Despite recent advances in antibiotic therapy and intensive care, sepsis remains the most common cause of death in the intensive care unit, annually claiming >225,000 victims in the U.S. alone. The pathogenesis of sepsis remains obscure, but is partly attributable to dys-regulated inflammation propagated by ?early? cytokines (e.g., TNF and IL-1?), but sustained by ?intermediate? (e.g., SAA) and ?late? (e.g., HMGB1) mediators. We recently discovered that LPS and SAA upregulated the expression of connexin 43 (Cx43) and/or pannexin 1 (Panx1) hemichannels to facilitate ATP-dependent PKR activation and HMGB1 release, but did not know whether LPS and SAA also induced procathepsin L (pCTS-L) secretion to mediate lethal bacterial infections (LBI). Our preliminary data indicated that LPS and SAA induced a marked expression and secretion of pCTS- L in both murine macrophage and human monocyte cultures. Consequently, pCTS-L was not detectable in the circulation of healthy animals or human subjects, but significantly elevated in the blood of septic animals and patients. Highly purified recombinant pCTS-L stimulated primary human monocytes to release various chemokines, as well as pro- (e.g., TNF and IL-1?) and anti-inflammatory cytokines (e.g., IL-10) in vitro, and exacerbated endotoxemic lethality in vivo. In contrast, pCTS-L-neutralizing antibodies significantly rescued mice from lethal sepsis, suggesting pCTS-L as another late mediator of LBI. Meanwhile, a semi-high throughput screening of a NatProduct Collection of 800 natural products and a US Drug Collection of 1360 bioactive compounds led to the finding of a few lead compounds [including lanosterol (LAN) and progesterone (PRO)] with striking structural resemblance and similar pCTS-L-inhibiting activities. Although LAN is an abundant secondary metabolite in some medicinal plants including wolfberry and aveloz, it also serves as a substrate for the synthesis of PRO in animals and humans. These exciting findings prompted the current proposal to investigate a novel role of pCTS-L in LBI, as well as intricate mechanisms by which a natural product, LAN, and its derivative, PRO, inhibit pCTS-L-induced inflammation. The experiments outlined in Aim 1 will test the hypothesis that pCTS-L systemically accumulates in the circulation of septic patients and correlates with other surrogate markers of sepsis.
In Aim 2, we will test the hypothesis that alterations of pCTS-L levels (by supplementation of pCTS-L or genetic knockout) or activities (by use of neutralizing IgGs or natural inhibitor LAN or PRO) divergently influence the outcomes of LBI. The experiments outlined in Aim 3 will test the hypothesis that LAN and/or PRO inhibit the pCTS-L-induced inflammation through impairing the TLR4/ RAGE-dependent hemichannel and PKR activation in vitro, and confer protection against LBI partly by attenuating systemic inflammation and associated dysregulated coagulation in vivo. Collectively, this project will improve our understanding of the role of pCTS-L in LBI, and shed light on the intricate mechanism underlying natural product LAN and PRO-mediated protection against lethal bacterial infections.

Public Health Relevance

The pathogenesis of sepsis remains obscure, but is partly attributable to dys-regulated systemic inflammation propagated by ?early? cytokines (e.g., TNF and IL-1?), but sustained by ?late? mediators such as HMGB1. The elucidation of procathepsin L (pCTS-L) as another possible ?late? mediator of lethal bacterial infections, and the intricate mechanism underlying a medicinal herbal product (lanosterol) and its animal derivative (progesterone)-mediated inhibition of pCTS-L-induced inflammation will significantly improve our understanding of the mechanisms underlying the pathogenesis of sepsis, as well as the protective effects of alternative natural therapies.

Agency
National Institute of Health (NIH)
Institute
National Center for Complementary & Alternative Medicine (NCCAM)
Type
Research Project (R01)
Project #
2R01AT005076-09A1
Application #
10058673
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Wang, Yisong
Project Start
2010-09-01
Project End
2024-08-31
Budget Start
2020-09-01
Budget End
2021-08-31
Support Year
9
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Feinstein Institute for Medical Research
Department
Type
DUNS #
110565913
City
Manhasset
State
NY
Country
United States
Zip Code
11030
Xiao, Hui-Wen; Ge, Chang; Feng, Guo-Xing et al. (2018) Gut microbiota modulates alcohol withdrawal-induced anxiety in mice. Toxicol Lett 287:23-30
Zhao, Xiaoling; Li, Renjia; Jin, Hui et al. (2018) Epigallocatechin-3-gallate confers protection against corticosterone-induced neuron injuries via restoring extracellular signal-regulated kinase 1/2 and phosphatidylinositol-3 kinase/protein kinase B signaling pathways. PLoS One 13:e0192083
Li, Wei; Bao, Guoqiang; Chen, Weiqiang et al. (2018) Connexin 43 Hemichannel as a Novel Mediator of Sterile and Infectious Inflammatory Diseases. Sci Rep 8:166
Xiao, Hui-Wen; Li, Yuan; Luo, Dan et al. (2018) Hydrogen-water ameliorates radiation-induced gastrointestinal toxicity via MyD88's effects on the gut microbiota. Exp Mol Med 50:e433
Deng, Wenjun; Zhu, Shan; Zeng, Ling et al. (2018) The Circadian Clock Controls Immune Checkpoint Pathway in Sepsis. Cell Rep 24:366-378
Lin, Nan; Shay, Jessica E S; Xie, Hong et al. (2018) Myeloid Cell Hypoxia-Inducible Factors Promote Resolution of Inflammation in Experimental Colitis. Front Immunol 9:2565
Lan, Xiqian; Wen, Hongxiu; Aslam, Rukhsana et al. (2018) Nicotine enhances mesangial cell proliferation and fibronectin production in high glucose milieu via activation of Wnt/?-catenin pathway. Biosci Rep 38:
Kang, Rui; Zeng, Ling; Zhu, Shan et al. (2018) Lipid Peroxidation Drives Gasdermin D-Mediated Pyroptosis in Lethal Polymicrobial Sepsis. Cell Host Microbe 24:97-108.e4
Chen, Ruochan; Zhu, Shan; Fan, Xue-Gong et al. (2018) High mobility group protein B1 controls liver cancer initiation through yes-associated protein -dependent aerobic glycolysis. Hepatology 67:1823-1841
Wen, Hongxiu; Kumar, Vinod; Lan, Xiqian et al. (2018) APOL1 risk variants cause podocytes injury through enhancing endoplasmic reticulum stress. Biosci Rep 38:

Showing the most recent 10 out of 50 publications