractive therapeutic strategy for ovarian cancer. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-023-01959-3.
format Online
Article
Text
id pubmed-10308639
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-103086392023-06-30 Metal-organic framework-encapsulated dihydroartemisinin nanoparticles induces apoptotic cell death in ovarian cancer by blocking ROMO1-mediated ROS production Yan, Yuanliang Yang, Xiaoxin Han, Ning Liu, Yuanhong Liang, Qiuju Li, Liu-Gen Hu, Jun Li, Tong-Fei Xu, Zhijie J Nanobiotechnology Research Dihydroartemisinin (DHA), a natural product derived from the herbal medicine Artemisia annua, is recently used as a novel anti-cancer agent. However, some intrinsic disadvantages limit its potential for clinical management of cancer patients, such as poor water solubility and low bioavailability. Nowadays, the nanoscale drug delivery system emerges as a hopeful platform for improve the anti-cancer treatment. Accordingly, a metal-organic framework (MOF) based on zeolitic imidazolate framework-8 was designed and synthesized to carry DHA in the core (ZIF-DHA). Contrast with free DHA, these prepared ZIF-DHA nanoparticles (NPs) displayed preferable anti-tumor therapeutic activity in several ovarian cancer cells accompanied with suppressed production of cellular reactive oxygen species (ROS) and induced apoptotic cell death. 4D-FastDIA-based mass spectrometry technology indicated that down-regulated reactive oxygen species modulator 1 (ROMO1) might be regarded as potential therapeutic targets for ZIF-DHA NPs. Overexpression of ROMO1 in ovarian cancer cells significantly reversed the cellular ROS-generation induced by ZIF-DHA, as well as the pro-apoptosis effects. Taken together, our study elucidated and highlighted the potential of zeolitic imidazolate framework-8-based MOF to improve the activity of DHA to treat ovarian cancer. Our findings suggested that these prepared ZIF-DHA NPs could be an attractive therapeutic strategy for ovarian cancer. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-023-01959-3. BioMed Central 2023-06-29 /pmc/articles/PMC10308639/ /pubmed/37386404 http://dx.doi.org/10.1186/s12951-023-01959-3 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Yan, Yuanliang
Yang, Xiaoxin
Han, Ning
Liu, Yuanhong
Liang, Qiuju
Li, Liu-Gen
Hu, Jun
Li, Tong-Fei
Xu, Zhijie
Metal-organic framework-encapsulated dihydroartemisinin nanoparticles induces apoptotic cell death in ovarian cancer by blocking ROMO1-mediated ROS production
title Metal-organic framework-encapsulated dihydroartemisinin nanoparticles induces apoptotic cell death in ovarian cancer by blocking ROMO1-mediated ROS production
title_full Metal-organic framework-encapsulated dihydroartemisinin nanoparticles induces apoptotic cell death in ovarian cancer by blocking ROMO1-mediated ROS production
title_fullStr Metal-organic framework-encapsulated dihydroartemisinin nanoparticles induces apoptotic cell death in ovarian cancer by blocking ROMO1-mediated ROS production
title_full_unstemmed Metal-organic framework-encapsulated dihydroartemisinin nanoparticles induces apoptotic cell death in ovarian cancer by blocking ROMO1-mediated ROS production
title_short Metal-organic framework-encapsulated dihydroartemisinin nanoparticles induces apoptotic cell death in ovarian cancer by blocking ROMO1-mediated ROS production
title_sort metal-organic framework-encapsulated dihydroartemisinin nanoparticles induces apoptotic cell death in ovarian cancer by blocking romo1-mediated ros production
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10308639/
https://www.ncbi.nlm.nih.gov/pubmed/37386404
http://dx.doi.org/10.1186/s12951-023-01959-3
work_keys_str_mv AT yanyuanliang metalorganicframeworkencapsulateddihydroartemisininnanoparticlesinducesapoptoticcelldeathinovariancancerbyblockingromo1mediatedrosproduction
AT yangxiaoxin metalorganicframeworkencapsulateddihydroartemisininnanoparticlesinducesapoptoticcelldeathinovariancancerbyblockingromo1mediatedrosproduction
AT hanning metalorganicframeworkencapsulateddihydroartemisininnanoparticlesinducesapoptoticcelldeathinovariancancerbyblockingromo1mediatedrosproduction
AT liuyuanhong metalorganicframeworkencapsulateddihydroartemisininnanoparticlesinducesapoptoticcelldeathinovariancancerbyblockingromo1mediatedrosproduction
AT liangqiuju metalorganicframeworkencapsulateddihydroartemisininnanoparticlesinducesapoptoticcelldeathinovariancancerbyblockingromo1mediatedrosproduction
AT liliugen metalorganicframeworkencapsulateddihydroartemisininnanoparticlesinducesapoptoticcelldeathinovariancancerbyblockingromo1mediatedrosproduction
AT hujun metalorganicframeworkencapsulateddihydroartemisininnanoparticlesinducesapoptoticcelldeathinovariancancerbyblockingromo1mediatedrosproduction
AT litongfei metalorganicframeworkencapsulateddihydroartemisininnanoparticlesinducesapoptoticcelldeathinovariancancerbyblockingromo1mediatedrosproduction
AT xuzhijie metalorganicframeworkencapsulateddihydroartemisininnanoparticlesinducesapoptoticcelldeathinovariancancerbyblockingromo1mediatedrosproduction