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 551A<br />

bution of GFP was inversely correlated with alpha-fetoprotein<br />

(AFP) expression, suggesting local insulin signaling mediated<br />

by IRS2 may play a role in tumor heterogeneity and cellular<br />

differentiation. Exogenous expression of IRS2 promoted tumorigenesis<br />

in HepG2 soft agar and clonogenic assays and also<br />

corresponded with decreased AFP immunostaining. In HepaRG<br />

pIRS2-GFP positive cells were observed in discrete sites within<br />

the cultures surrounding “islands” of hepatocyte differentiation.<br />

IRS2 enriched cells had progenitor-like properties and we show<br />

that insulin/IRS2 signaling at a cellular level determines the<br />

differentiation, proliferative expansion and survival of hepatocyte-like<br />

tumor cells within the cultures, whilst expression of<br />

IRS2 is dynamically regulated during the timecourse of hepatocyte<br />

differentiation such that levels diminished as cells matured.<br />

Taken together, our results underscore the heterogeneity of<br />

insulin sensitivity at the cellular level within “homogeneous”<br />

cancer cell lines. We show that IRS2 expression is spatially<br />

patterned both in monolayers in vitro and in tumors in vivo and<br />

serves as a proxy for insulin sensitivity. Further investigation of<br />

how this novel cellular niche reacts to aberrant insulin signaling<br />

in the context of metabolic disease may provide future insights<br />

into the links that underpin the association between type II diabetes<br />

and HCC risk.<br />

Disclosures:<br />

The following authors have nothing to disclose: Fátima Manzano Núñez, Carlos<br />

Acosta Umanzor, Aránzazu Leal Tassias, Deborah J. Burks, Luke A. Noon<br />

692<br />

Sonic Hedgehog-Containing Exosomes Mediate Biliary<br />

Differentiation and Fibronectin Deposition via Epigenetic<br />

Regulation<br />

Nidhi Jalan-Sakrikar, Thiago de Assuncao, Jie Lu, Gwen Lomberk,<br />

Martin E. Fernandez-Zapico, Raul A. Urrutia, Robert C. Huebert;<br />

Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN<br />

Background: Developmental morphogens play an important<br />

role in coordinating the ductular reaction and portal fibrosis<br />

that occur in the setting of cholangiopathies. However, little is<br />

known about how injured cholangiocytes signal to adjacent<br />

progenitor cells to promote repair after biliary injury. Recent<br />

<strong>studies</strong> show that sonic hedgehog (Shh) signaling is activated<br />

during liver regeneration and repair. In addition, cholangiocytes<br />

release exosomes that contain biologically active Shh and<br />

the secretion of these organelles increases during biliary fibrosis.<br />

Our lab has recently reported a differentiation protocol that<br />

generates induced pluripotent stem cell (iPSC)-derived cholangiocytes<br />

(iDC), a model useful for studying the stem cell to cholangiocyte<br />

transition. The purpose of this study was to dissect<br />

the mechanisms whereby injured cholangiocytes communicate<br />

with the neighboring progenitor cells via Shh-containing exosomes<br />

to initiate both cholangiocyte differentiation and extracellular<br />

matrix (ECM) remodeling. Methods and Results: Next<br />

generation RNA sequencing at each phase of the iPSC to iDC<br />

transition revealed significant upregulation of the Smoothened<br />

(SMO) receptor as well as Gli-1 and Gli-2 transcription factors<br />

during biliary specification and these changes correlated<br />

with a 60-fold increase in fibronectin (FN) levels. Exosomes<br />

isolated from LPS-injured cholangiocytes enhanced progenitor<br />

cell acquisition of cholangiocyte markers (CK7: 2.7±0.3-fold;<br />

CK19: 2.3±0.2-fold) as well as the expression and release<br />

of FN (8.3±2.4 fold). Cholangiocyte differentiation and FN<br />

release were both mediated by Shh as shown using pharmacologic<br />

(cyclopamine) or genetic (SMO shRNA) inhibition of<br />

Shh signaling. Concurrent alterations in epigenetic regulators<br />

(SetD7, KDM5D, EZH2) suggested that a common epigenetic<br />

regulatory program, driven by Shh, may mediate both cholangiocyte<br />

differentiation and ECM deposition. Cholangiocyte<br />

differentiation was associated with temporal alterations in histone<br />

methylation patterns that were Shh-responsive, including<br />

early increases in H3K4me3 (4.6±1.6-fold) and late decreases<br />

in H3K27me3 (-3.8±1.2-fold). In vivo, mice fed the choline-deficient,<br />

ethanolamine supplemented diet had an expansion of<br />

LGR5+ progenitor cells that was associated with Shh activation,<br />

increased FN deposition, and peri-portal fibrosis. Conclusions:<br />

We conclude that Shh-containing exosomes play an<br />

integral role in cholangiocyte differentiation from progenitor<br />

cells following biliary injury and that an epigenetically-driven<br />

cascade of gene regulation may simultaneously promote FN<br />

deposition and biliary fibrosis.<br />

Disclosures:<br />

The following authors have nothing to disclose: Nidhi Jalan-Sakrikar, Thiago de<br />

Assuncao, Jie Lu, Gwen Lomberk, Martin E. Fernandez-Zapico, Raul A. Urrutia,<br />

Robert C. Huebert<br />

693<br />

Interleukin-17 mediates liver progenitor cell transformation<br />

into cancer stem cells in hepatocellular carcinoma.<br />

Imène Gasmi 1,2 , Adrien Guillot 2,3 , Nabila Hamdaoui 1,2 , Arthur<br />

Brouillet 1,2 , Julien Calderaro 1,2 , Benoit Rousseau 1,2 , Jean-Michel<br />

Pawlotsky 1,2 , Fouad Lafdil 1,2 ; 1 INSERM U955 Henri Mondor Hospital,<br />

INSERM / University of Paris Est, Créteil, France; 2 INSERM<br />

/ University of Paris Est, Creteil, France; 3 NIH NIAAA, Bethesda,<br />

MD<br />

Introduction. Hepatocellular carcinoma (HCC) is the third leading<br />

cause of cancer-related death. HCC arises in the setting<br />

of cirrhotic livers in 80 to 90% of cases and progresses in an<br />

inflammatory context. Cancer stem cell biology has recently<br />

sparked the interest of scientists because of their capacity to<br />

initiate and to enhance tumor progression. However the underlying<br />

mechanisms by which expansion of CSCs is initiated are<br />

still unclear. After severe and chronic liver injury, liver progenitor<br />

cell compartment is activated under sustained inflammatory<br />

response. LPCs participate to the regenerative process and are<br />

also observed in HCC. Interestingly, among the large spectrum<br />

of released cytokines in HCC, recent <strong>studies</strong> identified<br />

IL-17-producing cells as a factor associated with a poor prognosis<br />

of the disease. In this study, we propose to determine<br />

whether IL-17 could be involved in tumorigenesis, in particular,<br />

by promoting the transformation of resident liver progenitor<br />

cells (LPCs) into CSCs. Materiels and methods. Serial sections<br />

from 70 HCC-patients were immuno-stained to identify LPCs<br />

(with anti-CK19) and IL-17-producing cells (with anti-IL-17). In<br />

vitro, a murine LPC line (BMOL) was used for long-term culture<br />

with or without IL-17 for 10, 20, 30 or 40 days. The expression<br />

of cancer stem cell markers were analyzed by quantitative<br />

RT-PCR and flow cytometry. Cell cycle controlling factors<br />

were assessed by western blot. Acquired self-renewal property<br />

of LPCs after long term culture with IL-17 was assessed by<br />

spheroid formation capacity analysis. Results. Identification<br />

and semi-quantitative analysis of CK19+ and IL-17+ cells in<br />

HCC patients showed a positive correlation between the number<br />

of infiltrated IL-17+ cells and the number of LPCs. In vitro,<br />

LPC stimulation with IL-17 led to increased expression of CSC<br />

marker mRNA expression including Klf4, ALDH1A1 or CD133,<br />

EpCAM and Glypican-3. CD133 protein and ALDH1A1 activity<br />

analyzed by flow cytometry, were enhanced in LPC cultured<br />

for 10, 20 or 30 days when compared to none treated LPCs.<br />

Cell cycle analysis showed that IL-17 induces the expression<br />

of the proto-oncogene c-Raf, and cyclin D1. In addition, LPCs<br />

cultured for 10 days in presence of IL-17 were able thereafter<br />

to form spheroids when transferred in low-attachment culture

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

Saved successfully!

Ooh no, something went wrong!