4.7 Article

Biodegradable hollow mesoporous organosilica nanotheranostics (HMON) for multi-mode imaging and mild photo-therapeutic-induced mitochondrial damage on gastric cancer

Journal

JOURNAL OF NANOBIOTECHNOLOGY
Volume 18, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s12951-020-00653-y

Keywords

Hollow mesoporous organosilica nanoparticles (HMON); Photo-thermal therapy (PTT); Photo-dynamic therapy (PDT); Multi-modal imaging; Mitochondrial damage

Funding

  1. National Natural Science Foundation of China [81601550, 81971746]
  2. China Postdoctoral Science Foundation [2019M662977, 2019M662986]
  3. Special Funds for the Cultivation of Guangdong College Students' Scientific and Technological Innovation [pdjh2020a0109, pdjh2020a0108, pdjh2020a0106]
  4. Guangzhou City Science and Technology Project-Zhujiang Technology New Star Project [20160521606488]
  5. Natural Science Foundation of Guangdong Province [2017A030306023, 2019A1515110635]
  6. Nature Science Foundation of Guangdong Province [A2020297, A2020131]
  7. Science and Technology Project of Guangdong Province [2018B090944002]
  8. National Key R&D Program of China [2017YFC1103400]
  9. Outstanding Youths Development Scheme of Nanfang Hospital, Southern Medical University [2017J006]

Ask authors/readers for more resources

Background CuS-modified hollow mesoporous organosilica nanoparticles (HMON@CuS) have been preferred as non-invasive treatment for cancer, as near infrared (NIR)-induced photo-thermal effect (PTT) and/or photo-dynamic effect (PDT) could increase cancer cells' apoptosis. However, the certain role of HMON@CuS-produced-PTT&PDT inducing gastric cancer (GC) cells' mitochondrial damage, remained unclear. Moreover, theranostic efficiency of HMON@CuS might be well improved by applying multi-modal imaging, which could offer an optimal therapeutic region and time window. Herein, new nanotheranostics agents were reported by Gd doped HMON decorated by CuS nanocrystals (called HMON@CuS/Gd). Results HMON@CuS/Gd exhibited appropriate size distribution, good biocompatibility,l-Glutathione (GSH) responsive degradable properties, high photo-thermal conversion efficiency (82.4%) and a simultaneous reactive oxygen species (ROS) generation effect. Meanwhile, HMON@CuS/Gd could efficiently enter GC cells, induce combined mild PTT (43-45 degrees C) and PDT under mild NIR power density (0.8 W/cm(2)). Surprisingly, it was found that PTT might not be the only factor of cell apoptosis, as ROS induced by PDT also seemed playing an essential role. The NIR-induced ROS could attack mitochondrial transmembrane potentials (MTPs), then promote mitochondrial reactive oxygen species (mitoROS) production. Meanwhile, mitochondrial damage dramatically changed the expression of anti-apoptotic protein (Bcl-2) and pro-apoptotic protein (Bax). Since that, mitochondrial permeability transition pore (mPTP) was opened, followed by inducing more cytochrome c (Cyto C) releasing from mitochondria into cytosol, and finally activated caspase-9/caspase-3-depended cell apoptosis pathway. Our in vivo data also showed that HMON@CuS/Gd exhibited good fluorescence (FL) imaging (wrapping fluorescent agent), enhanced T1 imaging under magnetic resonance imaging (MRI) and infrared thermal (IRT) imaging capacities. Guided by FL/MRI/IRT trimodal imaging, HMON@CuS/Gd could selectively cause mild photo-therapy at cancer region, efficiently inhibit the growth of GC cells without evident systemic toxicity in vivo. Conclusion HMON@CuS/Gd could serve as a promising multifunctional nanotheranostic platform and as a cancer photo-therapy agent through inducing mitochondrial dysfunction on GC.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available