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.

HEPATOLOGY, VOLUME 62, NUMBER 1 (SUPPL) AASLD ABSTRACTS 1031A<br />

We have shown previously that viral quasispecies evolution<br />

and diversity may play a pathogenic role in development of<br />

HBeAg seroconversion. However, the role of viral quasispecies<br />

in pathophysiology of reactivation in HBeAg-negative chronic<br />

hepatitis B is uncertain. Methods: Twenty-two HBeAg-negative<br />

chronic hepatitis B patients who had serial serum samples<br />

collected before and after HBV reactivation were analysed.<br />

Full length HBV genomes were amplified from serum extracted<br />

DNA and were cloned into pCR®2.1-TOPO vector. At least<br />

20 positive clones were randomly selected and sequenced<br />

using Big Dye Terminator. The samples were analyzed for point<br />

mutation in precore stop codon, core promoter- Enhancer II, S1<br />

promoter, S2 promoter and Enhancer I- X promoter regions.<br />

Ten inactive HBV carriers were also analysed as controls. Fulllength<br />

HBV genome collected before and after reactivation<br />

were transfected into Huh7 cells for functional analysis of their<br />

replication fitness. Results: In subjects with clear clinical reactivation<br />

profile, clonal selection was demonstrated (9 out of<br />

10 subjects) by DNA phylogenetic analysis (p=0.005). The<br />

genetic diversity and viral evolution rates were not statistically<br />

different from the inactive controls except except at the precore/core<br />

ORF (p=0.035, Mann-Whitney U test). 80% of reactivation<br />

patients had precore stop codon G to A substitution at<br />

nucleotide position 1896. In addition, other mutations in the<br />

promoter/enhancer regions were identified with more than fifty<br />

percent difference between time point 1 and time point 2 (and<br />

not found in control subjects). HBV clones isolated during HBV<br />

reactivation showed higher HBV DNA and HBsAg production<br />

rates than the clones isolated during inactive phase of the same<br />

patient in transfected Huh7 cell line. Conclusions: Significant<br />

clonal selection occurred during HBeAg-negative reactivation.<br />

Precore stop codon substitution G1896A occurs significantly<br />

in majority of the patients. The nucleotide variability in the precore<br />

promoter, and possibly other promoter/enhancer regions<br />

may play an important role in the progression of HBV disease,<br />

although more functional analyses of individual mutations are<br />

required.<br />

Disclosures:<br />

Seng Gee Lim - Advisory Committees or Review Panels: Bristol-Myers Squibb,<br />

Novartis Pharmaceuticals, Merck Sharp and Dohme Pharmaceuticals, Gilead<br />

Pharmaceuticals, Roche Pharmaceuticals; Speaking and Teaching: Bristol-Myers<br />

Squibb, Novartis Pharmaceuticals, Merck Sharp and Dohme Pharmaceuticals,<br />

Gilead Pharmaceuticals<br />

The following authors have nothing to disclose: Guan Huei Lee, Hui Heng Chong<br />

1687<br />

Hepatic macrophage IL-1 beta expression and bile and<br />

sterol transporter suppression precede cholestasis in a<br />

parenteral nutrition mouse model<br />

Karim C. El Kasmi, Aimee Anderson, Michael W. Devereaux,<br />

Ronald J. Sokol; Pediatrics, UC Denver, Aurora, CO<br />

Background: Parenteral Nutrition Associated Cholestasis<br />

(PNAC) is a serious complication of long term PN infusion in<br />

children and adults with intestinal failure. We have previously<br />

reported in a PNAC mouse model that activation of hepatic<br />

macrophages and up-regulation of pro-inflammatory cytokines<br />

(IL-1 beta) was associated with suppression of hepatic bile<br />

(Abcb11/BSEP, Abcc2/MRP2) and sterol (ABCG5/8/sterolin)<br />

transporters. IL-1R-/- mice were protected from PNAC, with<br />

normalization of hepatic transporter expression, supporting<br />

a causal role for IL-1 beta. The objective of this study was<br />

to determine the role of IL-1 beta at early stages of PNAC<br />

by examining the temporal relationship at early time points<br />

between IL-1 beta, transporter expression, and cholestasis.<br />

Methods and Results: PNAC model: wild type C57/B6 mice<br />

were exposed to dextran sulfate sodium (DSS) in drinking<br />

water (to induce intestinal injury) for 4 days followed by infusion<br />

of soy lipid emulsion-based PN through a central venous<br />

catheter for 3 or 7 days (DSS-PN mice); appropriate controls<br />

were conducted. Cholestasis (increased serum bile acids and<br />

bilirubin) and hepatocyte injury (increased AST and ALT)<br />

were absent in DSS/PN3d mice but developed in all DSS/<br />

PN7d mice. IL-1 beta transcription was significantly induced<br />

in liver homogenate and in isolated hepatic mononuclear cells<br />

(hMNCs) at 3 days in DSS/PN mice and persisted at 7 days.<br />

Concomitantly, transcription of Abcb11, Abcc2, and Abcg5/8<br />

was suppressed at both of these time points in DSS/PN mice,<br />

indicating that macrophage activation and transporter suppression<br />

preceded the onset of biochemical cholestasis. To further<br />

determine if IL-1 beta was responsible for the transcriptional<br />

suppression, wild type mice were injected i.p. with recombinant<br />

IL-1 beta (200ng/mouse) and sampled after 4 hrs, and<br />

HuH7 and HepG2 cells (human hepatocyte cell lines) were<br />

incubated with IL-1b (10-50ng/ml) for 4 hrs, and gene expression<br />

measured. IL-1 beta treatment significantly suppressed<br />

expression of Abcb11, Abcc2, and Abcg5/8 in the mice and<br />

in both hepatocyte cell lines. Incubation of these cell lines with<br />

LPS (100-1000ng/ml) had no effect on these transporters. Conclusions:<br />

Activation of hepatic macrophages and generation of<br />

IL-1 beta, as well as suppression of bile and sterol transporters,<br />

occurred after only 3 days of PN infusion and preceded onset<br />

of cholestasis. Furthermore, IL-1 beta was sufficient to suppress<br />

these transporters in cultured hepatocytes and in vivo. These<br />

data support IL-1 beta as a critical early mediator in the pathogenesis<br />

of PNAC and suggest IL1-beta signaling as a potential<br />

therapeutic target in this disorder.<br />

Disclosures:<br />

Ronald J. Sokol - Advisory Committees or Review Panels: Yasoo Health, Inc.; Consulting:<br />

Roche, Ikaria, Otsuka American Pharmaceuticals, Alnylam, Retrophin;<br />

Grant/Research Support: Mead Johnson Nutritionals, Lumena, FFF Enterprises<br />

The following authors have nothing to disclose: Karim C. El Kasmi, Aimee Anderson,<br />

Michael W. Devereaux<br />

1688<br />

Molecular mechanism of IL-1 beta mediated transcriptional<br />

suppression of the sterol transporter Abcg5/8 in<br />

a parenteral nutrition mouse model<br />

Karim C. El Kasmi, Aimee Anderson, Padade M. Vue, Michael<br />

W. Devereaux, Natarajan Balasubramaniyan, Frederick J. Suchy,<br />

Ronald J. Sokol; Pediatrics, UC Denver, Aurora, CO<br />

Background: Parenteral nutrition associated cholestasis (PNAC)<br />

is the leading indication for combined intestine/liver transplant.<br />

A possible role of intravenous soy lipid emulsions in PNAC<br />

pathogenesis has led to concern about the hepatic toxicity of<br />

phytosterols contained in soy lipid emulsions. We have demonstrated<br />

in a novel PNAC mouse model that soy lipid emulsions<br />

in PN are associated with induction of hepatic macrophage<br />

IL1b expression concomitant with accumulation of hepatic phytosterols<br />

and transcriptional suppression of hepatocyte Abcg5/<br />

g8, coding for the canalicular transporter of sterols, as well<br />

as BSEP and MRP2. The aim of this study was to elucidate the<br />

role of IL1b signaling in suppression of ABCG5/8 transcription<br />

during PNAC. Methods and Results: Wild type C57/B6 mice<br />

were exposed to dextran sulfate sodium (DSS) (to induce intestinal<br />

injury) for 4d followed by infusion of phytosterol-containing<br />

(soy lipid) PN solution through a central venous catheter for 14<br />

d (DSS-PN mice) and developed cholestasis (increased serum<br />

bile acids and bilirubin) and hepatocyte injury (increased AST<br />

and ALT). DSS-PN mice had hepatic IL1b induction and markedly<br />

reduced hepatic mRNA for Abcb11, Abcc2, Abcg5/8<br />

compared to control mice. To further elucidate the role of IL1b<br />

in this transcriptional suppression, mice with genetic deletion

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

Saved successfully!

Ooh no, something went wrong!