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.

458A AASLD ABSTRACTS HEPATOLOGY, October, 2015<br />

of four genes, c-myc, AKT, MET and Nr1h3, correlated with a<br />

subset of Opn-positive HCC tumors. This innovative paradigm<br />

allows for prospective testing of millions of combinations of<br />

overexpressed factors to determine the interaction of known<br />

and suspected liver oncogenes and tumor suppressors in a<br />

systematic fashion.<br />

Disclosures:<br />

The following authors have nothing to disclose: Monica Teta-Bissett, Kirk J. Wangensteen,<br />

Klaus H. Kaestner<br />

495<br />

Peretinoin, an Acyclic Retinoid, Inhibits Hepatocarcinogenesis<br />

through Suppression of Sphingosine Kinase 1<br />

Expression In vitro and In vivo<br />

Masaya Funaki 1 , Tetsuro Shimakami 1 , Tsuguhito Ota 2 , Masao<br />

Honda 1 , Takayoshi Shirasaki 1 , Shuichi Kaneko 1 ; 1 Disease Control<br />

and Homeostasis, Kanazawa university, Kanazawa, Japan;<br />

2 Brain/Liver Interface Medicine Research Center, Kanazawa University,<br />

Kanazawa, Japan<br />

[Objective] A sphingolipid, sphingosine-1-phospate (S1P) is<br />

a potent bioactive lipid which can regulate carcinogenesis<br />

and cancer progression. Both sphingosine kinase 1 (SPHK1)<br />

and SPHK2 are the essential kinases that produce S1P. Therefore,<br />

SPHK can be a therapeutic target by crucially regulating<br />

sphingolipid metabolism in several kinds of cancer. Peretinoin<br />

(NIK-333), an acyclic retinoid, was reported to inhibit the post<br />

therapeutic recurrence of hepatocellular carcinoma (HCC) in<br />

patients with chronic hepatitis C. However, the mechanism of<br />

its inhibitory effects against recurrent HCC remains unclear.<br />

We hypothesized that peretinoin could prevent hepatocarcinogenesis<br />

by modifying a SPHK-S1P axis. In the present study,<br />

we assessed the effect of peretinoin on SPHK activation and<br />

development of liver cancer in vivo and in vitro. [Method] We<br />

examined the effect of peretinoin on the expression and the<br />

enzymatic activity of SPHK1 in a human hepatoma cell line,<br />

Huh-7 cells. Next, using a liver cancer mice model with atherogenic<br />

high fat (AHF) diet-induced NASH we administrated<br />

an AHF diet with and without peretinoin (0.03%) for 48 weeks<br />

and examined the effect of peretinoin on hepatocarcinogenesis<br />

and expression of SPHK1. Finally, we clarified the effect of<br />

SPHK1 on hepatocarcinogenesis induced by Diethylnitrosoamine<br />

(DEN) using SPHK1 knockout mice. [Results] [In vitro] The<br />

treatment with peretinoin (10 to 40 μM) reduced the mRNA<br />

and protein expression of SPHK1 in Huh-7 cells in a time- and<br />

dose- dependent manner. However, peretinoin did not change<br />

the expression of SPHK2 in Huh-7 cells. Furthermore, peretinoin<br />

markedly reduced the enzymatic activity of SPHK1 assessed by<br />

in vitro 32P labeled SPHK activity assay. Next, we performed<br />

reporter gene assays by using constructs containing the SPHK1<br />

promoter region. Peretinoin reduced the promoter activity of<br />

SPHK1, and overexpression of SP1 restored promoter activity.<br />

In addition, three deletion constructs with and without SP1<br />

binding sites were created. The deletion constructs without SP1<br />

binding sites abolished the promoter activity. Interestingly, we<br />

previously reported that peretinoin suppresses the expression of<br />

SP1. Collectively, peretinoin reduced the expression of SPHK1<br />

mRNA via SP1. [In vivo] At week 48 week of treatment, peretinoin<br />

down-regulated SPHK1 mRNA expression and prevented<br />

AHF diet-induced hepatocarcinogenesis. The SPHK1 knockout<br />

mice developed significantly less numbers of HCC induced by<br />

DEN than did wild-type mice. [Conclusion] Our data indicate<br />

that peretinoin prevents hepatocarcinogenesis at least partly<br />

through reduction of expression and activation of SPHK1.<br />

Disclosures:<br />

Shuichi Kaneko - Grant/Research Support: MDS, Co., Inc, Chugai Pharma., Co.,<br />

Inc, Toray Co., Inc, Daiichi Sankyo., Co., Inc, Dainippon Sumitomo, Co., Inc,<br />

Ajinomoto Co., Inc, Bristol Myers Squibb., Inc, Pfizer., Co., Inc, Astellas., Inc,<br />

Takeda., Co., Inc, Otsuka„ÄÄPharmaceutical, Co., Inc, Eizai Co., Inc, Bayer<br />

Japan, Eli lilly Japan<br />

The following authors have nothing to disclose: Masaya Funaki, Tetsuro Shimakami,<br />

Tsuguhito Ota, Masao Honda, Takayoshi Shirasaki<br />

496<br />

Therapeutic overexpression of miR-122 protects mice<br />

from chronic alcoholic liver injury through regulation of<br />

hypoxia-inducible factor-1α<br />

Abhishek Satishchandran 1 , Aditya Ambade 2 , Banishree Saha 2 ,<br />

Benedek Gyongyosi 2 , Patrick Lowe 2 , Nicita Mehta 2 , James V. Zatsiorsky<br />

2 , Arvin Iracheta-Vellve 2 , Jia Li 3,4 , Donna Catalano 2 , Karen<br />

Kodys 2 , Li Zhong 3,4 , Jun Xie 3,5 , Shashi Bala 2 , Guangping Gao 3,5 ,<br />

Gyongyi Szabo 2 ; 1 University of Massachusetts Medical School,<br />

Worcester, MA; 2 Medicine, UMass Medical School, Worcester,<br />

MA; 3 Gene Therapy Center, UMass Medical School, Worcester,<br />

MA; 4 Pediatrics, UMass Medical School, Worcester, MA; 5 Microbiology<br />

and Physiology Systems, UMass Medical School, Worcester,<br />

MA<br />

Introduction: miR-122 regulates essential pathways in alcoholic<br />

liver disease (ALD). We have observed that chronic alcohol<br />

administration reduces miR-122 levels in murine hepatocytes.<br />

Given its essential role in hepatic homeostasis, we hypothesized<br />

that loss of miR-122 contributes to ALD and can be a therapeutic<br />

target. In this study, our goals were to asses: first, the<br />

role of miR-122 in the pathogenesis of ALD and the therapeutic<br />

potential of miR-122 restoration; second, the role of Hypoxia<br />

Inducible Factor 1-α (HIF-1α), a putative miR-122 target; and<br />

third, the mechanisms by which alcohol downregulates miR-<br />

122. Methods: Wild-type 8-week-old, C57Bl/6 or hepatocyte<br />

specific HIF-1α knockout (hepHIFKO) mice were injected intravenously<br />

with hepatocyte-tropic AAV (AAV8) containing antimiR-122<br />

TuD (TuD), or scrambled (Scr) vector. After 14 days,<br />

mice were started on a Lieber-DeCarli (LDC) alcohol diet (Et) or<br />

calorie-matched control diet (PF) for 4 weeks. Some WT mice<br />

were treated with AAV8 pri-miR-122 (OX) on day 7 of the<br />

LDC diet. Results: Anti-miR-122 TuD treatment alone resulted in<br />

significant increases in liver injury (ALT), steatosis, inflammation<br />

(TNFα, IL1-β, MCP-1), and fibrosis (Acta2, pro-col1α) in PF<br />

mice compared to Scr-treated controls. The co-administration of<br />

miR-122 TuD and alcohol resulted in a synergistic effect, further<br />

increasing liver injury, inflammation and fibrosis. Restoration of<br />

miR-122 in hepatocytes of alcohol-fed mice by treatment with<br />

scAAV8 OX resulted in a significant improvement in serum ALT,<br />

inflammation and fibrosis suggesting a protective role for miR-<br />

122 in ALD. The hepatic expression and DNA-binding activity<br />

of HIF-1α, a miR-122 target, was increased in TuD+PF mice<br />

equivalent to that of alcohol feeding alone. HIF1-α activity was<br />

the highest in TuD+Et mice compared to all other groups. Hep-<br />

HIF deficiency protected mice from liver damage, inflammation<br />

and fibrosis induced either by alcohol or miR-122 knockdown.<br />

We discovered that alcohol specifically inhibits expression<br />

of the precursor miR-122 transcript (pri-miR-122) at the transcriptional<br />

level. Furthermore, grainyhead-like 2 (GRHL2), an<br />

inhibitor of miR-122 transcription, was significantly increased<br />

in hepatocytes of alcohol fed mice compared to controls. Conclusions:<br />

Our findings dissect a novel mechanistic regulatory<br />

axis of miR-122 and demonstrate the therapeutic potential of<br />

miR-122 restoration in ALD. miR-122 inhibition alone mimics<br />

and together with alcohol augments the steatosis, inflammation<br />

and early fibrosis seen in ALD through its downstream target,

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

Saved successfully!

Ooh no, something went wrong!