02.10.2015 Views

studies

2015SupplementFULLTEXT

2015SupplementFULLTEXT

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

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

706A AASLD ABSTRACTS HEPATOLOGY, October, 2015<br />

Gastroenterology, 2014) shedding light on new pathways of<br />

IFN. It is now clear that JAK/STAT and the hundreds of Interferon<br />

Stimulated Genes (ISGs) it stimulates is not the only pathway<br />

mediating IFN’s antiviral effects in the liver. One of the<br />

genes we identified is IQ-motif containing GTPase activating<br />

protein 2 (IQGAP2). IQGAP2 is predominantly expressed in<br />

the liver and has been described as a tumor suppressor. Here<br />

we show that IQGAP2 also mediates IFN anti-HCV response<br />

in infected hepatoma cells. Indeed, IQGAP2 siRNA-mediated<br />

knock-down rescues HCV infection from IFN inhibition. We<br />

show that IQGAP2 is not itself an ISG and its knockdown<br />

does not affect JAK/STAT activation, suggesting that IQGAP2<br />

acts independently of the JAK/STAT pathway. We found that<br />

IQGAP2 interacts with RelA/p65, a subunit of NF-κB transcription<br />

factor. Interestingly, our data suggest that IFN alone<br />

is sufficient to activate RelA and stimulate NF-κB-dependent<br />

transcription in hepatoma cells; this process requires IQGAP2<br />

expression. Furthermore, we found that RelA silencing mimics<br />

IQGAP2 knock-down effects in rescuing HCV from IFN inhibition<br />

in IFN-deficient cells. This suggests that IQGAP2 and RelA<br />

mediate IFN anti-HCV response downstream of IFN binding to<br />

its receptor, a mechanism different from the well characterized<br />

role of NF-κB in IFN production. Finally, we investigated the<br />

effect of IQGAP2 or RelA knock-down on the induction by<br />

IFN of 23 ISGs with previously described anti-HCV properties.<br />

We found that IQGAP2 and RelA are both required for the<br />

full induction of two thirds of the tested genes suggesting that<br />

IQGAP2 and RelA mediate IFN anti-HCV response by controlling<br />

ISG induction in parallel of the JAK/STAT pathway.<br />

Altogether, our data demonstrate a previously unrecognized<br />

function for IQGAP2 in regulating innate antiviral response<br />

against HCV, and probably other hepatotropic pathogens.<br />

Since IQGAP2 is predominantly expressed in the liver, understanding<br />

the mechanism of RelA activation by IQGAP2 may<br />

open novel opportunities to prevent NF-kB associated liver disease.<br />

Disclosures:<br />

Stephane Chevaliez - Advisory Committees or Review Panels: Janssen; Speaking<br />

and Teaching: Gilead, BMS<br />

Dahlene Fusco - Grant/Research Support: Gilead<br />

Raymond T. Chung - Grant/Research Support: Gilead, Mass Biologics, Abbvie,<br />

Merck, BMS<br />

The following authors have nothing to disclose: Cynthia Brisac, Shadi Salloum,<br />

Victor Yang, Esperance A. Schaefer, Jian Hong, Charles Carlton-Smith, Nadia<br />

Alatrakchi, Anna Lidofsky, Xiao Liu, Dachuan Cai, Lee F. Peng, Wenyu Lin<br />

1016<br />

Single-strand RNA-induced monocyte differentiation<br />

generates pro-fibrogenic M2 macrophages in chronic<br />

hepatitis C Virus infection<br />

Banishree Saha, Karen Kodys, Gyongyi Szabo; Medicine, UMASS<br />

Med School, Worcester, MA<br />

Purpose: Innate immune responses, including monocyte and<br />

macrophage activation, contribute to the pathogenesis of<br />

chronic Hepatitis C Virus (HCV) infection and liver fibrosis.<br />

However the mechanisms that lead to liver fibrosis during<br />

HCV infection are not well understood. In response to the<br />

tissue microenvironment, monocytes undergo differentiation<br />

into polarized states including M1 (pro-inflammatory) or M2<br />

(anti-inflammatory/pro-fibrotic) macrophages. We hypothesized<br />

that during chronic HCV infection monocytes undergo<br />

differentiation that affect liver disease progression. Methods:<br />

Healthy human monocytes (n=10-15) were cultured with cellfree<br />

HCV or exosomes isolated from HCV-infected Huh7.5<br />

hepatoma cells for 5-7 days. Circulating monocytes and liver<br />

macrophages from HCV-infected patients and controls were<br />

analyzed by flow cytometry and western blotting. Results:<br />

Co-culture of healthy monocytes with cell-free HCV or exosomes<br />

derived from HCV-infected Huh7.5 cells resulted in differentiation<br />

of monocytes into macrophages (MΦ) with high CD14<br />

and CD68 expression. Exosomes from HCV-infected Huh7.5<br />

cells contained HCV single stranded (ss) RNA. These MΦ displayed<br />

M2 markers (CD206, CD163 and DC-SIGN) and produced<br />

pro- and anti-inflammatory cytokines. The HCV-induced<br />

M2-MΦ led to hepatic stellate cell (LX2) activation indicated<br />

by increased expression of collagen, TIMP-1 and α-SMA and<br />

this was prevented by anti-TGFβ blocking antibody administration.<br />

The HCV-induced monocyte activation and differentiation<br />

into M2-MΦ was prevented by TLR8 but not TLR3 or<br />

TLR7 knock-down. TLR8 ligands, independent of HCV, caused<br />

monocyte differentiation and M2-MΦ polarization suggesting<br />

a role for TLR8 in this process. Furthermore, HCV ssRNA alone<br />

induced monocyte to M2-MΦ differentiation and polarization,<br />

demonstrating that HCV ssRNA signals via TLR8 induce monocyte<br />

differentiation and polarization. In vivo in patients with<br />

chronic HCV infection, we observed a significant increase in<br />

the expression of M2 markers (CD206, CD163) on circulating<br />

monocytes and in the liver. Furthermore, we found a significant<br />

increase in collagen + CD206 + CD14 + circulating monocytes<br />

and the increased frequency of collagen + CD206 + CD14 + circulating<br />

monocytes correlated with liver fibrosis in HCV-infected<br />

patients. Conclusion: Our data identify a new mechanism by<br />

which cell-free and exosome-packaged HCV ssRNA interacts<br />

with monocytes and induces TLR8-mediated differentiation to a<br />

pro-fibrogenic, M2-MΦ. These M2-MΦ promote liver fibrosis.<br />

We also identified M2-marker plus collagen-expressing circulating<br />

monocytes as potential biomarkers of fibrosis in chronic<br />

HCV infection.<br />

Disclosures:<br />

The following authors have nothing to disclose: Banishree Saha, Karen Kodys,<br />

Gyongyi Szabo<br />

1017<br />

Elucidating Mechanisms Underlying Development of<br />

Liver Disease in HIV/HCV Coinfection<br />

Jinhee Hyun 3 , Masato Yoneda 1 , Eugene R. Schiff 1 , Emmanuel<br />

Thomas 1,2 ; 1 Schiff Center for Liver Diseases, University of Miami<br />

Miller School of Medicine, Miami, FL; 2 Sylvester Comprehensive<br />

Cancer Center, Miami, FL; 3 Cell Biology, University of Miami<br />

School of Medicine, Miami, FL<br />

BACKGROUND: Patients with HIV that are coinfected with<br />

HCV are at increased risk for rapidly progressive liver disease<br />

and subsequently the development of Hepatocellular Carcinoma<br />

(HCC). Specifically, HCC develops earlier in coinfected<br />

patients and these patients are more symptomatic than those<br />

with only HCV infection at diagnosis suggesting that both viruses<br />

increase the propensity for malignant transformation. However,<br />

the genetic and cellular based mechanisms underpinning how<br />

HCV initiates and subsequently induces liver pathology and<br />

why coinfection with HIV results in significantly worse hepatic<br />

disease remains to be clarified. In addition, the specific cell<br />

types that contribute to these clinical outcomes are unknown.<br />

In this study, we specifically are testing the hypothesis that the<br />

robust viral RNA dependent innate immune response that we<br />

have characterized, that drives inflammation in HCV infected<br />

livers, is augmented with HIV coinfection through activation<br />

of viral DNA and RNA sensing pathways. METHODS: To this<br />

end, we have developed novel in-vitro models that utilize HCV<br />

infected primary human hepatocytes (PHHs) and HIV infected<br />

primary kupffer cells (PKCs). Specifically, the JFH-1 strain was<br />

used to study HCV while the BAL (R5) and IIIB (X4) strains were

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

Saved successfully!

Ooh no, something went wrong!