4.7 Article

Neuropeptide-Functionalized Gold Nanorod Enhanced Cellular Uptake and Improved In Vitro Photothermal Killing in LRP1-Positive Glioma Cells

期刊

PHARMACEUTICS
卷 14, 期 9, 页码 -

出版社

MDPI
DOI: 10.3390/pharmaceutics14091939

关键词

glioma; blood-brain barrier; neuropeptide (ANGIOPEP-2); gold nanorod; photothermal therapy; hyperthermia

资金

  1. Ministry of Science and Technology [MOST108-2221-B-037-002-MY3, MOST109-2320-B-037017-MY3]
  2. Kaohsiung Medical University [NK111P26, DK109001]

向作者/读者索取更多资源

This study synthesized a neuropeptide-functionalized gold nanorod and evaluated its cellular uptake and photothermal effects on glioma cells. The results showed that the functionalized nanorods had higher cellular uptake and induced higher rates of apoptosis and ROS production under laser irradiation. The study provides insights into the effective time point for maximum cellular uptake and potential improvements in anti-glioma therapy.
The therapeutic modalities for glioblastoma multiforme fail badly due to the limitations of poor penetration through the blood-brain barrier and the lack of tumor targeting. In this study, we synthesized a neuropeptide (ANGIOPEP-2)-functionalized gold nanorod (GNR-ANGI-2) and systemically evaluated the cellular uptake and photothermal effects enhanced by the neuropeptide functionalization of the gold nanorod under laser or sham exposure. The expression of LRP1, the specific ligand for ANGIOPEP-2, was the highest in C6 cells among five studied glioma cell lines. The cellular internalization studies showed higher uptake of gold nanorods functionalized with ANGIOPEP-2 than of those functionalized with scrambled ANGIOPEP-2. The in vitro photothermal studies of C6 cells treated with GNR-ANGI-2 and laser showed a higher rate of apoptosis at early and late stages than cells treated with GNR-ANGI-2 without laser. Correspondingly, in vitro ROS evaluation showed a higher intensity of ROS production in cells treated with GNR-ANGI-2 under laser irradiation. The Western blotting results indicated that GNR-ANGI-2 with laser exposure activated the caspase pathway of apoptosis, and GNR-ANGI-2 with sham exposure induced autophagy in C6 cells. The current study provides in-depth knowledge on the effective time point for maximum cellular uptake of GNR-ANGI-2 to achieve a better anti-glioma effect. Moreover, by exploring the molecular mechanism of cell death with GNR-ANGI-2-mediated photothermal therapy, we could modify the nanoshuttle with multimodal targets to achieve more efficient anti-glioma therapy in the future.

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