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 547A<br />

including Nanog, Sox2 and Oct4. (4) Increased levels of several<br />

stem-ness genes and telomerase (TERT) (25-45 fold), c-Myc<br />

(20-100 fold), IGF2 (~20 fold), with decreased levels of molecules<br />

implicated in BWS- P57 and CTCF were observed in liver<br />

tumors that developed in Sptbn1 +/- /Smad3 +/- mice. (5) Disruption<br />

of the β2SP/SMAD3/CTCF complex raises TERT levels<br />

in BWS and in Sptbn1 +/− /Smad3 +/− mice. (6) We observed<br />

that disruption of the β2SP/SMAD3/CTCF complex increases<br />

stem-like properties (increased ALDH-positive population and<br />

Sphere formation) and enhances tumorigenesis in human HCC<br />

cell lines. (7) We identified a mechanism by which the long<br />

range chromatin modulator CTCF facilitates TGF-β-mediated<br />

repression of hTERT transcription via β2S/SMAD3/CTCF interactions.<br />

Conclusions: We present new insights into liver tumor<br />

formation through the human stem cell disorder BWS and our<br />

mutant models in the TGF-β pathway, demonstrating a potential<br />

shift arising from disruption of TGF-β/CTCF signaling leading<br />

to BWS-associated tumorigenesis, telomerase activation,<br />

and human stem cell associated cancer development. This syndrome<br />

can provide new strategies for preventing and targeting<br />

liver cancers.<br />

Disclosures:<br />

The following authors have nothing to disclose: Jian Chen, Jiun-Sheng Chen,<br />

Young Jin Gi, H. Franklin Herlong, Yun Seong Jeong, Nipun Mistry, Xiaoping<br />

Su, Asif Rashid, Bibhuti Mishra, Jon White, Milind Javle, Marta L. Davila, John<br />

R. Stroehlein, Rosanna Weksbergc, Jerry W. Shay,, Keigo Machida, Hidekazu<br />

Tsukamoto, Lopa Mishra<br />

683<br />

Self-renewing diploid Axin2+ cells fuel homeostatic<br />

renewal of the liver<br />

Bruce M. Wang 1 , Roel Nusse 2,3 ; 1 Medicine, UCSF Liver Center,<br />

San Francisco, CA; 2 Developmental Biology, Stanford University,<br />

Stanford, CA; 3 Howard Hughes Medical Institute, Stanford, CA<br />

The cellular source of new hepatocytes in the adult liver and<br />

the molecular regulation of hepatocyte renewal are fundamental<br />

unanswered questions in liver biology. While it has been<br />

shown that new hepatocytes in the uninjured liver arise from<br />

pre-existing hepatocytes, hepatocytes are known to be heterogenous<br />

with striking differences in age and function across<br />

the liver lobule. It is unknown whether a specific subpopulation<br />

of hepatocytes serves homeostatic renewal in the liver. Using<br />

a novel mouse model for stably labeling Wnt-responsive cells<br />

in vivo, we have discovered a unique population of Wnt-controlled<br />

hepatocytes with stem cell characteristics surrounding<br />

the central vein. These pericentral cells express the early liver<br />

progenitor marker Tbx3, proliferate at a faster rate than other<br />

hepatocytes and are diploid, and thus differ from mature<br />

hepatocytes, which are mostly polyploid. Over time, these<br />

cells self-renew and give rise to descendants that differentiate<br />

into Tbx3-negative, polyploid hepatocytes and can replace all<br />

hepatocytes along the liver lobule during homeostatic renewal.<br />

Importantly, these Wnt-responsive cells are present in the normal<br />

liver, thereby distinguishing them from injury-induced multipotent<br />

progenitor cells (oval cells) associated with the portal<br />

region. Adjacent central vein endothelial cells provide essential<br />

Wnt signals that maintain the pericentral cell population,<br />

thereby constituting the hepatocyte stem cell niche. Thus, we<br />

describe for the first time a subset of hepatocytes that subserves<br />

homeostatic hepatocyte renewal, characterize its anatomical<br />

niche, and identify molecular signals that regulate its activity.<br />

Disclosures:<br />

The following authors have nothing to disclose: Bruce M. Wang, Roel Nusse<br />

684<br />

FAP disease modelling using patient-specific iPS cells<br />

derived from urine<br />

Christoph J. Niemietz, Vanessa Sauer, Jacquelline Stella, Gursimran<br />

Chandhok, Andree Zibert, Hartmut H. Schmidt; Klinik für<br />

Transplantationsmedizin, Universitätsklinikum Münster, Münster,<br />

Germany<br />

Transthyretin-related familial amyloid polyneuropathy (TTR-<br />

FAP) is a rare genetic neurodegenerative disease caused by<br />

mutations of TTR. TTR is primarily secreted by the liver and<br />

misfolding ultimately leads to aggregation and fibril deposition<br />

in peripheral tissues and organs. In order to prevent expression<br />

of TTR, gene silencing strategies are currently under clinical<br />

investigation. The use of primary cells derived from FAP<br />

patients for evaluation of antisense strategies is difficult. As<br />

an alternate, stem cell technology is used here to generate<br />

patient specific primary cells. Urine collections of FAP patients<br />

were processed for isolation of renal epithelial cells, followed<br />

by reprogramming into iPS cells (iPSCs) using non-integrating<br />

episomal vectors. After characterization of pluripotent cell lines,<br />

differentiation towards hepatocyte-like cells was accomplished<br />

after treatment with factors for 14 days. iPSC-Heps were characterized<br />

by analysis of typical hepatic markers via qRT-PCR,<br />

flow cytometry, and immunocytochemistry. ASOs and siRNAs<br />

were introduced into iPSC-Heps in order to evaluate gene<br />

silencing of TTR via qRT-PCR analysis, ELISA and Western-blot.<br />

Typically, 2-3 stable cell colonies emerged upon cultivation<br />

of urine-derived cells from FAP patients (n=12). iPSC-Heps<br />

showed similar morphology and gene expression in comparison<br />

to HepG2 control. TTR gene expression was high. ASOs or<br />

siRNA treatment resulted in downregulation of TTR. Our results<br />

indicate that iPSCs derived from urine cells of FAP patients can<br />

be routinely reprogrammed and differentiated into iPSC-Heps<br />

expressing TTR at comparable levels as control cells. TTR gene<br />

silencing analysis of iPSC-Heps allows the establishment of a<br />

patient-specific in vitro platform for evaluating drug efficiency.

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

Saved successfully!

Ooh no, something went wrong!