4.6 Article

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

Journal

CANCER IMMUNOLOGY RESEARCH
Volume 3, Issue 8, Pages 902-914

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/2326-6066.CIR-14-0219

Keywords

-

Funding

  1. Ministry of Education, Science, and Culture (MEXT)
  2. Carcinogenic Spiral a MEXT Grant-in-Project
  3. Ministry of Health, Labor, and Welfare of Japan
  4. Takeda Foundation
  5. Yasuda Cancer Research Foundation
  6. Kato Memorial Bioscience Foundation
  7. Grants-in-Aid for Scientific Research [15K08517] Funding Source: KAKEN

Ask authors/readers for more resources

Double-stranded RNA directly acts on fibroblast and myeloid lineages to induce necroptosis as in TNF alpha. 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 TNF alpha 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 mg) 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.

Authors

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

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available