Theranostics 2018; 8(11):3138-3152. doi:10.7150/thno.21693 This issue Cite

Research Paper

Negative regulation of cationic nanoparticle-induced inflammatory toxicity through the increased production of prostaglandin E2 via mitochondrial DNA-activated Ly6C+ monocytes

Li Liu*, Yantong Liu*, Bocheng Xu, Chuyu Liu, Yanpeng Jia, Ting Liu, Chunju Fang, Wei Wang, Jun Ren, Zhiyao He, Ke Men, Xiao Liang, Min Luo, Bin Shao, Ye Mao, Henyi Xiao, Zhiyong Qian, Jia Geng, Birong Dong, Peng Mi, Yu Jiang, Yuquan Wei, Xiawei Wei

Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
*These authors contributed equally to this work.

Citation:
Liu L, Liu Y, Xu B, Liu C, Jia Y, Liu T, Fang C, Wang W, Ren J, He Z, Men K, Liang X, Luo M, Shao B, Mao Y, Xiao H, Qian Z, Geng J, Dong B, Mi P, Jiang Y, Wei Y, Wei X. Negative regulation of cationic nanoparticle-induced inflammatory toxicity through the increased production of prostaglandin E2 via mitochondrial DNA-activated Ly6C+ monocytes. Theranostics 2018; 8(11):3138-3152. doi:10.7150/thno.21693. https://www.thno.org/v08p3138.htm
Other styles

File import instruction

Abstract

Graphic abstract

Rationale: Cationic nanocarriers present with well-known toxicities, including inflammatory toxicity, which limit their clinical application. How the cationic nanocarrier-induced inflammatory response is negatively regulated is unknown. Herein, we found that following a sublethal dose of cationic nanocarriers, the induced inflammatory response is characterized by early neutrophil infiltration and spontaneous resolution within 1 week.

Methods: C57BL/6 mice were intravenously injected with a dosage of 1-100 mg/kg cationic DOTAP liposomes as well as other cationic materials. Cell necrosis was detected by flow cytometry. Release of mitochondrial DNA was quantified by qPCR via Taqman probes. Signal proteins were detected by Western blotting. PGE2 production in the supernatant was quantitated using an enzyme immunoassay (EIA). The infiltrated inflammatory cells were observed in WT mice, Ccr2-/- mice, Sting-/-mice and Tlr9-/-mice.

Results: The early stage (24-48 h) inflammatory neutrophil infiltration was followed by an increasing percentage of monocytes; and, compared with WT mice, Ccr2-/- mice presented with more severe pulmonary inflammation. A previously uncharacterized population of regulatory monocytes expressing both inflammatory and immunosuppressive cytokines was identified in this model. The alteration in monocyte phenotype was directly induced by mtDNA release from cationic nanocarrier-induced necrotic cells via a STING- or TLR9-dependent pathway. Neutrophil activation was specifically inhibited by PGE2 from Ly6C+ inflammatory monocytes, and intravenous injections of dual-phenotype monocytes beneficially modified the immune response; this inhibitory effect was abolished after treatment with indomethacin. Moreover, we provide clear evidence that mitochondrial DNA activated Ly6C+ monocytes and increased PGE2 production through TLR9- or STING-mediated MAPK-NF-κB-COX2 pathways.

Conclusion: Our findings suggest that Ly6C+ monocytes and mtDNA-induced Ly6C+ monocyte PGE2 production may be part of a feedback mechanism that contributes to the resolution of cationic nanocarrier-induced inflammatory toxicity and may have important implications for understanding nanoparticle biocompatibility and designing better, safer drug delivery systems.

Keywords: cationic nanocarriers, inflammatory toxicity, prostaglandin E2, monocytes, mitochondrial DNA (mtDNA)


Citation styles

APA
Liu, L., Liu, Y., Xu, B., Liu, C., Jia, Y., Liu, T., Fang, C., Wang, W., Ren, J., He, Z., Men, K., Liang, X., Luo, M., Shao, B., Mao, Y., Xiao, H., Qian, Z., Geng, J., Dong, B., Mi, P., Jiang, Y., Wei, Y., Wei, X. (2018). Negative regulation of cationic nanoparticle-induced inflammatory toxicity through the increased production of prostaglandin E2 via mitochondrial DNA-activated Ly6C+ monocytes. Theranostics, 8(11), 3138-3152. https://doi.org/10.7150/thno.21693.

ACS
Liu, L.; Liu, Y.; Xu, B.; Liu, C.; Jia, Y.; Liu, T.; Fang, C.; Wang, W.; Ren, J.; He, Z.; Men, K.; Liang, X.; Luo, M.; Shao, B.; Mao, Y.; Xiao, H.; Qian, Z.; Geng, J.; Dong, B.; Mi, P.; Jiang, Y.; Wei, Y.; Wei, X. Negative regulation of cationic nanoparticle-induced inflammatory toxicity through the increased production of prostaglandin E2 via mitochondrial DNA-activated Ly6C+ monocytes. Theranostics 2018, 8 (11), 3138-3152. DOI: 10.7150/thno.21693.

NLM
Liu L, Liu Y, Xu B, Liu C, Jia Y, Liu T, Fang C, Wang W, Ren J, He Z, Men K, Liang X, Luo M, Shao B, Mao Y, Xiao H, Qian Z, Geng J, Dong B, Mi P, Jiang Y, Wei Y, Wei X. Negative regulation of cationic nanoparticle-induced inflammatory toxicity through the increased production of prostaglandin E2 via mitochondrial DNA-activated Ly6C+ monocytes. Theranostics 2018; 8(11):3138-3152. doi:10.7150/thno.21693. https://www.thno.org/v08p3138.htm

CSE
Liu L, Liu Y, Xu B, Liu C, Jia Y, Liu T, Fang C, Wang W, Ren J, He Z, Men K, Liang X, Luo M, Shao B, Mao Y, Xiao H, Qian Z, Geng J, Dong B, Mi P, Jiang Y, Wei Y, Wei X. 2018. Negative regulation of cationic nanoparticle-induced inflammatory toxicity through the increased production of prostaglandin E2 via mitochondrial DNA-activated Ly6C+ monocytes. Theranostics. 8(11):3138-3152.

This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
Popup Image