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.

266A AASLD ABSTRACTS HEPATOLOGY, October, 2015<br />

ASH. Inhibition of peroxisome proliferator-activated receptor<br />

g (PPARg) or cyclic-AMP-responsive-element binding protein H<br />

(CREBH) attenuated chronic-plus-binge ethanol-induced elevation<br />

of Fsp27a and FSP27b mRNA, respectively, and subsequently<br />

ameliorated liver injury. Overexpression of Fsp27 and<br />

ethanol exposure synergistically induced mitochondrial reactive<br />

oxygen species production and hepatocyte injury in vivo and<br />

in vitro, which is likely owing to the mitochondrial location of<br />

FSP27 leading to the decreased mitochondrial complex I activity.<br />

Finally, hepatic expression of CIDEC mRNA was elevated<br />

and positively correlated with hepatic steatosis, disease severity,<br />

and mortality in AH patients. Conclusion: FSP27/CIDEC<br />

gene product promotes ASH in chronic-plus-binge ethanol-fed<br />

mice and in human AH. Targeting CIDEC gene is likely to be a<br />

novel therapeutic targets for the treatment of ASH.<br />

overgrowth was similar to WT mice, alcohol-fed Reg3b and<br />

Reg3g deficient mice showed a significantly higher number of<br />

mucosa-associated bacteria in the small intestine as compared<br />

with their respective WT littermates. This was accompanied<br />

by a significantly increased number of bacteria translocating<br />

through intestinal epithelial cells and more positive mesenteric<br />

lymph node cultures in alcohol-fed Reg3b -/- and Reg3g -/- mice.<br />

Interestingly, absence of Reg3b or Reg3g did not affect the<br />

intestinal paracellular barrier function as determined by systemic<br />

LPS level and fecal albumin. Most importantly, Reg3b<br />

and Reg3g deficient mice showed more alcoholic liver disease<br />

as assessed by higher plasma ALT levels, increased hepatic<br />

triglycerides and inflammation. To corroborate our data, we<br />

generated transgenic mice overexpressing Reg3g under the<br />

control of the intestine specific villin promoter. Consistent with<br />

our results, Reg3g transgenic mice showed significantly less<br />

mucosa-associated bacteria than WT littermates after alcohol<br />

feeding. Overexpression of intestinal Reg3g suppressed<br />

translocation of bacteria through intestinal epithelial cells to<br />

mesenteric lymph nodes, while paracellular permeability and<br />

systemic LPS levels were not changed after alcohol feeding.<br />

Reg3g transgenic mice were protected from alcohol-induced<br />

liver injury, steatosis and inflammation. Conclusion: Intestinal<br />

Reg3b and Reg3g prevent bacterial colonization of epithelial<br />

surfaces, which reduces translocation of viable bacteria and<br />

prevents alcoholic liver disease. Reg3 proteins are novel and<br />

promising targets in the treatment of alcohol-induced liver disease.<br />

Disclosures:<br />

The following authors have nothing to disclose: Lirui Wang, Peng Chen, Lora V.<br />

Hooper, Bernd Schnabl<br />

Disclosures:<br />

Ramon Bataller - Advisory Committees or Review Panels: Sandhill; Consulting:<br />

VTI, Oncozyme Pharma<br />

The following authors have nothing to disclose: Ming-Jiang Xu, Yan Cai, Hua<br />

Wang, José T. Altamirano, Gemma Odena, Frank J. Gonzalez, Bin Gao<br />

111<br />

Antimicrobial proteins Reg3b and Reg3g protect mice<br />

from alcoholic liver disease by preventing bacterial<br />

translocation<br />

Lirui Wang 1 , Peng Chen 1 , Lora V. Hooper 2 , Bernd Schnabl 1 ;<br />

1 University of California San Diego, La Jolla, CA; 2 Department of<br />

Immunology, The University of Texas Southwestern Medical Center,<br />

Dallas, TX<br />

Background: Antimicrobial proteins are secreted by intestinal<br />

epithelial cells and Paneth cells. They represent the first<br />

line of defense against pathogens and maintain homeostasis<br />

with commensal bacteria. We have previously shown that the<br />

expression of the C-type lectin regenerating islet derived-3<br />

(Reg3) is suppressed in the small intestine after chronic alcohol<br />

use in mice and humans, yet functional consequences of<br />

lower Reg3 expression on the intestinal microbiota, bacterial<br />

translocation and alcoholic liver disease are unknown. The<br />

aim of our study was to investigate the role of Reg3b and<br />

Reg3g in chronic alcoholic liver disease. Methods and Results:<br />

A Lieber-DeCarli model was used to induce intestinal dysbiosis,<br />

bacterial translocation and liver disease in mice. After alcohol<br />

feeding for 8 weeks, wild-type (WT) littermate mice showed<br />

bacterial overgrowth of the luminal and the mucosa-associated<br />

microbiota in the small intestine. While luminal bacterial<br />

112<br />

Hepatocytes from mice on intragastric feeding model of<br />

alcoholic steatohepatitis release extracellular vesicles<br />

with specific microRNA cargo that modulate hepatic<br />

stellate cell and macrophage phenotype<br />

Akiko Eguchi 1 , Jihoon Kim 1 , Lucila Ohno-Machado 1 , Hidekazu<br />

Tsukamoto 2 , Ariel E. Feldstein 1 ; 1 UCSD, La Jolla, CA; 2 USC, Los<br />

Angeles, CA<br />

Liver inflammation and fibrosis are key histological features<br />

associated with prognosis in patients with alcoholic steatohepatitis<br />

(ASH). Extracellular vesicles (EVs) are released during cell<br />

stress or demise, can contain a barcode of the cell of origin<br />

including specific microRNAs and are growingly recognized<br />

as key cell-to-cell communicators. Here we tested the hypothesis<br />

that during ASH development, hepatocyte damage release EVs<br />

with a microRNA signature that can fuse with hepatic stellate<br />

cells (HSC) and Kupffer cells (KC) to regulate their phenotype.<br />

Methods: C57/B6 mice were placed on intragastric feeding<br />

model of continuous ethanol infusion or control diet for 4 weeks<br />

to reproduce a physiologically relevant model of ASH. The<br />

extent of steatosis, inflammation, and fibrosis was assessed<br />

by histological and molecular analyses on liver specimens.<br />

Isolated hepatocytes (HC) or KC from ASH or control mice<br />

were incubated in medium-EV free serum and HC- or KC-derived<br />

EVs were isolated by ultracentrifugation from culture<br />

medium. A complete characterization of EVs was performed<br />

by FACS, electron microscopy, dynamic light scattering. HC-EV<br />

biological function was investigated in isolated primary HSCs<br />

or KCs derived from C57/B6 mice by cell morphology or<br />

qPCR. Comprehensive encapsulated miRNA in HC-EVs were<br />

assessed via transcriptome using illumina miRNA-seq. Results:<br />

We observed highly significant differences in the levels of HCor<br />

KC-EVs between control group and ASH (1.3x10 6 HC-EV/

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

Saved successfully!

Ooh no, something went wrong!