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.

680A AASLD ABSTRACTS HEPATOLOGY, October, 2015<br />

and a decrease in mRNA for markers of inflammation and stellate<br />

cell activation CD-68 and α-SMA, respectively, in a DEN<br />

model of cirrhosis. Furthermore, ND-654 markedly decreased,<br />

in a dose dependent manner, DEN-induced protein expression<br />

of inflammatory cytokines including MCP1, TNFα and LIF, and<br />

fibrotic markers such as TIMP1 and FGF21 and other markers<br />

including Serpin E1, Leptin and Resistin. Conclusions: These<br />

data demonstrate that liver-directed inhibition of ACC can<br />

impact multiple steps in the pathogenesis of fatty liver disease<br />

to NASH and cirrhosis .<br />

Disclosures:<br />

Geraldine Harriman - Employment: Nimbus Discovery<br />

Jeremy R. Greenwood - Employment: Schrodinger Inc.; Patent Held/Filed: Nimbus<br />

Therapeutics; Stock Shareholder: Schrodinger Inc.<br />

Kenneth K. Tanabe - Patent Held/Filed: EGF SNP and risk for HCC, EGFR inhibition<br />

and HCC, gene signature for prognosis in cirrhosis<br />

Bryan C. Fuchs - Consulting: Collagen Medical; Grant/Research Support: Nimbus<br />

Therapeutics<br />

Rosana Kapeller - Employment: Nimbus Therapeutics; Stock Shareholder: Aileron<br />

Therapeutics<br />

The following authors have nothing to disclose: Danielle K. DePeralta, Omeed<br />

Moaven, Lan Wei, Sathesh P. Bhat, William Westlin, H. James Harwood<br />

958<br />

Metformin targets a phosphoSTAT3-miRNAs pathway<br />

to inhibit lipid droplets accumulation and intracellular<br />

inflammation in vitro and in vivo<br />

Natalia Pediconi 1 , Silvia Di Cocco 3 , Silvia Piconese 3 , Federica<br />

Mori 4 , Laura Belloni 2 , Abigail D. Nunn 2 , Tullio Scopigno 2 , Vincenzo<br />

Barnaba 3 , Giovanni Blandino 4 , Sabrina Strano 4 , Massimo<br />

Levrero 3,2 ; 1 Dept of Molecular Medicine, Sapienza University,<br />

Rome, Italy; 2 Center for Life NanoScience (CNLS), IIT-Sapienza,<br />

Rome, Italy; 3 Dept of Internal Medicine - DMISM, Sapienza University,<br />

Rome, Italy; 4 Translational Oncogenomic Unit, Regina Elena<br />

Cancer Institute, Rome, Italy<br />

Background and aim: Accumulation of triglyceride-containing<br />

lipid droplets (LDs) within hepatocytes in NAFLD patients is a<br />

potentially reversible process, although sustained activation of<br />

inflammatory signaling pathways leads to non-alcoholic steatohepatitis<br />

(NASH) that can evolve into cirrhosis and HCC.<br />

The prevalence of NAFLD is increased with aging, however<br />

the molecular mechanism for aging-induced fatty liver remains<br />

poorly understood. Here we investigated the role of a phosphoSTAT3-miRNAs<br />

pathway in LD accumulation and the<br />

activation of intracellular inflammatory pathways in vescicular<br />

steatosis. Methods: DMSO-differentiated human non-transformed<br />

HepaRG cells treated with oleic acid were used as a<br />

cellular model of vescicolar steatosis. C57BL/6 mice treated or<br />

not with metformin for 78 weeks were used as an in vivo model<br />

of aging-induced fatty liver. Results: dHepaRG cells treated<br />

with oleic acid show: a) an accumulation of intracellular lipids<br />

with an increase of both total cellular lipid volume and lipid<br />

droplets number, as evaluated by picosecond CARS (coherent<br />

anti-stoke Raman spettroscopy) mycroscopy; b) the generation<br />

of reactive oxygen species (ROS); c) deregulated lipid metabolism<br />

and liver-specific genes, including PDK4, PLIN4, SLC2A1,<br />

ALB and ALDOB; d) activation of an intracellular inflammatory<br />

response, with the upregulation of NFkB and IFN-a regulated<br />

genes and the activation of the phosphoSTAT3/IL6 pathway.<br />

Treatment of oleate-exposed HepaRG with the S3I STAT3<br />

inhibitor reduces bothe tryglicerides and ROS accumulation<br />

and inhibits phosphoSTAT3 accumulation. We also found that<br />

several STAT3/IL6 responsive miRNAs, including miR21 and<br />

miR24, are upregulated after lipid overload, paralleling STAT3<br />

activation. Chromatin immuno-precipitation (ChIP) experiments<br />

showed that the oleate-dependent transcriptional deregulation<br />

of these miRNAs correlates with phosphoSTAT3 bound to their<br />

promoters. Treatment with S3I inhibits both oleate-dependent<br />

miR21 and miR24 upregulation and phosphoSTAT3 binding to<br />

their promoter. Treatment with metformin, an AMPK activator<br />

widely used as anti-diabetic drug and known to reduce liver<br />

steatosis in vivo, reduces phosphoSTAT3 accumulation, inhibits<br />

miRNAs upregulation and reduces trygliceride accumulation in<br />

fatty HepaRG. Importantly, a chronic 78 weeks treatment with<br />

metformin prevents aging-induced steatosis in C57BL/6 mice<br />

and reduces both liver and serum miR21 levels. Conclusions:<br />

We show that a phosphoSTAT3-miRNAs intracellular inflammatory<br />

pathway amenable to therapeutic targeting by metformin<br />

is activated in response to lipid accumulation in differentiated<br />

hepatocytes in vitro and in vivo.<br />

Disclosures:<br />

Massimo Levrero - Advisory Committees or Review Panels: BMS, Jansen, Gilead,<br />

Tekmira, Galapagos, Medimmune; Speaking and Teaching: MSD, Roche<br />

The following authors have nothing to disclose: Natalia Pediconi, Silvia Di<br />

Cocco, Silvia Piconese, Federica Mori, Laura Belloni, Abigail D. Nunn, Tullio<br />

Scopigno, Vincenzo Barnaba, Giovanni Blandino, Sabrina Strano<br />

959<br />

Exacerbation of non-alcoholic fatty liver disease<br />

(NAFLD) in mice with myeloid cell-specific deletion of<br />

FXR<br />

Rachel McMahan 1 , Cara Porsche 1 , Moshe Levi 2 , Hugo R. Rosen 1 ;<br />

1 Gastroenterology, University of Colorado, Aurora, CO; 2 Renal<br />

Diseases & Hypertension, University of Colorado, Aurora, CO<br />

Background: Nonalcoholic fatty liver disease (NAFLD) is estimated<br />

to affect a third of the U.S. population. The progression<br />

from benign steatosis to steatohepatitis in NAFLD has been<br />

linked to increased recruitment of pro-inflammatory monocytes<br />

and activation of liver macrophages. We have previously<br />

shown that activation of the nuclear bile acid receptor FXR can<br />

improve disease in murine models of NALFD and that in vitro<br />

activation of FXR in macrophages inhibits pro-inflammatory<br />

cytokine production. However, FXR is also expressed in other<br />

cell types including hepatocytes, and the protective effects of<br />

FXR may be through non-macrophage FXR signaling. Therefore,<br />

we aimed to determine the specific role of macrophage<br />

FXR expression in NALFD. Methods: To evaluate to role of<br />

macrophage expression of FXR in NAFLD, we used the Cre-<br />

Lox system to create myeloid-cell specific targeted mutants of<br />

the FXR gene (FXR fl/fl LyzMCre). FXR fl/fl LyzMCre or littermate<br />

controls were fed a western or control diet for 3 months and<br />

NAFLD severity was determined by histological analysis,<br />

hepatic gene expression and flow cytometric analysis of intrahepatic<br />

immune cells isolated by collagenase and mechanical<br />

digestion. Results: Loss of FXR expression in myeloid cells led to<br />

a more severe histological phenotype with increased macrovesicular<br />

steatosis and inflammatory foci compared to littermate<br />

controls. In addition, western diet feeding increased hepatic<br />

expression of pro-inflammatory genes (CCL2, TNF-alpha) and<br />

pro-fibrotic genes (TIMP-1) in FXR fl/fl LyzMCre mice. Myeloid<br />

cell deletion of FXR also led to increased recruitment of intrahepatic<br />

pro-inflammatory monocytes and an increase in IL-17<br />

producing T cells. Conclusions: Myeloid cell-specific deletion of<br />

FXR leads to increased liver inflammation in a murine model of<br />

NAFLD. Targeting FXR in macrophages may be important for<br />

inhibiting hepatic inflammation and NAFLD development.<br />

Disclosures:<br />

Moshe Levi - Grant/Research Support: Intercept, Genzyme-Sanofi, Daiichi Sankyo,<br />

Merck, Novartis<br />

The following authors have nothing to disclose: Rachel McMahan, Cara Porsche,<br />

Hugo R. Rosen

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

Saved successfully!

Ooh no, something went wrong!