HUSCAP logo Hokkaido Univ. logo

Hokkaido University Collection of Scholarly and Academic Papers >
Graduate School of Medicine / Faculty of Medicine >
Peer-reviewed Journal Articles, etc >

PolyI:C-Induced, TLR3/RIP3-Dependent Necroptosis Backs Up Immune Effector-Mediated Tumor Elimination In Vivo

Files in This Item:
141387_1_merged_1426906273.pdf7.03 MBPDFView/Open
Please use this identifier to cite or link to this item:http://hdl.handle.net/2115/62581

Title: PolyI:C-Induced, TLR3/RIP3-Dependent Necroptosis Backs Up Immune Effector-Mediated Tumor Elimination In Vivo
Other Titles: TICAM-1-RIP3 necroptosis in tumor
Authors: Takemura, Ryo Browse this author
Takaki, Hiromi Browse this author
Okada, Seiji Browse this author →KAKEN DB
Shime, Hiroaki Browse this author →KAKEN DB
Akazawa, Takashi Browse this author →KAKEN DB
Oshiumi, Hiroyuki Browse this author →KAKEN DB
Matsumoto, Misako Browse this author →KAKEN DB
Teshima, Takanori Browse this author →KAKEN DB
Seya, Tsukasa Browse this author →KAKEN DB
Keywords: RNA (polyI:C)
TLR3
TICAM-1
necroptosis
RIP kinases
Issue Date: Aug-2015
Publisher: American Association for Cancer Research
Journal Title: Cancer immunology research
Volume: 3
Issue: 8
Start Page: 902
End Page: 914
Publisher DOI: 10.1158/2326-6066.CIR-14-0219
PMID: 25898986
Abstract: Double-stranded RNA directly acts on fibroblast and myeloid lineages to induce necroptosis as in TNFα. Here, we investigated whether this type of cell death occurred in cancer cells in response to polyinosinic-polycytidylic acid (polyI:C) and the pan-caspase inhibitor z-Val-Ala-Asp fluromethyl ketone (zVAD). We found that the colon cancer cell line CT26 is highly susceptible to necroptosis, as revealed by staining with annexin V/propidium iodide. CT26 cells possess RNA sensors, TLR3 and MDA5, which are upregulated by interferon (IFN)-inducing pathways and linked to receptor-interacting protein kinase (RIP) 1/3 activation via TICAM-1 or MAVS adaptor, respectively. Although exogenously added polyI:C alone marginally induced necroptosis in CT26 cells, a combined regimen of polyI:C and zVAD induced approximately 50% CT26 necroptosis in vitro without secondary effects of TNFα or type I IFNs. CT26 necroptosis depended on the TLR3-TICAM-1-RIP3 axis in the tumor cells to produce reactive oxygen species, but not on MDA5, MAVS, or the caspases/inflammasome activation. However, the RNA-derived necroptosis was barely reproduced in vivo in a CT26 tumor-implanted Balb/c mouse model with administration of polyI:C + zVAD. Significant shrinkage of CT26 tumors was revealed only when polyI:C (100 μg) was injected intraperitoneally and zVAD (1 mg) subcutaneously into tumor-bearing mice that were depleted of cytotoxic T lymphocytes and natural killer cells. The results were confirmed with immune-compromised mice with no lymphocytes. Although necroptosis-induced tumor growth retardation appears mechanistically complicated and dependent on the injection routes of polyI:C and zVAD, anti-caspase reagent directed to tumor cells will make RNA adjuvant immunotherapy more effective by modulating the formation of the tumoricidal microenvironment and dendritic cell-inducing antitumor immune system.
Type: article (author version)
URI: http://hdl.handle.net/2115/62581
Appears in Collections:医学院・医学研究院 (Graduate School of Medicine / Faculty of Medicine) > 雑誌発表論文等 (Peer-reviewed Journal Articles, etc)

Submitter: 瀬谷 司

Export metadata:

OAI-PMH ( junii2 , jpcoar_1.0 )

MathJax is now OFF:


 

 - Hokkaido University