[HTML][HTML] The HMGB1/RAGE axis triggers neutrophil-mediated injury amplification following necrosis

P Huebener, JP Pradere, C Hernandez… - The Journal of …, 2019 - Am Soc Clin Investig
P Huebener, JP Pradere, C Hernandez, GY Gwak, JM Caviglia, X Mu, JD Loike…
The Journal of clinical investigation, 2019Am Soc Clin Investig
In contrast to microbially triggered inflammation, mechanisms promoting sterile inflammation
remain poorly understood. Damage-associated molecular patterns (DAMPs) are considered
key inducers of sterile inflammation following cell death, but the relative contribution of
specific DAMPs, including high–mobility group box 1 (HMGB1), is ill defined. Due to the
postnatal lethality of Hmgb1-knockout mice, the role of HMGB1 in sterile inflammation and
disease processes in vivo remains controversial. Here, using conditional ablation strategies …
In contrast to microbially triggered inflammation, mechanisms promoting sterile inflammation remain poorly understood. Damage-associated molecular patterns (DAMPs) are considered key inducers of sterile inflammation following cell death, but the relative contribution of specific DAMPs, including high–mobility group box 1 (HMGB1), is ill defined. Due to the postnatal lethality of Hmgb1-knockout mice, the role of HMGB1 in sterile inflammation and disease processes in vivo remains controversial. Here, using conditional ablation strategies, we have demonstrated that epithelial, but not bone marrow–derived, HMGB1 is required for sterile inflammation following injury. Epithelial HMGB1, through its receptor RAGE, triggered recruitment of neutrophils, but not macrophages, toward necrosis. In clinically relevant models of necrosis, HMGB1/RAGE-induced neutrophil recruitment mediated subsequent amplification of injury, depending on the presence of neutrophil elastase. Notably, hepatocyte-specific HMGB1 ablation resulted in 100% survival following lethal acetaminophen intoxication. In contrast to necrosis, HMGB1 ablation did not alter inflammation or mortality in response to TNF- or FAS-mediated apoptosis. In LPS-induced shock, in which HMGB1 was considered a key mediator, HMGB1 ablation did not ameliorate inflammation or lethality, despite efficient reduction of HMGB1 serum levels. Our study establishes HMGB1 as a bona fide and targetable DAMP that selectively triggers a neutrophil-mediated injury amplification loop in the setting of necrosis.
The Journal of Clinical Investigation