[HTML][HTML] Glutaminolysis mediated by MALT1 protease activity facilitates PD-L1 expression on ABC-DLBCL cells and contributes to their immune evasion

X Xia, W Zhou, C Guo, Z Fu, L Zhu, P Li, Y Xu… - Frontiers in …, 2018 - frontiersin.org
X Xia, W Zhou, C Guo, Z Fu, L Zhu, P Li, Y Xu, L Zheng, H Zhang, C Shan, Y Gao
Frontiers in Oncology, 2018frontiersin.org
Previous studies have demonstrated that programmed death-1 ligand 1 (PD-L1) expressed
in an aggressive activated B-cell (ABC)/non-germinal center B cell–like (GCB) subtype of
diffuse large B-cell lymphoma (DLBCL) is associated with inhibition of the tumor-associated
T cell response. However, the molecular mechanism underlying PD-L1 expression in ABC-
DLBCL remains unclear. Here, we report that MALT1 protease activity is required for ABC-
DLBCL cells to evade cytotoxity of Vγ9Vδ2 T lymphocytes by generating substantial PD-L1+ …
Previous studies have demonstrated that programmed death-1 ligand 1 (PD-L1) expressed in an aggressive activated B-cell (ABC)/non-germinal center B cell–like (GCB) subtype of diffuse large B-cell lymphoma (DLBCL) is associated with inhibition of the tumor-associated T cell response. However, the molecular mechanism underlying PD-L1 expression in ABC-DLBCL remains unclear. Here, we report that MALT1 protease activity is required for ABC-DLBCL cells to evade cytotoxity of Vγ9Vδ2 T lymphocytes by generating substantial PD-L1+ ABC-DLBCL cells. While, NF-κB was dispensable for the PD-L1 expression induced by MALT1 protease activity in ABC-DLBCL cells. Furthermore, we showed that GLS1 expression was profoundly reduced by MALT1 protease activity inhibition, which resulted in insufficiency of glutaminolysis-derived mitochondrial bioenergetics. Activation of the PD-L1 transcription factor STAT3, which was strongly suppressed by glutaminolysis blockade, was rescued in a TCA (tricarboxylic acid) cycle-dependent manner by glutamate addition. Collectively, MALT1 protease activity coupled with glutaminolysis-derived mitochondrial bioenergetics plays an essential role in PD-L1 expression on ABC-DLBCL cells under immunosurveillance stress. Thus, our research sheds light on a mechanism underlying PD-L1 expression and highlights a potential therapeutic target to vanquish immune evasion by ABC-DLBCL cells.
Frontiers