In vivo OVA-specific Cytotoxic CD8+ T Cell Killing Assay

BIO-PROTOCOL ◽  
2016 ◽  
Vol 6 (12) ◽  
Author(s):  
Nada Chaoul ◽  
Catherine Fayolle ◽  
Claude Leclerc
Keyword(s):  
T Cell ◽  
BIO-PROTOCOL ◽  
2018 ◽  
Vol 8 (22) ◽  
Author(s):  
Mahdia Benkhoucha ◽  
Nicolas Molnarfi ◽  
Elodie Belnoue ◽  
Madiha Derouazi ◽  
Patrice Lalive
Keyword(s):  
T Cell ◽  

BIO-PROTOCOL ◽  
2014 ◽  
Vol 4 (13) ◽  
Author(s):  
Myoungjoo Kim ◽  
Weiming Ouyang ◽  
Will Liao ◽  
Michael Zhang ◽  
Ming Li
Keyword(s):  
T Cell ◽  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A257-A257
Author(s):  
Thomas Gajewski ◽  
Emily Higgs ◽  
Shuyin Li ◽  
Blake Flood ◽  
Ken Hatogai

BackgroundCheckpoint blockade therapies have transformed the landscape of cancer care. Durable clinical responses have been observed in a subset of patients. However, many patients do not respond, and understanding the mechanisms that determine tumor resistant to checkpoint blockade drugs could potentially benefit more patients. Ferroptosis is a relatively newly described form of regulated cell death distinct from apoptosis and necroptosis. Recently, T cell-promoted tumor ferroptosis was shown to be an anti-tumor mechanism and targeting this pathway could be a potential therapeutic approach.MethodsTo identify genes critical to immunotherapy resistance, B16.SIY cells were transduced with a genome-scale gRNA lentivirus to generate loss of function mutants. In vitro-primed CD8+ T cells isolated from 2C/Rag2–/– TCR transgenic mice specific for the SIY antigen were co-cultured with transduced B16.SIY tumor cells. Resistant mutants were identified by sequencing the gRNAs of survival clones. The gene encoding Decr2, a peroxisomal 2,4-dienoyl-CoA reductase, was identified. To investigate the role of Decr2 in tumor growth and immune responses in vivo, the Decr2 knock-down or Decr2 overexpressed tumors were transplanted into B6 mice and the mice were subsequently treated with anti-PD-L1 antibody. The tumor microenvironments were analyzed by flow cytometry. To understand the resistance mechanism of Decr2 knock-down tumors, RNA-seq was performed and analyzed. The CD8+ T cell mediated tumor ferroptosis in vitro and in vivo was analyzed for lipid reactive oxygen species.ResultsDecr2 mutants were relatively resistant to CD8+ T cell killing in vitro. Consistent with this resistance to CD8+ T cell killing, Decr2 knock-down tumors showed minimal response to anti-PDL1 therapy in vivo. RNA-seq analysis of Decr2 knock-down B16.SIY tumors revealed upregulation of ferroptosis-related genes, including slc7a11. Further mechanistic studies showed that Decr2 knock-down tumors displayed defects in ferroptosis in vitro and in vivo.ConclusionsDecr2-deficient tumors were relatively resistant to CD8+ T cell killing in vitro and anti-PD-L1 immunotherapy in vivo by modulating CD8+ T cell-induced ferroptosis.


2020 ◽  
Vol 8 (2) ◽  
pp. e001037
Author(s):  
Ruiyan Wu ◽  
Caiqin Wang ◽  
Zhiming Li ◽  
Jian Xiao ◽  
Chunyan Li ◽  
...  

BackgroundImmune checkpoint inhibitors (ICIs) induce better tumor regression in melanoma with programmed cell death 1 ligand 1 (PD-L1) high expression, but there has been an upsurge of failed responses. In this study, we aimed to explore the additional mechanisms possibly accounting for ICIs resistance and interventional strategies to overcome the resistance in melanoma with PD-L1 high expression.MethodsMelanoma xenografts and cytotoxicity assays were used to investigate function of SOX2 in regulating antitumor immunity. The activity of the janus kinase-signal transducer and activator of transcriptions (JAK-STAT) pathway was investigated by western blots, quantitative PCR and luciferase assay. Epigenetic compounds library screen was employed to identify inhibitors that could decrease SOX2 level. The effect of histone deacetylase inhibitor SAHA in antitumor immunity alone or in combination with immunotherapy was also determined in vitro and in vivo. Prognostic impact of SOX2 was analyzed using transcriptional profiles and clinical data download from the Gene Expression Omnibus and The Cancer Genome Atlas repository.ResultsWe uncovered a role of SOX2 in attenuating the sensitivity of melanoma cells to CD8+ T-cell killing. Mechanistically, SOX2 inhibited phosphatases suppressor of cytokine signaling 3 (SOCS3) and protein tyrosine phosphatase non-receptor type 1 (PTPN1) transcription, induced duration activation of the JAK-STAT pathway and thereby overexpression of interferon stimulated genes resistance signature (ISG.RS). By targeting the SOX2-JAK-STAT signaling, SAHA promoted the antitumor efficacy of IFNγ or anti-PD-1 in vitro and in vivo. Moreover, SOX2 was an independent prognostic factor for poor survival and resistant to anti-PD-1 therapy in melanoma with PD-L1 high expression.ConclusionsOur data unveiled an additional function of SOX2 causing immune evasion of CD8+ T-cell killing through alleviating the JAK-STAT pathway and ISG.RS expression. We also provided a rationale to explore a novel combination of ICIs with SAHA clinically, especially in melanoma with PD-L1 and SOX2 high expression.


Sign in / Sign up

Export Citation Format

Share Document