TY - JOUR A1 - Knethen, Andreas von A1 - Schäfer, Anne A1 - Kuchler, Laura A1 - Knape, Tilo A1 - Christen, Urs A1 - Hintermann, Edith A1 - Fißlthaler, Beate A1 - Schröder, Katrin A1 - Brandes, Ralf A1 - Genz, Berit A1 - Abshagen, Kerstin A1 - Pützer, Brigitte M. A1 - Sha, Lisa K. A1 - Weigert, Andreas A1 - Syed, Shahzad Nawaz A1 - Schulz, Martin A1 - Shah, Ajay M. A1 - Erns, Andreas A1 - Putyrski, Mateusz A1 - Finkelmeier, Fabian A1 - Pešić, Marina A1 - Greten, Florian A1 - Hogardt, Michael A1 - Kempf, Volkhard A. J. A1 - Gunne, Sandra A1 - Parnham, Michael J. A1 - Brüne, Bernhard T1 - Tolerizing CTL by sustained hepatic PD-L1 expression provides a new therapy spproach in mouse sepsis T2 - Theranostics N2 - Cytotoxic T lymphocyte (CTL) activation contributes to liver damage during sepsis, but the mechanisms involved are largely unknown. Understanding the underlying principle will permit interference with CTL activation and thus, provide a new therapeutic option. Methods: To elucidate the mechanism leading to CTL activation we used the Hepa1-6 cell line in vitro and the mouse model of in vivo polymicrobial sepsis, following cecal-ligation and -puncture (CLP) in wildtype, myeloid specific NOX-2, global NOX2 and NOX4 knockout mice, and their survival as a final readout. In this in vivo setting, we also determined hepatic mRNA and protein expression as well as clinical parameters of liver damage - aspartate- and alanine amino-transaminases. Hepatocyte specific overexpression of PD-L1 was achieved in vivo by adenoviral infection and transposon-based gene transfer using hydrodynamic injection. Results: We observed downregulation of PD-L1 on hepatocytes in the murine sepsis model. Adenoviral and transposon-based gene transfer to restore PD-L1 expression, significantly improved survival and reduced the release of liver damage, as PD-L1 is a co-receptor that negatively regulates T cell function. Similar protection was observed during pharmacological intervention using recombinant PD-L1-Fc. N-acetylcysteine blocked the downregulation of PD-L1 suggesting the involvement of reactive oxygen species. This was confirmed in vivo, as we observed significant upregulation of PD-L1 expression in NOX4 knockout mice, following sham operation, whereas its expression in global as well as myeloid lineage NOX2 knockout mice was comparable to that in the wild type animals. PD-L1 expression remained high following CLP only in total NOX2 knockouts, resulting in significantly reduced release of liver damage markers. Conclusion: These results suggest that, contrary to common assumption, maintaining PD-L1 expression on hepatocytes improves liver damage and survival of mice during sepsis. We conclude that administering recombinant PD-L1 or inhibiting NOX2 activity might offer a new therapeutic option in sepsis. KW - sepsis KW - cytotoxic T cells KW - reactive oxygen species KW - PD-L1 KW - liver Y1 - 2019 UR - http://publikationen.ub.uni-frankfurt.de/frontdoor/index/index/docId/49948 UR - https://nbn-resolving.org/urn:nbn:de:hebis:30:3-499486 N1 - 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. VL - 9 SP - 2003 EP - 2016 ER -