Controlling inflammation and cellular damage is the key to preventing and treating multiple organ failure (MOF) following trauma. However, many of the mechanisms that regulate inflammation and cell death in specific organs remain unknown. This means that despite advances in supportive measures for patients with MOF, there have been few advances in MOF treatments, or in our ability to adequately prevent the onset of MOF. Our overarching goal is to ultimately develop new therapeutics for trauma patients based on regulating the inflammatory response and its effects on cellular death and survival pathways following trauma and hemorrhagic shock with resuscitation (HS/R). In this proposal we will continue to investigate novel molecular pathways following HS/R that lead to activation of caspase-11 in end-organs, particularly liver. We will also determine how activation of inflammatory caspases regulates cell death and inflammation, which in turn determines organ cell survival after HS/R. Knowing how these cell-signaling pathways function and interact may guide us toward future therapeutic targets for prevention and treatment of MOF after trauma. Inflammatory caspases include caspases-1, 11 (mice), 4/5 (human), and are associated with inflammatory cell death (pyroptosis) and release of inflammatory cytokines (IL-1?, IL-18). Caspase-11 was recently identified as the intracellular receptor for lipopolysaccharide (LPS) and forms what has been termed the non-canonical inflammasome. LPS-mediated caspase-11 activation in macrophages results in cleavage of gasdermin D (GsdmD) and subsequent release of IL-1?, induction of pyroptosis, and initiation of NLRP3 inflammasome and caspase-1 activation. Caspase-11 can also be activated by endogenous oxidized phospholipid (oxPAPC) in LPS-primed dendritic cells, although this interaction does not result in pyroptosis. Our exciting new data show that caspase-11 is activated during HS/R, a non-infectious/sterile injury model, suggesting a novel mechanism of activation of caspase-11 by endogenous damage associated molecular patterns (DAMPs) without a requirement for LPS. We show that once again mitochondria are integral in activation of inflammatory responses after HS/R, and cardiolipin externalization on damaged and stressed mitochondria can activate caspase-11. Surprisingly activation of caspase-11 in HC after HS/R does not lead to pyroptosis, but is vital for active release of HC HMGB1 in exosomes, and caspase-11 activation is detrimental in HS/R. Our preliminary findings lead us to our main hypothesis that DAMP-induced caspase-11 activation has cell specific functions during HS/R, and is an important regulator of cell death and organ damage. We expect to confirm novel, mitochondrial DAMP-mediated pathways of activation of caspase-11, and novel functions of caspase-11 in liver that may make it an attractive therapeutic target during trauma/HS.

Public Health Relevance

Trauma and shock activate pathways of inflammation that can lead to organ failure and death. The mechanisms of activation of inflammation in different organs and cell-types is not well understood, but knowing how organs respond to shock may improve our future ability to prevent and treat organ failure in trauma patients. Our research investigates effects of activation of an inflammatory protein, caspase-11, which may regulate organ damage and inflammation during shock, and may be a future treatment target that may improve survival in severely injured patients.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
2R01GM102146-06
Application #
9447275
Study Section
Surgery, Anesthesiology and Trauma Study Section (SAT)
Program Officer
Somers, Scott D
Project Start
2013-01-01
Project End
2022-03-31
Budget Start
2018-04-10
Budget End
2019-03-31
Support Year
6
Fiscal Year
2018
Total Cost
Indirect Cost
Name
University of Pittsburgh
Department
Surgery
Type
Schools of Medicine
DUNS #
004514360
City
Pittsburgh
State
PA
Country
United States
Zip Code
15213
Zhou, Hui; Deng, Meihong; Liu, Yingjie et al. (2018) Platelet HMGB1 is required for efficient bacterial clearance in intra-abdominal bacterial sepsis in mice. Blood Adv 2:638-648
Wang, Yifei; Sedlacek, Abigail L; Pawaria, Sudesh et al. (2018) Cutting Edge: The Heat Shock Protein gp96 Activates Inflammasome-Signaling Platforms in APCs. J Immunol 201:2209-2214
Li, Zhi-Gang; Scott, Melanie J; Brzóska, Tomasz et al. (2018) Lung epithelial cell-derived IL-25 negatively regulates LPS-induced exosome release from macrophages. Mil Med Res 5:24
Jiao, Yang; Li, Zhigang; Loughran, Patricia A et al. (2018) Frontline Science: Macrophage-derived exosomes promote neutrophil necroptosis following hemorrhagic shock. J Leukoc Biol 103:175-183
Lei, Zhao; Deng, Meihong; Yi, Zhongjie et al. (2018) cGAS-mediated autophagy protects the liver from ischemia-reperfusion injury independently of STING. Am J Physiol Gastrointest Liver Physiol 314:G655-G667
Lai, Dengming; Tang, Jing; Chen, Linsong et al. (2018) Group 2 innate lymphoid cells protect lung endothelial cells from pyroptosis in sepsis. Cell Death Dis 9:369
Kader, Muhamuda; Alaoui-El-Azher, Mounia; Vorhauer, Jennie et al. (2017) MyD88-dependent inflammasome activation and autophagy inhibition contributes to Ehrlichia-induced liver injury and toxic shock. PLoS Pathog 13:e1006644
Sun, Qian; Fan, Jie; Billiar, Timothy R et al. (2017) Inflammasome and autophagy regulation - a two-way street. Mol Med 23:188-195
Zettel, Kent; Korff, Sebastian; Zamora, Ruben et al. (2017) Toll-Like Receptor 4 on both Myeloid Cells and Dendritic Cells Is Required for Systemic Inflammation and Organ Damage after Hemorrhagic Shock with Tissue Trauma in Mice. Front Immunol 8:1672
Li, Zhigang; Jiao, Yang; Fan, Erica K et al. (2017) Aging-Impaired Filamentous Actin Polymerization Signaling Reduces Alveolar Macrophage Phagocytosis of Bacteria. J Immunol 199:3176-3186

Showing the most recent 10 out of 26 publications