02.10.2015 Views

studies

2015SupplementFULLTEXT

2015SupplementFULLTEXT

SHOW MORE
SHOW LESS
  • No tags were found...

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

302A AASLD ABSTRACTS HEPATOLOGY, October, 2015<br />

In contrast, IL-4Rα ∆LysM mice displayed retarded fibrosis resolution<br />

as evidenced by slower clearance of hydroxyproline<br />

and Sirius-Red stained collagen at 2 weeks off CCL 4<br />

. This was<br />

accompanied by a reduction of Ly-6c lo restorative and M2<br />

macrophages and an increase of M1 macrophages. In a therapeutic<br />

approach, we applied an ASO targeting IL-4Rα (IL-4Rα<br />

ASO) to CCL 4<br />

-treated mice. IL-4Rα ASO strongly attenuated<br />

fibrosis progression but mitigated fibrosis resolution after CCL 4<br />

withdrawal. Conclusion: Our <strong>studies</strong> demonstrate that IL-4Rα<br />

modulates fibrosis progression and reversal in discordant ways<br />

through macrophages. During progression, IL-4Rα signaling<br />

increases inflammation and fibrosis by activating Ly6c hi macrophages;<br />

during reversal, IL-4Rα potentiates restorative (M2,<br />

Ly6c lo ) macrophage signaling. Accordingly, IL-4Rα ASO treatment<br />

attenuates fibrosis progression, but retards reversal.<br />

Disclosures:<br />

Jeff R. Crosby - Employment: ISIS Pharmaceuticals<br />

Michael L. McCaleb - Employment: Isis Pharmaceuticals; Stock Shareholder: Isis<br />

Pharmaceuticals<br />

The following authors have nothing to disclose: Shih-Yen Weng, Santosh Vijayan,<br />

Xiaoyu Wang, Yilang Tang, Kornelius Padberg, Yong Ook Kim, Brombacher<br />

Frank, Ari Waisman, Ernesto Bockamp, Detlef Schuppan<br />

184<br />

Liver sinusoidal endothelial cells induce neutrophil<br />

extracellular traps in liver sterile inflammation via<br />

IL-33/ST2 axis<br />

Hai Huang, Hamza Yazdani, Patricia Loughran, Heth R. Turnquist,<br />

Allan Tsung; Department of Suegery, University of Pittsburgh Medical<br />

Center, Pittsburgh, PA<br />

Both liver sinusoidal endothelial cells (LSECs) and neutrophils<br />

are involved and interact in liver ischemia/reperfusion (I/R)<br />

injury. Damaged LSECs, as the major sources of IL-33, play<br />

important role in neutrophil infiltration during liver I/R. We<br />

recently found that in response to damage associated molecular<br />

patterns (DAMPs), infiltrated neutrophils forming neutrophil<br />

extracellular traps (NETs), exacerbate sterile inflammatory<br />

injury during liver I/R. We here sought to determine the role<br />

of IL-33 released from LSECs in NET formation during liver I/R.<br />

WT or IL-33 KO mice were subjected to a non-lethal warm liver<br />

I/R model. Recombinant IL-33 (rIL-33) or soluble ST2 (sST2,<br />

decoy receptor of IL-33) was administered in select groups.<br />

IL-33 KO mice or sST2-treated WT mice were significantly protected<br />

from I/R injury, having significantly less serum sALT and<br />

hepatic necrosis, lower levels of systemic cytokines, abrogation<br />

of proinflammatory signaling pathways compared to WT mice.<br />

rIL-33 administered during I/R exacerbated hepatotoxicity and<br />

systemic inflammation. Although significant neutrophils infiltration<br />

was found in both IL-33 KO and WT mice, NETs formation<br />

decreased in IL-33 KO mice during liver I/R compared<br />

with WT mice, associated with significant less serum level of<br />

myeloperoxidase (MPO)-DNA complexes and tissue level of<br />

citrullinated-histone H3 (NET markers). In addition, treatment<br />

of sST2 reduced NET formation whereas significant increased<br />

NETs were found in rIL-33 treated mice after liver I/R. In vitro,<br />

IL-33 released from LSECs that were subjected to hypoxia (1%<br />

O 2<br />

) promotes NET formation. Directly using rIL-33 stimulates<br />

neutrophils confirmed IL-33/ST2-MyD88 signaling pathway in<br />

NET formation. Using either IL-33 KO LSECs, or co-stimulated<br />

neutrophil with IL-33 neutralizing antibody or sST2 blocked<br />

NET formation. Our study demonstrates IL-33 from LSECs initiates<br />

a feed-forward mechanism to neutrophils inducing NETs<br />

formation in excessive inflammation and hepatotoxicity during<br />

liver I/R.<br />

Disclosures:<br />

The following authors have nothing to disclose: Hai Huang, Hamza Yazdani,<br />

Patricia Loughran, Heth R. Turnquist, Allan Tsung<br />

185<br />

15-PGDH prevents LPS/GalN-induced acute liver injury<br />

through inhibiting Kupffer cell activation<br />

Lu Yao, Chang Han, Tong Wu; pathology and laboratory medicine,<br />

tulane university, New Orleans, USA Minor Outlying Islands<br />

The NAD+-dependent 15-hydroxyprostaglandin dehydrogenase<br />

(15-PGDH) catalyzes the oxidation of the 15(S)-hydroxyl<br />

group of Prostaglandin E 2<br />

(PGE 2<br />

), converting the pro-inflammatory<br />

PGE 2<br />

to the anti-inflammatory 15-keto-PGE 2<br />

(an<br />

endogenous ligand for peroxisome proliferator-activated receptor-gamma<br />

[PPAR-γ]). PPAR-γ is a ligand-dependent transcriptional<br />

factor, which plays an important role in regulation of<br />

inflammatory cell activation. To evaluate the significance of<br />

15-PGDH/PPAR-γ cascade in liver inflammation, we generated<br />

transgenic mice with targeted expression of 15-PGDH in the<br />

liver (15-PGDH Tg) and the animals were subjected to lipopolysaccharide<br />

(LPS)/Galactosamine (GalN) induced acute liver<br />

inflammation and tissue damage. Compared to the wild type<br />

mice, the 15-PGDH Tg mice showed lower levels of alanine<br />

aminotransferase (ALT) and aspartate aminotransferase (AST),<br />

less liver tissue damage, less hepatic apoptosis/necrosis, less<br />

macrophage activation, and lower inflammatory cytokine production.<br />

In cultured Kupffer cells, treatment with 15-keto-PGE 2<br />

or the conditioned medium (CM) from 15-PGDH Tg hepatocyes<br />

inhibited LPS-induced cytokine production. Both 15-keto-PGE 2<br />

and the CM from15-PGDH Tg hepatocyes also significantly<br />

up-regulated the expression of PPAR-γ down-stream genes in<br />

Kupffer cells. On the other hand, 15-PGDH overexpression<br />

or 15-keto-PGE 2<br />

treatment of hepatocytes did not influence<br />

TNF-α-induced hepatocyte apoptosis. These results suggest<br />

that the resistance of 15-PGDH Tg mice to LPS/GalN-induced<br />

liver injury is mediated through 15-keto-PGE 2<br />

, which activates<br />

PPAR-γ in Kupffer cells and inhibited inflammatory cytokine<br />

production. Consistent with this assertion, we observed that the<br />

PPAR-γ antagonist, GW9662, reversed the effect of 15-keto-<br />

PGE 2<br />

in Kupffer cell (in vitro) and restored the susceptibility<br />

of 15-PGDH Tg mice to LPS/GalN-induced acute liver injury<br />

(in vivo). Taken together, our findings provide novel evidence<br />

that hepatic overexpression of 15-PGDH protects against<br />

LPS/GalN-induced acute liver injury by inhibiting Kupffer cell<br />

inflammatory response. Thus, 15-keto-PGE 2<br />

is an endogenous<br />

PPAR-γ ligand that may be utilized as a pharmacological agent<br />

to ameliorate liver inflammation and tissue damage.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!