4.7 Article

Dual Targeting of Endoplasmic Reticulum by Redox-Deubiquitination Regulation for Cancer Therapy

期刊

INTERNATIONAL JOURNAL OF NANOMEDICINE
卷 16, 期 -, 页码 5193-5209

出版社

DOVE MEDICAL PRESS LTD
DOI: 10.2147/IJN.S321612

关键词

endoplasmic reticulum stress; unfolded protein response; nanocatalytic medicine; reactive oxygen species; deubiquitinase inhibitor; apoptosis

资金

  1. National Natural Science Foundation of China [81901747, 81571729]
  2. National Key Research and Development Program of China [2017YFE0124400, 2020YFA0909000]
  3. Shanghai Science and Technology Commission Research Project [18ZR1423200]
  4. Shanghai Municipal Education Commission [2019-01-07-00-02E00064]
  5. Med-Engineering Crossing Foundation from Shanghai Jiao Tong University [YG2017ZD05]
  6. Bethune Charitable Foundation [BCF-NH-ZL-20201119-024]
  7. Incubating Program for Clinical Research and Innovation of Renji Hospital [PYXJS16-008, PYIII20-07]

向作者/读者索取更多资源

The study utilized a phase change material to encapsulate iron oxide nanoparticles and a UPR modulator for enhanced cancer therapy. The theranostic platform showed excellent tumor-targeting properties and effective photothermal-enhanced tumor inhibition both in vitro and in vivo. By inducing ER stress and apoptosis in cancer cells through redox imbalance, the strategy offers novel directions for treating therapy-resistant cancers.
Background: Recently, nanocatalyst-induced endoplasmic reticulum (ER) stress for cancer therapy has been attracting considerable attention. However, cancer cells are often able to overcome ER stress-induced death by activating the unfolded protein response (UPR), making nanocatalytic monotherapy a poor defense against cancer progression. Purpose: In this study, to improve the nanocatalytic treatment efficacy, a phase change material (PCM) was used to encapsulate the upstream ER stress initiator, iron oxide nanoparticles (Fe3O4 NPs), and the downstream UPR modulator, PR-619. Subsequently, the tumor-homing peptide tLyP-1 was coupled to it to form tLyP-1/PR-619/Fe3O4@PCM (tPF@PCM) theranostic platform. Materials and Methods: tPF@PCM was synthesized using nanoprecipitation and resolidification methods followed by the EDC/NHS cross-linking method. The targeting capacity of tPF@PCM was evaluated in vitro and in vivo using flow cytometry and magnetic resonance imaging, respectively. The therapeutic efficacy of tPF@PCM was investigated in a renal cell carcinoma mouse model. Moreover, we explored the synergistic anti-tumor mechanism by examining the intracellular reactive oxygen species (ROS), aggregated proteins, ER stress response levels, and type of cell death. Results: tPF@PCM had excellent tumor-targeting properties and exhibited satisfactory photothermal-enhanced tumor inhibition efficacy both in vitro and in vivo. Specifically, the phase transition temperature (45 degrees C) maintained using 808 nm laser irradiation significantly increased the release and catalytic activity of the peroxidase mimic Fe3O4 NPs. This strongly catalyzed the generation of hydroxyl radicals (center dot OH) via the Fenton reaction in the acidic tumor microenvironment. The redox imbalance subsequently resulted in an increase in the level of damaged proteins in the ER and initiated ER stress. Moreover, the pan-deubiquitinase inhibitor PR-619 blocked the adaptive UPR-mediated degradation of these damaged proteins, exacerbating the ER burden. Consequently, irremediable ER stress activated the terminal UPR, leading to apoptosis in cancer cells. Conclusion: This ER stress-exacerbating strategy effectively suppresses tumorigenesis, offering novel directions for advances in the treatment of conventional therapy-resistant cancers.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据